WO2008131374A1 - Use of low temperature and/or low ph in cell culture - Google Patents

Use of low temperature and/or low ph in cell culture Download PDF

Info

Publication number
WO2008131374A1
WO2008131374A1 PCT/US2008/061123 US2008061123W WO2008131374A1 WO 2008131374 A1 WO2008131374 A1 WO 2008131374A1 US 2008061123 W US2008061123 W US 2008061123W WO 2008131374 A1 WO2008131374 A1 WO 2008131374A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell culture
protein
culture
cell
cells
Prior art date
Application number
PCT/US2008/061123
Other languages
French (fr)
Inventor
Jose Manuel Gomes
Gregory Walter Hiller
Original Assignee
Wyeth
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39493848&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2008131374(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to JP2010506424A priority Critical patent/JP5557736B2/en
Priority to EP08746526.6A priority patent/EP2139986B1/en
Priority to BRPI0810511-1A2A priority patent/BRPI0810511A2/en
Priority to AU2008242632A priority patent/AU2008242632B2/en
Priority to PL08746526T priority patent/PL2139986T3/en
Priority to CN200880013275A priority patent/CN101679943A/en
Priority to KR20147030274A priority patent/KR20140132016A/en
Application filed by Wyeth filed Critical Wyeth
Priority to DK08746526.6T priority patent/DK2139986T3/en
Priority to CA2684727A priority patent/CA2684727C/en
Priority to MX2009011362A priority patent/MX2009011362A/en
Priority to SI200831866T priority patent/SI2139986T1/en
Priority to ES08746526.6T priority patent/ES2646090T3/en
Priority to RU2009139054/10A priority patent/RU2478702C2/en
Publication of WO2008131374A1 publication Critical patent/WO2008131374A1/en
Priority to IL201720A priority patent/IL201720A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/02Preparation of peptides or proteins having a known sequence of two or more amino acids, e.g. glutathione
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • C12N2500/33Amino acids other than alpha-amino carboxylic acids, e.g. beta-amino acids, taurine
    • 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 provides methods of improving protein production by cultured cells, especially mammalian cells. Specifically, the present invention relates to methods of preparing a protein product(s), e.g., a glycoprotein product(s), wherein the protein product characteristics are controlled by manipulating the cell culture environment. The invention also relates to methods of improving the therapeutic efficacy and/or immunogenicity of the protein product(s), e.g., the glycoprotein product(s), produced in mammalian cells, e.g., by manipulating protein glycosylation and decreasing protein aggregation and misfolding, by reducing the temperature and/or the pH of the cell culture.
  • a protein product(s) e.g., a glycoprotein product(s)
  • the invention also relates to methods of improving the therapeutic efficacy and/or immunogenicity of the protein product(s), e.g., the glycoprotein product(s), produced in mammalian cells, e.g., by manipulating protein glycosylation and decreasing protein aggregation
  • HMWA high molecular weight aggregate
  • recombinant overexpression of polypeptides in cells may overload the endoplasmic reticulum (ER) machinery, allowing an increased number of misfolded and/or aggregated proteins to evade degradative pathways and to exit the ER.
  • ER endoplasmic reticulum
  • current protein production methods may result in a large proportion of product which is aggregated, nonfunctional, and thus, unusable as produced.
  • misfolded and/or aggregated protein is undesirable because it may lead to adverse events upon administration, including but not limited to, a potential for immunogenicity upon administration (e.g., complement activation or anaphylaxis).
  • a potential for immunogenicity upon administration e.g., complement activation or anaphylaxis
  • excessive therapeutic protein misfolding and/or aggregation may lead to failure in, e.g., clinical trials. Therefore, there is a need in the art for new methods of limiting or reducing protein misfolding and/or aggregation.
  • Protein glycosylation is a common posttranslational modification process by which complex sugar moieties are added onto a protein through the actions of a series of specialized enzymes - glycotransferases and glycosidases - in the ER. This process can control proper folding of the newly synthesized polypeptides such that only the correctly folded polypeptides exit the ER, whereas the misfolded proteins are degraded. Patterns of protein glycosylation affect protein targeting, structure, thermodynamic stability, and enzymatic activity (see, e.g., Sola et al. (2007) Cell. MoI. Life ScL 64:2133-52; Sola and Griebenow (2006) FEBS Lett. 580:1685-90).
  • iV-glycan sialylation is associated with an increase in the lifetime of glycoproteins because such glycoproteins are not recognized by asialo glycoprotein receptors, which target nonsialylated proteins for degradation (see, e.g., Bork et al. (2007) FEBS Letters 581:4195-98).
  • alterations in protein glycosylation may affect the quality and efficacy of the product.
  • aberrant protein glycosylation may result in immunogenicity of the final therapeutic protein product.
  • Proteins produced under nonnatural, suboptimal conditions can acquire sugars and sugar patterns not found naturally on human proteins, and thus result in immunogenic reaction in the subject
  • Fc-fusion protein therapeutics e.g., soluble receptor Fc-fusion protein therapeutics.
  • the invention is a method of producing a protein in a cell culture comprising at least one of: (a) growing cells in the cell culture at a reduced temperature; and (b) growing cells in the cell culture at a reduced pH; so that production of misfolded proteins and/or aggregated proteins is reduced.
  • cells are grown in the cell culture at a reduced temperature and at a reduced pH.
  • the invention is directed to modifying pH and temperature parameters to reduce production of misfolded and/or aggregated proteins in mammalian cell culture, and especially Chinese Hamster Ovary ("CHO") cell culture.
  • the cell culture produces a protein (the "produced protein") that is a soluble receptor, such as, without limitation TNFR-Fc or sIL-13R proteins.
  • the reduced temperature may be in a range of 27.O 0 C to less than 30.0 0 C.
  • the reduced pH is in a range of 6.80 to less than 7.00.
  • a combination of pH and temperature in the aforesaid ranges may also be used.
  • the invention is a method of producing a protein in a cell culture, wherein the level of protein glycosylation of the produced protein is controlled by modifying the temperature and/or the pH of the cell culture.
  • the level of glycosylation such as, without limitation, iV-glycan sialylation, may be increased by increasing temperature and/or pH, or decreased by decreasing temperature and/or pH.
  • the invention is a method of producing a therapeutic protein controlling the parameters described above.
  • the invention is a pharmaceutical composition comprising a therapeutic protein made by such method and a pharmaceutically acceptable carrier.
  • FIG. IA represents integrated viable cell numbers (Y-axis; IVC [normalized e 9 cells * day/L]), normalized to average harvest day IVC, of CHO cells transfected with TNFR-Fc grown either at 27.O 0 C [ ⁇ ], 28.O 0 C [A], 29.O 0 C [ ⁇ ], or 30.0 0 C[O] over time (X-axis; culture time in days [d]);
  • FIG. IB represents cell viability (Y-axis) of the same cells over time (X-axis; culture time [d]).
  • FIG. 2A represents the residual glucose profile (Y-axis; glucose [g/L]) in the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O 0 C [ ⁇ ], 28.O 0 C [A], 29.O 0 C [ ⁇ ], or 30.0 0 C[O] over time (X-axis; culture time [d]);
  • FIG. 2B represents the glutamine profile (Y-axis; glutamine [mM]) of the same cells over time (X-axis; culture time [d]).
  • FIG. 3A represents lactate concentration in the media (Y-axis; lactate [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O 0 C [ ⁇ ], 28.O 0 C [A], 29.O 0 C [ ⁇ ], or 30.0 0 C[O] over time (X-axis; culture time [d]);
  • FIG. 3B represents the ammonium profile (Y-axis; NH 4 + [mM]) of the same cells over time (X-axis; culture time [d]).
  • FIG. 4A depicts cell-specific productivities, represented by cumulative average Qp (X-axis; Cum. Avg. Qp [normalized mg/e 9 cells/day]), normalized to average harvest day cumulative average Qp, of the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O 0 C [ ⁇ ], 28.O 0 C [A], 29.O 0 C [ ⁇ ], or 30.0 0 C[O] over time (X-axis; culture time [d]); while FIG. 4B represents TNFR-Fc titer (X-axis; product titer [normalized mg/L]), normalized to average harvest day titer, of the same cells over time (culture time [d]).
  • FIG. 5A The effect of varying production temperatures (X-axis; [ 0 C]) in the cell culture of CHO cells transfected with TNFR-Fc on the production of misfolded and/or aggregated TNFR-Fc (Y-axis; % misfolded / aggregated product) is demonstrated in FIG. 5A; while the effect of temperatures on the production of HMWA (Y-axis; % high molecular weight species) is demonstrated in FIG. 5B.
  • FIG. 6A The effect of varying production temperatures (X-axis; temperature [ 0 C]) in the cell culture of CHO cells transfected with TNFR-Fc on the percentage of total sialylation (N- and 0-linked sialylation) of TNFR-Fc (Y-axis; percentage of total sialylation of reference material) is demonstrated in FIG. 6A.
  • the reference material used was a specific aliquot of TNFR-Fc with known and preferable glycosylation pattern to which the assay results may be compared.
  • 6B demonstrates the effect of varying production temperatures (X-axis; temperature [ 0 C]) in the cell culture of the same cells on the percentage of total sialylated (D) or non-sialylated (•) iV-linked glycans (Y-axis; Percentage of Total iV-linked Glycans).
  • FIG. 7 demonstrates the effect of growing cells at different temperatures (27.O 0 C [ ⁇ ], 28.O 0 C [A], 29.O 0 C [ ⁇ ], or 30.0 0 C[O]) on the pH of the CHO cell culture (Y-axis) over time (X-axis; culture time [d]). [0019] FIG.
  • FIG. 8A represents integrated viable cell numbers (Y-axis; IVC [normalized e 9 cells *day/L]), normalized to average harvest day IVC, of the CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]); while FIG. 8B represents cell viability (Y-axis) of the same cells over time (X-axis; culture time [d]).
  • FIG. 9A represents glucose profile (Y-axis; glucose [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc, grown at either pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]); while FIG. 9B represents glutamine profile (Y-axis; glutamine [mM]) of the same cells over time (X-axis; culture time [d]).
  • FIG. 9A represents glucose profile (Y-axis; glucose [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc, grown at either pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]); while FIG. 9B represents glutamine profile (Y-axis; glutamine [mM]) of the same cells over time (X-axis;
  • FIG. 1OA represents lactate concentration in the media (Y-axis; lactate [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X- axis; culture time [d]); while FIG. 1OB represents ammonium profile (Y-axis; NH 4 + [mM]) of the same cells over time (X-axis; culture time [d]). [0022] FIG.
  • FIG. HB represents deviations of the pH of the cell culture from the pH set point (X-axis; pH of culture), where the CHO cells transfected with TNFR-Fc were grown at a pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]).
  • FIG. HB represents osmolality (X-axis; osmolality [mOsm/kg]) of the same cells grown at a pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]).
  • FIG. 12A represents cell specific productivities, represented by cumulative average Qp (X-axis; Cum. Avg. Qp [normalized mg/e 9 cells/day]), normalized to average harvest day cumulative average Qp, of the cell culture of CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ⁇ ], 7.00 [ ⁇ ], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]).; while FIG. 12B represents TNFR-Fc titer (X-axis; product titer [normalized mg/L]), normalized to average harvest day titer, of the same cells over time (culture time [d]).
  • FIG. 13A represents the effect of varying the pH set point of the culture (X-axis; pH) of CHO cells transfected with TNFR-Fc on the production of misfolded / aggregated TNFR-Fc (Y-axis; % misfolded / aggregated product).
  • FIG. 13B represents the effect of varying the pH set point of the cell culture (X- axis; pH) on the production of HMWA (Y-axis; % high molecular weigh species).
  • FIG. 14A The effect of varying the pH set point (X-axis; pH) in the culture of CHO cells transfected with the TNFR-Fc on the percentage of total sialylation of TNFR-Fc (Y-axis; percentage of total sialylation of reference material) is demonstrated in FIG. 14A.
  • Fig. 14B demonstrates the effect of varying cell culture pH set point (X-axis; pH) in the cell culture of the same cells on the percentage of total sialylated (D) or non-sialylated (•) iV-linked glycans (Y-axis;
  • FIG. 15 depicts a typical fluorescence (Y-axis; fluorescence, measured in mV) versus retention time (X-axis; minutes) profile depicting iV-linked glycan sialylation as observed by subjecting hydrazaine released 2-aminobenzamide-
  • FIG. 16 depicts the fluorescence (Y-axis; mV) versus retention time (X- axis; minutes) profiles depicting iV-linked glycan sialylation as observed by subjecting hydrazaine released 2-aminobenzamide- (2-AB)-labeled protein glycoforms to Normal Phase Chromatography of cell culture of CHO cells transfected with TNFR-Fc at varying pH set points of the culture.
  • FIG. 17 represents (A) viable cell density (Y-axis; cells/mL), (B) total cell density , which includes both viable and non-viable cells, (Y-axis; cells/mL),
  • C cell viability (Y-axis), and (D) integrated viable cell numbers (IVC) (Y-axis; e9 cells*day/L) of the cell culture of sIL-13R overexpressing cells grown at
  • FIG. 18 represents sIL-13R titer, both dimer and HMWA, (Y-axis; sIL-
  • FIG. 19A represents daily specific sIL-13R production rate (Y-axis; Qp
  • FIG. 19B represents cumulative average cell specific productivities (Y- axis; Cum. Avg. Qp [mg/e9 cells/day]) over time (X-axis; culture time [d]) for cell cultures overexpressing sIL-13R, grown at 37.O 0 C, 33.O 0 C, 32.O 0 C, 31.O 0 C,
  • FIG. 20 represents daily specific glucose consumption rate (Y-axis; QgIc
  • FIG. 21 represents daily specific glutamine consumption rate (Y-axis; QgIn [mmol/e9 cell/d]) for different time intervals (X-axis) for cell cultures overexpressing sIL-13R, grown at 37.O 0 C, 33.O 0 C, 32.O 0 C, 31.O 0 C, 29.O 0 C, or RT.
  • FIG. 22 represents lactate concentration (Y-axis; lactate[g/L]) in the cell culture medium of the cells overexpressing sIL-13R, grown at 37.O 0 C [ ⁇ ], 33.O 0 C [ ⁇ ], 32.O 0 C [•], 31.O 0 C [O], 29.O 0 C [ ⁇ ], or RT [D] over time (X-axis; culture time [d]).
  • FIG. 23 represents ammonium concentration (Y-axis; ammonia [mM]) in the cell culture medium of the cells overexpressing sIL-13R, grown at 37.O 0 C [ ⁇ ], 33.O 0 C [ ⁇ ], 32.O 0 C [•], 31.O 0 C [O], 29.O 0 C [ ⁇ ], or RT [D] over time (X- axis; culture time [d]).
  • FIG. 24 represents the effect of cell culture temperature (X-axis; production temperature [ 0 C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 9 of culture of cells overexpressing sIL-13R.
  • FIG. 25 represents the effect of cell culture temperature (X-axis; production temperature [ 0 C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 18 of culture of cells overexpressing sIL-13R.
  • FIG. 24 represents the effect of cell culture temperature (X-axis; production temperature [ 0 C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 9 of culture of cells overexpressing sIL-13R.
  • FIG. 25 represents the effect of cell culture temperature (X-axis; production temperature [ 0 C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 18 of culture of cells overexpressing sIL-13R.
  • 26A represents percentage of the sIL-13R dimer (Y-axis; % sIL- 13R dimer) recovered from the total sIL-13R protein produced in the conditioned media of sIL-13R overexpressing cells grown at either 37.O 0 C [ ⁇ ], 33.O 0 C [ ⁇ ], 32.O 0 C [•], 31.O 0 C [O], 29.O 0 C [ ⁇ ], or RT [D] over time (X-axis; culture time [d]).
  • FIG. 26B represents the percentage of HMWA (Y-axis; % High Molecular Weight Species) relative to total sIL-13R in the conditioned medium of sIL-13R overexpressing cells over time (X-axis; culture time [d]).
  • FIG. 27 represents the sIL-13R dimer titer (Y-axis; sIL-13R dimer only [mg/L]) in the conditioned media of cells grown either 37.O 0 C [ ⁇ ], 33.O 0 C [ ⁇ ], 32.O 0 C [•], 31.O 0 C [O], 29.O 0 C [ ⁇ ], or RT [D] over time (X-axis; culture time [d]).
  • DETAILED DESCRIPTION OF THE INVENTION [0039] The prevailing wisdom for producing therapeutic proteins in a mammalian cell culture is that the temperature during the production phase should be at least 30.0 0 C and the pH should be at least 7.00.
  • HMWA high molecular weight aggregate
  • the present invention provides a novel method of protein production in a cell culture, wherein the method results in decreased protein misfolding and/or decreased protein aggregation.
  • the invention provides for methods of controlling the level of protein glycosylation.
  • polypeptide or “polypeptide product” are synonymous with the terms “protein” and “protein product,” respectively, and, as is generally understood in the art, refer to at least one chain of amino acids linked via sequential peptide bonds.
  • a "protein of interest” or a “polypeptide of interest” or the like is a protein encoded by an exogenous nucleic acid molecule that has been transfected or transformed into a host cell, e.g., transiently or stably transfected or transformed into a host cell.
  • the nucleic acid sequence of the exogenous DNA determines the sequence of amino acids. This sequence may be a sequence that occurs in nature, or may alternatively be a sequence engineered by man.
  • a "protein of interest” is a protein encoded by a nucleic acid molecule that is endogenous to the host cell. The expression of such an endogenous protein of interest may be altered by transfecting a host cell with an exogenous nucleic acid molecule that may, for example, contain one or more regulatory sequences and/or encode a protein that enhances expression of the protein of interest.
  • a polypeptide of interest is produced in the cell culture, e.g., for subsequent purification.
  • glycoprotein refers to proteins that have sugar moieties, e.g., oligosaccharide moieties, attached to either their asparagine side chains (iV-glycosylation) or their serine and/or threonine side chains (0-glycosylation).
  • iV-glycosylation protein sialylation
  • iV-glycan sialylation protein sialylation
  • secretory and membrane proteins are glycosylated in the ER and/or the Golgi apparatus. It is known in the art that glycosylation controls protein folding and release in the ER.
  • the polypeptide of interest is a glycoprotein of interest
  • the glycoprotein of interest is produced in the cell culture, e.g., for subsequent purification.
  • the glycoprotein of interest is a receptor, and may be a soluble receptor.
  • Methods and compositions of the present invention may be used to produce any protein of interest including, but not limited to, proteins having pharmaceutical, diagnostic, agricultural, and/or any of a variety of other properties that are useful in commercial, experimental and/or other applications.
  • a protein of interest can be a protein therapeutic.
  • a protein therapeutic or a therapeutic protein
  • proteins produced using methods and/or compositions of the present invention may be processed and/or modified, before administering to a subject as a therapeutic protein.
  • the present invention may be used to culture cells for the advantageous production of any therapeutic protein, such as pharmaceutically or commercially relevant enzymes, receptors, receptor fusions, soluble receptors, soluble receptor fusions, antibodies (e.g., monoclonal and/or polyclonal antibodies), antigen- binding fragments of an antibody, Fc fusion proteins, SMIPs, cytokines, hormones, regulatory factors, growth factors, coagulation / clotting factors, or antigen-binding agents.
  • therapeutic protein such as pharmaceutically or commercially relevant enzymes, receptors, receptor fusions, soluble receptors, soluble receptor fusions, antibodies (e.g., monoclonal and/or polyclonal antibodies), antigen- binding fragments of an antibody, Fc fusion proteins, SMIPs, cytokines, hormones, regulatory factors, growth factors, coagulation / clotting factors, or antigen-binding agents.
  • therapeutic protein such as pharmaceutically or commercially relevant enzymes, receptors, receptor fusions,
  • the term "antibody” includes a protein comprising at least one, and typically two, VH domains or portions thereof, and/or at least one, and typically two, VL domains or portions thereof.
  • the antibody is a tetramer of two heavy immunoglobulin chains and two light immunoglobulin chains, wherein the heavy and light immunoglobulin chains are inter-connected by, e.g., disulfide bonds.
  • the antibodies, or a portion thereof can be obtained from any origin, including, but not limited to, rodent, primate (e.g., human and non-human primate), camelid, shark as well as recombinantly produced, e.g., chimeric, humanized, and/or in vitro generated, e.g., by methods well known to those of skill in the art.
  • rodent e.g., human and non-human primate
  • camelid e.g., camelid, shark
  • recombinantly produced e.g., chimeric, humanized, and/or in vitro generated, e.g., by methods well known to those of skill in the art.
  • This invention also encompasses "antigen-binding fragments of antibodies", which include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHl domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHl domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment, which consists of a VH domain; (vi) a camelid or camelized variable domain, e.g., a VHH domain; (vii) a single chain Fv (scFv); (viii) a bispecific antibody; and (ix) one or more antigen binding fragments of an immunoglobulin fused to an Fc region.
  • a Fab fragment a monovalent fragment consisting of
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-26; Huston et al. (1988) Proc. Natl. Acad. ScL U.S.A. 85:5879-83).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody.
  • the invention also encompasses single domain antibodies.
  • Single domain antibodies can include antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies.
  • Single domain antibodies may be any of the art, or any future single domain antibodies.
  • Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, goat, rabbit, cow and shark.
  • a single domain antibody as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains.
  • Such single domain antibodies are disclosed in WO 9404678 for example.
  • this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins.
  • VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention.
  • Single domain antibodies also include shark IgNARs; see, e.g., Dooley et al., Proc. Natl. Acad. ScL U.S.A., 103:1846-1851 (2006).
  • an antibody is understood to have each of its binding sites identical.
  • a "bispecific” or “bifunctional antibody” is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol.
  • the protein in embodiments where the protein is an antibody or a fragment thereof, it can include at least one, or two full-length heavy chains, and at least one, or two light chains.
  • the antibodies or fragments thereof can include only an antigen-binding fragment (e.g., an Fab, F(ab')2, Fv or a single chain Fv fragment).
  • the antibody or fragment thereof can be a monoclonal or single specificity antibody.
  • the antibody or fragment thereof can also be a human, humanized, chimeric, CDR-grafted, or in vitro generated antibody.
  • the antibody has a heavy chain constant region chosen from, e.g., IgGl, IgG2, IgG3, or IgG4.
  • the antibody has a light chain chosen from, e.g., kappa or lambda.
  • the constant region is altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function).
  • the antibody or fragment thereof specifically binds to a predetermined antigen, e.g., an antigen associated with a disorder, e.g., a neurodegenerative, metabolic, inflammatory, autoimmune and/or a malignant disorder.
  • a predetermined antigen e.g., an antigen associated with a disorder, e.g., a neurodegenerative, metabolic, inflammatory, autoimmune and/or a malignant disorder.
  • Proteins described herein optionally, further include a moiety that enhances one or more of, e.g., stability, effector cell function or complement fixation.
  • an antibody or antigen-binding protein can further include a pegylated moiety, albumin, or a heavy and/or a light chain constant region.
  • Antibodies are generally made, for example, via traditional hybridoma techniques (Kohler et al., Nature 256:495 499 (1975)), recombinant DNA methods (U.S. Patent No. 4,816,567), or phage display techniques using antibody libraries (Clackson et al., Nature 352:624 628 (1991); Marks et al., J. MoI. Biol. 222:581 597 (1991)).
  • phage display techniques using antibody libraries.
  • the antibodies may be tagged with a detectable or functional label.
  • labels include radiolabels (e.g., 1311 or 99Tc), enzymatic labels (e.g., horseradish peroxidase or alkaline phosphatase), and other chemical moieties (e.g., biotin).
  • SMIPTM Mal Modular Immunopharmaceutical drugs
  • SMIPTM Single-chain polypeptides composed of a binding domain for a cognate structure such as an antigen, a counterreceptor or the like, a hinge region polypeptide having either one or no cysteine residues, and immunoglobulin CH2 and CH3 domains (see also www.trubion.com).
  • SMIPs and their uses and applications are disclosed in, e.g., U.S. Published Patent Application Nos.
  • the protein of interest is a soluble receptor, e.g., a soluble receptor fusion protein.
  • Membrane proteins e.g., receptors
  • Soluble proteins can be produced according to methods well known in the art.
  • a soluble receptor comprises an extracellular region of the receptor, or a fragment of the extracellular region of the receptor.
  • a soluble receptor comprises two polypeptides.
  • the first polypeptide comprises a full-length receptor; alternatively, the first polypeptide comprises less than the full length of the receptor, e.g., an extracellular portion of the receptor.
  • the first polypeptide is a full-length cytokine receptor; alternatively the first polypeptide is less than the full length of the cytokine receptor, e.g., an extracellular portion of the cytokine receptor.
  • Such a soluble receptor can also comprise an additional polypeptide, e.g., a GST, Lex-A, MBP polypeptide sequence or an immunoglobulin chain, including, e.g., an Fc fragment, a heavy chain constant region of the various isotypes, including: IgGl, IgG2, IgG3, IgG4, IgM, IgAl, IgA2, IgD, and IgE.
  • a second polypeptide is preferably soluble.
  • the second polypeptide enhances the half-life, (e.g., the serum or circulatory half-life) of the linked polypeptide.
  • the second polypeptide includes at least a region of an immunoglobulin polypeptide.
  • Immunoglobulin fusion polypeptides are known in the art and are described in, e.g., U.S. Patent Nos. 5,516,964; 5,225,538; 5,428,130; 5,514,582; 5,714,147; and 5,455,165. Soluble fusion proteins are known to be susceptible to aggregation during production, and therefore the methods according to the invention provide a particular benefit in connection with cell cultures that produce proteins of this type.
  • the second polypeptide comprises a full-length immunoglobulin polypeptide.
  • the second polypeptide comprises less than the full length of immunoglobulin polypeptide, e.g., a heavy chain, light chain, Fab, Fab 2 , Fv, or Fc.
  • the second polypeptide can include the heavy chain of an immunoglobulin polypeptide.
  • the second polypeptide can include the Fc region of an immunoglobulin polypeptide.
  • a soluble receptor fusion protein comprises a tumor necrosis factor inhibitor.
  • tumor necrosis factor inhibitors in the form of tumor necrosis factor alpha and beta receptors (TNFR-I; EP 417,563 published Mar. 20, 1991; and TNFR-2, EP 417,014 published Mar. 20, 1991, each of which is incorporated herein by reference in its entirety) are expressed in accordance with systems and methods of the present invention (for review, see Naismith and Sprang, J. Inflamm. 47(1- 2): 1-7, 1995-96, incorporated herein by reference in its entirety).
  • a tumor necrosis factor inhibitor comprises a soluble TNF receptor.
  • TNF inhibitors of the present invention are soluble forms of TNFRI and TNFRII. In certain embodiments, TNF inhibitors of the present invention are soluble TNF binding proteins. In certain embodiments, TNF inhibitors of the present invention are TNFR fusion proteins, e.g., TNFR-Ig or TNFR-Fc. As used herein, "etanercept,” refers to a TNFR-Fc, which is a dimer of two molecules of the extracellular portion of the p75 TNF-alpha receptor, each molecule consisting of a 235 amino acid Fc portion of human IgGl.
  • cells expressing TNFR-Fc are grown in a cell culture at a reduced temperature and/or reduced pH to decrease the amount of misfolded protein and/or high molecular weight aggregates during the production of TNFR-Fc.
  • cells expressing TNFR-Fc are grown in a cell culture at a reduced temperature and/or reduced pH to modulate glycosylation during the production of TNFR-Fc.
  • the soluble receptor fusion protein is the sIL-13R.
  • soluble IL- 13 receptor refers to a recombinant fusion protein which includes extracellular domain (ECD) of the human interleukin (IL)-13-alpha2 receptor and the Fc region of the human IgGl heavy chain.
  • ECD extracellular domain
  • sIL-13R is composed of two identical polypeptide chains (i.e., dimer of two polypeptide chains) which appear to be linked by intermolecular disulfide bonds.
  • sIL-13R soluble receptor fusion protein and its uses are disclosed in U.S. Patent No. 5,710,023, incorporated herein in its entirety by reference.
  • the second polypeptide has less effector function than the effector function of an Fc region of a wild-type immunoglobulin heavy chain.
  • Fc effector function includes, for example, Fc receptor binding, complement fixation and T cell-depleting activity (see, for example, U.S. Patent No. 6,136,310). Methods for assaying T cell-depleting activity, Fc effector function, and antibody stability are known in the art.
  • the second polypeptide has low or no affinity for the Fc receptor. In an alternative embodiment, the second polypeptide has low or no affinity for complement protein CIq.
  • the fusion proteins may additionally include a linker sequence joining the soluble receptor or a fragment thereof to the second moiety.
  • the fusion protein can include a peptide linker, e.g., a peptide linker of about 2 to 20, more preferably about 5 to 10, amino acids in length.
  • additional amino acid sequences can be added to the N- or C-terminus of the fusion protein to facilitate expression, detection and/or isolation or purification.
  • soluble receptor fusion protein may be linked to one or more additional moieties, e.g., GST, His6 tag, FLAG tag.
  • the fusion protein may additionally be linked to a GST fusion protein in which the fusion protein sequences are fused to the C-terminus of the GST (i.e., glutathione S-transferase) sequences.
  • Such fusion proteins can facilitate the solubility, i.e., increase accurate folding, and thus improve purification of the fusion protein.
  • culture refers to a cell population that is contacted with a cell culture medium under conditions suitable to survival and/or growth of the cell population. As used herein, these terms may refer to the combination comprising the cell population (e.g., the animal cell culture) and the medium with which the population is in contact.
  • the cells used in the present invention may be recombinant host cells, e.g., eukaryotic host cells, i.e., cells transfected with an expression construct containing a nucleic acid that encodes a polypeptide of interest, including animal cells.
  • animal cells encompasses invertebrate, nonmammalian vertebrate (e.g., avian, reptile and amphibian), and mammalian cells.
  • invertebrate cells include the following insect cells: Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori (silkworm / silk moth).
  • the cell culture is a mammalian cell culture.
  • a number of mammalian cell lines are suitable host cells for recombinant expression of polypeptides of interest.
  • Mammalian host cell lines include, for example, COS, PER.C6, TM4, VERO076, MDCK, BRL-3A, W138, Hep G2, MMT, MRC 5, FS4, CHO, 293T, A431, 3T3, CV-I, C3H10T1/2, Colo205, 293, HeLa, L cells, BHK, HL-60, FRhL-2, U937, HaK, Jurkat cells, Rat2, BaF3, 32D, FDCP-I, PC12, Mix, murine myelomas (e.g., SP2/0 and NSO) and C2C12 cells, as well as transformed primate cell lines, hybridomas, normal diploid cells, and cell strains derived from in vitro culture of primary tissue and primary explants.
  • murine myelomas e.g., SP2/0 and NSO
  • C2C12 cells
  • any eukaryotic cell that is capable of expressing the polypeptide of interest may be used in the disclosed methods.
  • Numerous cell lines are available from commercial sources, such as the American Type Culture Collection (ATCC).
  • ATCC American Type Culture Collection
  • the cell culture e.g., the large-scale cell culture, employs CHO cells.
  • the cell culture comprises mammalian cells
  • a cell culture comprises mammalian cells
  • yeast eukaryotes
  • prokaryotes such as bacteria
  • Suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium, or any bacterial strain capable of expressing the polypeptide of interest. Expression in bacteria may result in formation of inclusion bodies incorporating the recombinant protein.
  • refolding of the recombinant protein may be required in order to produce active, or more active, material.
  • Several methods for obtaining correctly folded heterologous proteins from bacterial inclusion bodies are known in the art. These methods generally involve solubilizing the protein from the inclusion bodies, then denaturing the protein completely using a chaotropic agent.
  • cysteine residues are present in the primary amino acid sequence of the protein, it is often necessary to accomplish the refolding in an environment that allows correct formation of disulfide bonds (a redox system).
  • General methods of refolding are known in the art and disclosed in, e.g., Kohno (1990) Meth. Enzymol. 185:187-95, EP 0433225, and U.S. Patent No. 5,399,677.
  • Suitable yeast strains for polypeptide production include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Pichia pastoris, Kluyveromyces strains, Candida, or any yeast strain capable of expressing polypeptide of interest.
  • the term "bioreactor” as used herein refers to any vessel used for the growth of a eukaryotic cell culture, e.g., an animal cell culture (e.g., a mammalian cell culture).
  • the bioreactor can be of any size as long as it is useful for the culturing of cells, e.g., mammalian cells.
  • the bioreactor will be at least 30 ml and may be at least 1, 10, 100, 250, 500, 1000, 2500, 5000, 8000, 10,000, 12,000 liters or more, or any intermediate volume.
  • the internal conditions of the bioreactor including but not limited to pH and temperature, are typically controlled during the culturing period.
  • production bioreactor refers to the final bioreactor used in the production of the polypeptide or protein of interest.
  • the volume of a large-scale cell culture production bioreactor is generally greater than about 100 ml, typically at least about 10 liters, and may be 500, 1000, 2500, 5000, 8000, 10,000, 12,000 liters or more, or any intermediate volume.
  • a suitable bioreactor or production bioreactor may be composed of (i.e., constructed of) any material that is suitable for holding cell cultures suspended in media under the culture conditions of the present invention and is conducive to cell growth and viability, including glass, plastic or metal; the material(s) should not interfere with expression or stability of the produced product, e.g., a therapeutic protein product.
  • suitable bioreactors for use in practicing the present invention.
  • medium refers to a solution containing nutrients that nourish growing animal cells, e.g., mammalian cells, and can also refer to medium in combination with cells.
  • the term “inoculation medium” refers to the medium that is used to form a cell culture. Inoculation medium may or may not differ in composition from the medium used during the rest of the cell growth phase.
  • medium solutions provide, without limitation, essential and nonessential amino acids, vitamins, energy sources, lipids, and trace elements required by the cell for at least minimal growth and/or survival.
  • the solution may also contain components that enhance growth and/or survival above the minimal rate, including hormones and growth factors.
  • the medium is a defined medium.
  • defined media are media in which all components have a known chemical structure.
  • the medium may contain an amino acid(s) derived from any source or method known in the art, including, but not limited to, an amino acid(s) derived either from single amino acid addition(s) or from a peptone or protein hydrolysate addition(s) (including animal or plant source(s)).
  • the medium used during the cell growth phase may contain concentrated medium, i.e., medium that contains higher concentration of nutrients than is normally necessary and normally provided to a growing culture.
  • cell media, inoculation media, etc. is appropriate to culture a particular cell, e.g., animal cell (e.g., CHO cells), and the amount of glucose and other nutrients (e.g., glutamine, iron, trace D elements) or agents designed to control other culture variables (e.g., the amount of foaming, osmolality) that the medium should contain (see, e.g., Mather, J.P., et al. (1999) "Culture media, animal cells, large scale production," Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis, and Bioseparation, Vol. 2:777-85; U.S. Published Patent Application No.
  • animal cell e.g., CHO cells
  • nutrients e.g., glutamine, iron, trace D elements
  • agents designed to control other culture variables e.g., the amount of foaming, osmolality
  • cell density refers to the number of cells present in a given volume of medium.
  • viable cell density refers to the number of live cells present in a given volume of medium under a given set of experimental conditions.
  • cell viability refers to the ability of cells in culture to survive under a given set of culture conditions or experimental variations.
  • the term as used herein also refers to that portion of cells that are alive at a particular time in relation to the total number of cells, living and dead, in the culture at that time.
  • integrated viable cell density refers to the average density of viable cells over the course of the culture multiplied by the amount of time the culture has run.
  • integrated viable cell density is a useful tool for estimating the amount of protein produced over the course of the culture.
  • the methods of the present invention are applicable to cells grown in a batch culture, fed batch culture, perfusion culture, modified fed-batch culture (see U.S. Provisional Application Number 60/954,922), batch refeed culture, or any combination thereof.
  • batch culture refers to a method of culturing cells in which all the components that will ultimately be used in culturing the cells, including the medium as well as the cells themselves, are provided at the beginning of the culturing process.
  • a batch culture is typically stopped at some point and the cells and/or components in the medium are harvested and optionally purified.
  • fed-batch culture refers to a method of culturing cells in which additional components are provided to the culture at some time subsequent to the beginning of the culture process.
  • the provided components typically comprise nutritional supplements for the cells that have been depleted during the culturing process.
  • a fed-batch culture is typically stopped at some point and the cells and/or components in the medium are harvested and optionally purified.
  • perfusion culture refers to a method of culturing cells in which additional fresh medium is provided, either continuously over some period of time or intermittently over some period of time, to the culture (subsequent to the beginning of the culture process), and simultaneously spent medium is removed.
  • the fresh medium typically provides nutritional supplements for the cells that have been depleted during the culturing process.
  • Polypeptide product which may be present in the spent medium, is optionally purified.
  • Perfusion also allows for removal of cellular waste products (flushing) from the cell culture growing in the bioreactor.
  • modified fed-batch culture refers to a method of culturing cells which combines both the fed-batch culture method and the perfusion culture method.
  • the modified fed-batch culture method is described in U.S. Provisional Application Number 60/954,922, incorporated herein in its entirety by reference.
  • the invention can also be practiced with batch re-feed processes. It is believed that reduced pH will provide a better handle for reducing protein misfolding and/or aggregation and for modifying glycosylation in this type of cell culture.
  • the method comprises first inoculating the cell culture with the inoculation medium during the growth phase of the cell culture, and subsequently switching cells into the production phase, wherein the temperature and/or pH of the cell culture is adjusted to a reduced temperature and/or a reduced pH.
  • Growth phase refers to a stage in the cell culture wherein the cells are grown to achieve the cell density optimal for protein production.
  • the cells may be switched into the production phase, which may occur at different temperature and/or different pH than the growth phase, for example, reduced temperature and/or reduced pH.
  • the cell culture may be switched from growth phase to production phase one day after the inoculation.
  • the cell culture may be switched from growth phase to production phase five days after the inoculation.
  • the shift may be relatively gradual.
  • the shift may be abrupt.
  • the temperature and/or pH may be steadily adjusted, e.g., decreased.
  • the temperature and/or pH may be adjusted by discrete intervals.
  • Production phase of the cell culture is a stage in the cell culture wherein the cells are grown under conditions optimal for producing the polypeptide of interest, e.g., a therapeutic protein.
  • the reduced temperature and/or pH of the cell culture may be selected based on the temperature and/or pH at which the cell culture remains viable, at which high level of protein is produced, at which production and accumulation of metabolic waste products, e.g., lactic acid and ammonia, is minimized, at which the product quality of the protein has been appropriately controlled, and/or any combination of these or other factors deemed important by the practitioner.
  • the method of the invention comprises inoculating the cell culture at a reduced temperature and/or reduced pH, so no temperature or pH shift is necessary at the initiation of the production phase.
  • a skilled artisan would know to monitor the temperature and the pH of the cell culture such that the temperature and the pH do not deviate from the established temperature and pH set points.
  • a skilled artisan will know to use a base, e.g., sodium carbonate base, to prevent cultures from deviating below the set pH.
  • the cell culture medium started at a pH above the target pH set point and no deliberate adjustment to pH set point occurred.
  • a pH adjustment during the course of the cell culture could also be included as would be understood by those of skill in the art.
  • initiating the culture at low temperature without an adjustment is also possible.
  • the process may be comprised of a production phase only.
  • Reduced temperature refers to a temperature below the conventional temperature for cell growth (the temperature at which the cell are typically grown) for that type of cell.
  • the cell culture in the production phase is preferably in a range of 24.O 0 C to less than 30.0 0 C, and more preferably in a range of 27.O 0 C to less than 30.0 0 C.
  • reduced temperature of the cell culture is 24.O 0 C, 24.5 0 C, 25.O 0 C, 25.5 0 C, 26.O 0 C, 26.5 0 C. 27.O 0 C, 27.5 0 C, 28.O 0 C, 28.5 0 C, 29.O 0 C, 29.5 0 C, 29.6 0 C, 29.7 0 C, 29.8 0 C, and 29.9 0 C.
  • reduced temperature of the cell culture is a temperature of about 29.5 0 C. Reduced temperature for cells other than mammalian may be determined on a case by case basis by a person of ordinary skill in the art.
  • Reduced pH refers to a pH set point below the conventional pH for cell growth (the pH at which the cells are typically grown) of that particular cell type. In embodiments of the present invention, where the cells are mammalian cells, reduced pH of the cell culture in the production phase is below 7.00. In embodiments of the invention, reduced pH of the cell culture is in a range of 6.50 to less than 7.00, preferably in a range of 6.80 to less than 7.00.
  • reduced pH of the cell culture is 6.80, 6.85, 6.90, 6.95, 6.96, 6.97, 6.98, and 6.99. In the most preferred embodiment of the invention, reduced pH of the cell culture is about 6.95.
  • Reduced pH for cells other than mammalian may be determined on a case by case basis by a person of ordinary skill in the art. [0086] A skilled artisan will understand that depending on the cell type of the cell culture, conventional temperature and pH (as distinguished from the reduced temperature and pH) will differ. For example, conventional temperature and pH for most mammalian cells, e.g., CHO cells, is above 30.0° C (such as, for example, 37.O 0 C) and above 7.00 (such as, for example, 7.20), respectively.
  • the methods of the invention will utilize a different reduced temperature and a different reduced pH for such cells.
  • the practitioner may find it beneficial or necessary to periodically monitor particular conditions of the growing cell culture. As nonlimiting examples, it may be beneficial or necessary to monitor, e.g., temperature, pH, dissolved oxygen, cell density, cell viability, integrated viable cell density, lactate levels, ammonium levels, glucose levels, glutamine levels, osmolality, titer of the expressed polypeptide, etc. Numerous techniques are well known to those of skill in the art for measuring such conditions/criteria.
  • cell density may be measured using a hemocytometer, an automated cell-counting device (e.g., a Coulter counter, Beckman Coulter Inc., Fullerton, CA), or cell-density examination (e.g., CEDEX®, Innovatis, Malvern, PA).
  • Viable cell density may be determined by staining a culture sample with Trypan blue. Lactate, ammonium, glucose and glutamine levels, as well as dissolved oxygen and pH may be measured, e.g., with the BioProfile 400 Chemistry Analyzer (Nova Biomedical, Waltham, MA), which takes measurements of key nutrients, metabolites, and gases in cell culture media.
  • Dissolved oxygen and pH may also be measured using, e.g., a blood gas analyzer (e.g., a Bayer Rapidlab 248 pH/blood gas analyzer (Bayer HealthCare LLC, East Walpole, MA)). Temperature, pH, and dissolved oxygen may also be measured by, e.g., various types of in-situ probes. Osmolality of the cell culture may be measured by, e.g., a freezing point osmometer.
  • HPLC can be used to determine, e.g., the levels of lactate, ammonium, or the expressed polypeptide or protein. In one embodiment of the invention, the levels of expressed polypeptide can be determined by using, e.g., protein A HPLC.
  • the level of the expressed polypeptide or protein can be determined by standard techniques such as Coomassie staining of SDS-PAGE gels, Western blotting, Bradford assays, Lowry assays, biuret assays, and UV absorbance. It may be necessary to monitor the posttranslational modifications of the expressed polypeptide or protein, e.g., glycosylation. It also may be beneficial to monitor other posttranslational modifications of the protein, e.g., phosphorylation, etc. In order to monitor certain cell culture conditions, it may be necessary to remove small aliquots of the culture for analysis. One of ordinary skill in the art will understand that such removal may potentially introduce contamination into the cell culture, and will take appropriate care to minimize the risk of such contamination.
  • a practitioner will monitor integrated viable cell number at the reduced cell culture temperature and/or reduced pH.
  • growing cells at the reduced temperature and/or reduced pH reduces integrated viable cell density by 20%, more preferably less than 20% (e.g., 15%).
  • a practitioner will monitor cell viability at the reduced cell culture temperature and/or reduced pH.
  • growing cells at the reduced temperature and/or reduced pH reduces cell viabilities by less than 15%, more preferably by less than 5%.
  • Glucose is a primary source of energy for the cell culture. Significant deviations in glucose consumption in the cell culture could indicate a negative effect of cell culture conditions on the health of the cell culture. Thus, in a preferred embodiment of the invention, growing cells at the reduced temperature and/or reduced pH results in minimal change, e.g., reduction, in glucose consumption of the cell culture.
  • Glutamine is an alternative source of energy for the cell culture, and is an important source of nitrogen for various molecules, e.g., amino acids, in the cell culture.
  • growing cells at the reduced temperature and/or reduced pH results in a minimal change, e.g., reduction, in the glutamine consumption of the cell culture.
  • waste product production e.g., lactate and ammonia production
  • growing cells at the reduced temperature and/or pH results in a minimal change, e.g., increase, in production of lactate and ammonia.
  • growing cells at a reduced temperature an/or pH can minimize lactate and ammonia production.
  • growing cells at a reduced temperature and/or pH results in minimal reduction in cumulative average productivity and product titer.
  • Titer refers to the total amount of polypeptide of interest, e.g., a glycoprotein of interest, produced by a cell culture (e.g., an animal cell culture), divided by a given amount of medium volume; thus "titer” refers to a concentration. Titer is typically expressed in units of milligrams of polypeptide per liter of medium.
  • any reduction in cell productivity and product titer experienced in the cell culture is offset by decreased production of aggregated or misfolded protein product, e.g., decreased production of HMWA.
  • aggregated protein refers to the protein groupings, i.e., high molecular weight aggregates, that produce nonfunctional, sub-optimal or undesired protein product.
  • misfolded protein refers to an improperly folded protein, often a protein that can no longer display normal biological activity, e.g., normal enzymatic activity.
  • protein aggregates may comprise either or both correctly folded and/or misfolded proteins.
  • Aggregated or misfolded proteins are commonly formed in overexpressing cell culture, e.g., a cell culture overexpressing a protein of interest, e.g., a glycoprotein of interest.
  • aggregation can be caused either by nonspecific hydrophobic interactions of unfolded polypeptide chains or by interaction of folding intermediates.
  • growing cells at the reduced temperature and/or reduced pH results in at least 50% reduction in protein misfolding/aggregation, preferably about 60% reduction in protein misfolding/aggregation.
  • hydrophobic interaction HPLC HIC-HPLC
  • HMWA high molecular weight aggregate
  • High molecular weight aggregate refers to undesirable byproduct of protein production which results from association between at least two proteins.
  • “High molecular weight aggregate” may be an association between at least two of the same proteins and/or association between the protein of interest and other proteins found in the cell culture, e.g., host cell proteins.
  • the association may arise by any method including, but not limited to, covalent, noncovalent, disulfide, and/or nonreducible crosslinking.
  • a protein that is active in a multimer form is a receptor, e.g., a cytokine receptor (e.g., sIL-13R).
  • a cytokine receptor e.g., sIL-13R
  • growing cells at a reduced temperature and/or pH results in at least about a 10% reduction, preferably about a 40% reduction, more preferably about a 50% reduction, and even more preferably about an 80% or more reduction in high molecular weight aggregates, or any intermediate values.
  • a 10% reduction preferably about a 40% reduction, more preferably about a 50% reduction, and even more preferably about an 80% or more reduction in high molecular weight aggregates, or any intermediate values.
  • SEC-HPLC size- exclusion high performance liquid chromatography
  • temperature and/or pH in a cell culture are used to change protein glycosylation, e.g., protein sialylation, to a predetermined level.
  • decreasing pH and/or temperature of the cell culture may lead to the decrease in both N- and 0-linked glycosylation, e.g., decrease in iV-glycan sialylation).
  • Change in protein glycosylation e.g., sialylation
  • One skilled in the art may monitor the degree of glycosylation (e.g., sialylation) to ensure that the changing temperature and/or pH achieves a desired type and level of protein glycosylation.
  • Chromatographic techniques e.g., Normal Phase Chromatography (NPC) may be used to monitor protein glycosylation of the protein product.
  • the cells are harvested and the polypeptide of interest is collected and purified.
  • the polypeptide of interest displays reduced misfolding and/or aggregation, while retaining an acceptable glycosylation pattern.
  • the polypeptide of interest at the end of the production process is in a soluble form (e.g., the polypeptide of interest is a soluble receptor, e.g., a soluble cytokine receptor).
  • soluble forms of the polypeptide can be purified from conditioned media.
  • Membrane-bound forms of the polypeptide can be purified by preparing a total membrane fraction from the expressing cells and extracting the membranes with a nonionic detergent such as TRITON ® X-IOO (EMD Biosciences, San Diego, CA). Cytosolic or nuclear proteins may be prepared by lysing the host cells (via mechanical force, Parr-bomb, sonication, detergent, etc.), removing the cell membrane fraction by centrifugation, and retaining the supernatant. [0098] The polypeptide can be purified using other methods known to those skilled in the art.
  • a polypeptide produced by the disclosed methods can be concentrated using a commercially available protein concentration filter, for example, an AMICON ® or PELLICON ® ultrafiltration unit (Millipore, Billerica, MA).
  • the concentrate can be applied to a purification matrix such as a gel filtration medium.
  • an anion exchange resin e.g., a MonoQ column, Amersham Biosciences, Piscataway, NJ
  • such resin contains a matrix or substrate having pendant diethylaminoethyl (DEAE) or polyethylenimine (PEI) groups.
  • the matrices used for purification can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step may be used for purification of proteins.
  • Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups (e.g., S-SEPHAROSE ® columns, Sigma-Aldrich, St. Louis, MO).
  • the purification of the polypeptide from the culture supernatant may also include one or more column steps over affinity resins, such as concanavalin A- agarose, AF-HEPARIN650, heparin-TOYOPEARL ® or Cibacron blue 3GA SEPHAROSE ® (Tosoh Biosciences, San Francisco, CA); hydrophobic interaction chromatography columns using such resins as phenyl ether, butyl ether, or propyl ether; or immunoaffinity columns using antibodies to the labeled protein.
  • affinity resins such as concanavalin A- agarose, AF-HEPARIN650, heparin-TOYOPEARL ® or Cibacron blue 3GA SEPHAROSE ® (Tosoh Biosciences, San Francisco, CA)
  • one or more HPLC steps employing hydrophobic HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups (e.g., Ni-NTA columns), can be employed to further purify the protein.
  • the polypeptides may be recombinantly expressed in a form that facilitates purification.
  • the polypeptides may be expressed as a fusion with proteins such as maltose-binding protein (MBP), glutathione-5-transferase (GST), or thioredoxin (TRX); kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ), and Invitrogen (Carlsbad, CA), respectively.
  • MBP maltose-binding protein
  • GST glutathione-5-transferase
  • TRX thioredoxin
  • kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ), and Invitrogen (Carlsbad, CA), respectively.
  • the proteins can also be tagged with a small epitope (e.g., His, myc or Flag tags) and subsequently identified or purified using a specific antibody to the chosen epitope.
  • proteins produced according to one or more methods of the present invention can be useful in the preparation of pharmaceuticals.
  • Proteins produced according to one or more methods of the present invention may be administered to a subject or may first be formulated for delivery by any available route including, but not limited to, e.g., parenteral (e.g., intravenous), intradermal, subcutaneous, oral, nasal, bronchial, ophthalmic, transdermal (topical), transmucosal, rectal, and vaginal routes.
  • compositions typically include a purified protein expressed from a mammalian cell line, a delivery agent (e.g., a cationic polymer, peptide molecular transporter, surfactant, etc., as described above) in combination with a pharmaceutically acceptable carrier.
  • a delivery agent e.g., a cationic polymer, peptide molecular transporter, surfactant, etc., as described above
  • pharmaceutically acceptable carrier includes nontoxic materials that do not interfere with the effectiveness of the biological activity of the active ingredient(s), e.g., solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The characteristics of the carrier will depend on the route of administration. Supplementary active compounds can also be incorporated into the compositions.
  • a pharmaceutical composition is formulated to be compatible with its intended route of administration.
  • the pharmaceutical composition When the therapeutic protein produced according to one or more methods of the present invention is administered in an oral form, the pharmaceutical composition will be in the form of a tablet, capsule, powder, solution or elixir.
  • the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant.
  • the tablet, capsule, and powder will contain from about 5 to 95% binding agent, and preferably from about 25 to 90% binding agent.
  • a liquid carrier such as water, petroleum, oils of animal or plant origin, such as sesame oil, peanut oil (taking into consideration the occurrence of allergic reactions in the population), mineral oil, or soybean oil, or sesame oil, or synthetic oils may be added.
  • the liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol.
  • the pharmaceutical composition contains from about 0.5 to 90% by weight of the binding agent, and preferably from about 1 to 50% by weight of the binding agent.
  • the therapeutic protein produced according to one or more methods of the present invention is administered by intravenous, cutaneous or subcutaneous injection
  • the therapeutic protein will be in the form of a pyrogen- free, parenterally acceptable aqueous solution.
  • a preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the therapeutic protein, an isotonic vehicle such as sodium chloride injection, Ringer's injection, dextrose injection, dextrose and sodium chloride injection, lactated Ringer's injection, or other vehicle as known in the art.
  • the pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additive known to those of skill in the art.
  • Example 1.1 Effects of Reduced Temperature on Cell Culture Performance of Recombinant CHO Cells and on Product Quality of TNFR-Fc Fusion Protein
  • Chinese Hamster Ovary (CHO) cells overexpressing recombinant glycoprotein TNFR-Fc were inoculated at identical concentrations and conditions in four separate bench-scale bioreactors at 37.O 0 C. Cells were grown in a fed- batch culture for one day, followed by a temperature- shift to either 30.0 0 C, 29.O 0 C, 28.O 0 C, or 27.O 0 C. The initial pH set point lower limit of the culture was 7.00.
  • a cell sample was removed from the culture each day to measure various conditions of the cells culture, e.g., integrated viable cell numbers, cell viabilities, residual glucose profile, residual glutamine profile, lactate and ammonia concentrations, cumulative average cell productivity (Qp), product titer, levels of misfolded / aggregated product, levels of high molecular weight aggregates, levels of sialylation, and pH.
  • various conditions of the cells culture e.g., integrated viable cell numbers, cell viabilities, residual glucose profile, residual glutamine profile, lactate and ammonia concentrations, cumulative average cell productivity (Qp), product titer, levels of misfolded / aggregated product, levels of high molecular weight aggregates, levels of sialylation, and pH.
  • IVC Integrated viable cell numbers
  • Cell viabilities were measured by staining with Trypan blue using cell- density examination (e.g., CEDEX®, Innovatis, Malvern, PA). Additionally, the residual glucose profile and the residual glutamine profile in the cell culture were measured using the BioProfile Chemistry Analyzer (Nova Biomedical, Waltham, MA). Concentrations of the waste products, lactate and ammonia, were measured using the BioProfile Chemistry Analyzer (Nova Biomedical, Waltham, MA).
  • Cumulative average cell productivity (Qp) and product titer were measured using protein A HPLC and were normalized to the average harvest day cumulative average Qp and titer.
  • the average harvest day titer was determined by calculating the arithmetic mean of the harvest day (e.g., day 10) titer for all experimental conditions tested. All individual titer values were then scaled to the average harvest day titer to generate normalized values.
  • the average harvest day cumulative average Qp was determined by calculating the arithmetic mean of the harvest day cumulative average Qp for all experimental conditions tested. All individual values were then scaled to the average harvest day titer to generate normalized values.
  • Levels of misfolded / aggregated product were measured using HIC- HPLC, and levels of high molecular weight aggregates were measured using SEC-HPLC.
  • Levels of total and iV-linked glycan sialylation were measured by subjecting 2-aminobenzamide- (2-AB)-labeled protein glycoforms to Normal Phase Chromatography (NPC).
  • NPC Normal Phase Chromatography
  • the amount of total sialylation (N- and O- linked) was defined as the percentage relative to the amount of total sialylation of reference material, i.e., TNFR-Fc aliquot with known and preferable sialylation pattern.
  • pH of the cell culture was determined by an off-line measurement via blood gas analyzer (Bayer Rapidlab 248 pH/blood gas analyzer (Bayer Healthcare LLC, East Walpole, MA)).
  • lactic acid and ammonia may also be consumed by the cell culture.
  • the cessation of the production of lactic acid and ammonia or the consumption of lactic acid and ammonia promote cell viability, cell productivity, and can have an effect of increasing polypeptide product titer.
  • Growing cells at the reduced temperature had an effect of altering the net consumption of lactate in the later half of the fed-batch culture (FIG. 3A).
  • Reduced temperatures also resulted in higher ammonia production (FIG. 3B).
  • Reduced temperatures resulted in lower cell-specific productivities (lower Qp) (FIG. 4A) and lower product titer (FIG. 4B).
  • Example 1.2 Effects of Reduced pH on Cell Culture Performance of Recombinant CHO Cells and on Product Quality of TNFR-Fc Fusion Protein
  • CHO cells overexpressing recombinant glycoprotein TNFR-Fc were inoculated at identical concentrations at 37.O 0 C and at pH set point of either 7.20, 7.10, 7.00, 6.90 or 6.80. Addition of sodium carbonate base was utilized to prevent cultures from deviating below the set point pH. No pH control was utilized above the set point pH. Bioreactors were temperature- shifted to 30.0 0 C on day 1 (one day post inoculation, i.e., one day after the start of the experiment). Various conditions of the cell culture were measured as described in Example 1.1.1.
  • pH of the cell culture must be chosen in such as way that the detrimental effects on protein glycosylation are balanced by the beneficial effects on reduction of misfolded and/or aggregated proteins. For instance, growing cells at a reduced pH of 6.95 has significantly reduced the proportion of misfolded and aggregated TNFR-Fc protein, while having minimal effects on glycosylation of TNFR-Fc.
  • Stably transfected Chinese Hamster Ovary (CHO) cells overexpressing recombinant glycoprotein sIL-13R were seeded in Applikon bioreactors at 3x10 5 cells/mL in an inoculation medium with Antifoam (Dow Corning Corporation, Midland, MI). Additional Antifoam was added as required. A concentrated nutrient medium was used as a feed medium. The pH set point lower limit was 6.80. The d ⁇ 2 set point was 23% utilizing 7% CO 2 /93% air sparge gas, while agitation was 200 rpm. Bioreactors were temperature- shifted from 37.O 0 C on day 5.
  • the production phase temperature was either 37.O 0 C, 33.O 0 C, 32.O 0 C, 28.O 0 C, or room temperature (RT; about 24.O 0 C).
  • Feeding schedules for experimental conditions are summarized in Table 1. Feed volumes are listed as percentage of culture volume in bioreactor. The temperature shift for cell cultures occurred at about 6x10 6 cells/mL.
  • the viable cell densities achieved after the temperature shift were slightly lower at RT (about 6xl0 6 cells/mL) and 29.O 0 C (about 7xl0 6 cells/mL) than at higher temperatures (about 8xl0 6 cells/mL) (FIG. 17A).
  • the total cell densities achieved after the temperature shift were slightly lower at RT (about 6.5xl0 6 cells/mL) and 29.O 0 C (about 7.5xlO 6 cells/mL) than at higher temperatures (about 8 to 9xl0 6 cells/mL) (FIG. 17B). Viabilities were better maintained throughout the fed-batch at lower temperatures (FIG.
  • the highest titer (combined sIL-13R dimer and HMWA titer) was achieved by the 31.O 0 C culture followed by the 32.O 0 C culture (188 mg/L), 29.O 0 C culture (178 mg/L), and 33.O 0 C culture (151 mg/L) (FIG. 18).
  • the RT and 37.O 0 C cultures had significantly lower titers.
  • the cell specific productivity, or daily specific sIL-13R production rate (Qp), of the sIL-13R-producing cell line was significantly diminished at RT and 37.O 0 C (FIG. 19A). Additionally, the cumulative average specific productivities, or cumulative average Qp, were also lower at 37.O 0 C and RT (FIG. 19B).
  • Lactate concentration profiles for the RT, 29.O 0 C, 31.O 0 C, and 32.O 0 C cultures were similar with lactate concentration peaking on or around the day of the temperature shift, then decreasing to between 0.7 and 2.0 g/L by day 18 (FIG. 22).
  • the lactate profiles were similar to the other cultures during the growth phase, but were significantly different during the production phase.
  • lactate concentration did not decrease after the temperature shift, and instead remained essentially constant.
  • lactate increased throughout the entire duration of the fed-batch.
  • the ammonia concentration peaked on or around the day of the temperature shift, decreased during the middle stage of the fed-batch, and then increased during the late stage of the fed-batch.
  • the increase in ammonia concentration in the late stage of the fed-batch was greater and occurred sooner.
  • ammonia concentration increased throughout the entire course of the fed-batch.
  • the ammonia levels remained approximately constant after the temperature shift.
  • HMWA high molecular weight aggregates
  • SEC size-exclusion chromatography

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides a novel method of reducing protein misfolding and aggregation in the cell culture by growing the cell culture at a reduced temperature and/or reduced pH. As a result, the quality of the protein produced in the cell culture is greatly improved. Thus, the present invention facilitates improvements in the efficacy of therapeutic proteins produced in cell culture.

Description

TΓΓLE
USE OF LOW TEMPERATURE AND/OR LOW PH IN CELL CULTURE
[0001] This application claims the benefit of U.S. Provisional Application Ser. No. 60/913,382, filed April 23, 2007, which is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
Field of the Invention
[0002] The present invention provides methods of improving protein production by cultured cells, especially mammalian cells. Specifically, the present invention relates to methods of preparing a protein product(s), e.g., a glycoprotein product(s), wherein the protein product characteristics are controlled by manipulating the cell culture environment. The invention also relates to methods of improving the therapeutic efficacy and/or immunogenicity of the protein product(s), e.g., the glycoprotein product(s), produced in mammalian cells, e.g., by manipulating protein glycosylation and decreasing protein aggregation and misfolding, by reducing the temperature and/or the pH of the cell culture.
Related Background Art
[0003] A large proportion of biotechnology products, whether commercially available or in development, are protein therapeutics. There is a large and increasing demand for production of proteins in animal cell cultures and for improved methods related to such production. Such improved methods are needed because the cellular machinery of an animal cell is generally required to produce many forms of protein therapeutics, such as posttranslationally modified proteins, particularly glycosylated proteins.
[0004] A common problem encountered in large-scale therapeutic protein production methods is that a significant portion of protein product is produced in either misfolded or aggregated form, i.e., in high molecular weight aggregate ("HMWA") form. For instance, recombinant overexpression of polypeptides in cells may overload the endoplasmic reticulum (ER) machinery, allowing an increased number of misfolded and/or aggregated proteins to evade degradative pathways and to exit the ER. Thus, current protein production methods may result in a large proportion of product which is aggregated, nonfunctional, and thus, unusable as produced. The presence of misfolded and/or aggregated protein is undesirable because it may lead to adverse events upon administration, including but not limited to, a potential for immunogenicity upon administration (e.g., complement activation or anaphylaxis). Thus, excessive therapeutic protein misfolding and/or aggregation may lead to failure in, e.g., clinical trials. Therefore, there is a need in the art for new methods of limiting or reducing protein misfolding and/or aggregation.
[0005] Protein glycosylation is a common posttranslational modification process by which complex sugar moieties are added onto a protein through the actions of a series of specialized enzymes - glycotransferases and glycosidases - in the ER. This process can control proper folding of the newly synthesized polypeptides such that only the correctly folded polypeptides exit the ER, whereas the misfolded proteins are degraded. Patterns of protein glycosylation affect protein targeting, structure, thermodynamic stability, and enzymatic activity (see, e.g., Sola et al. (2007) Cell. MoI. Life ScL 64:2133-52; Sola and Griebenow (2006) FEBS Lett. 580:1685-90). For instance, iV-glycan sialylation is associated with an increase in the lifetime of glycoproteins because such glycoproteins are not recognized by asialo glycoprotein receptors, which target nonsialylated proteins for degradation (see, e.g., Bork et al. (2007) FEBS Letters 581:4195-98). Thus, alterations in protein glycosylation may affect the quality and efficacy of the product.
[0006] Moreover, aberrant protein glycosylation may result in immunogenicity of the final therapeutic protein product. Proteins produced under nonnatural, suboptimal conditions can acquire sugars and sugar patterns not found naturally on human proteins, and thus result in immunogenic reaction in the subject
(Jefferis (2006) Biotechnol. Prog. 21:11-16). Such nonnatural glycosylations are especially prevalent in protein therapeutics, such as antibody therapeutics or
Fc-fusion protein therapeutics, e.g., soluble receptor Fc-fusion protein therapeutics.
[0007] For these reasons, the FDA requires that glycoform profiles of therapeutic proteins be maintained within strict limits. Thus, there is a need in the pharmaceutical industry for a method of therapeutic protein production in the cell culture that enables control of the level of protein glycosylation.
SUMMARY OF THE INVENTION
[0008] Thus, the invention is a method of producing a protein in a cell culture comprising at least one of: (a) growing cells in the cell culture at a reduced temperature; and (b) growing cells in the cell culture at a reduced pH; so that production of misfolded proteins and/or aggregated proteins is reduced. In embodiments, cells are grown in the cell culture at a reduced temperature and at a reduced pH.
[0009] In preferred embodiments, the invention is directed to modifying pH and temperature parameters to reduce production of misfolded and/or aggregated proteins in mammalian cell culture, and especially Chinese Hamster Ovary ("CHO") cell culture. In preferred embodiments the cell culture produces a protein (the "produced protein") that is a soluble receptor, such as, without limitation TNFR-Fc or sIL-13R proteins. In these preferred embodiments, the reduced temperature may be in a range of 27.O0C to less than 30.00C. In still other preferred embodiment, the reduced pH is in a range of 6.80 to less than 7.00. A combination of pH and temperature in the aforesaid ranges may also be used. [0010] In another aspect, the invention is a method of producing a protein in a cell culture, wherein the level of protein glycosylation of the produced protein is controlled by modifying the temperature and/or the pH of the cell culture. Thus, the level of glycosylation, such as, without limitation, iV-glycan sialylation, may be increased by increasing temperature and/or pH, or decreased by decreasing temperature and/or pH.
[0011] In another aspect, the invention is a method of producing a therapeutic protein controlling the parameters described above. In still another aspect, the invention is a pharmaceutical composition comprising a therapeutic protein made by such method and a pharmaceutically acceptable carrier.
BRIEF DESCRIPTION OF THE DRAWINGS
[0012] FIG. IA represents integrated viable cell numbers (Y-axis; IVC [normalized e9 cells * day/L]), normalized to average harvest day IVC, of CHO cells transfected with TNFR-Fc grown either at 27.O0C [♦], 28.O0C [A], 29.O0C [ ■ ], or 30.00C[O] over time (X-axis; culture time in days [d]); FIG. IB represents cell viability (Y-axis) of the same cells over time (X-axis; culture time [d]).
[0013] FIG. 2A represents the residual glucose profile (Y-axis; glucose [g/L]) in the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O0C [♦], 28.O0C [A], 29.O0C [ ■ ], or 30.00C[O] over time (X-axis; culture time [d]); FIG. 2B represents the glutamine profile (Y-axis; glutamine [mM]) of the same cells over time (X-axis; culture time [d]).
[0014] FIG. 3A represents lactate concentration in the media (Y-axis; lactate [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O0C [♦], 28.O0C [A], 29.O0C [ ■ ], or 30.00C[O] over time (X-axis; culture time [d]); FIG. 3B represents the ammonium profile (Y-axis; NH4 + [mM]) of the same cells over time (X-axis; culture time [d]).
[0015] FIG. 4A depicts cell-specific productivities, represented by cumulative average Qp (X-axis; Cum. Avg. Qp [normalized mg/e9 cells/day]), normalized to average harvest day cumulative average Qp, of the cell culture of CHO cells transfected with TNFR-Fc grown either at 27.O0C [♦], 28.O0C [A], 29.O0C [ ■ ], or 30.00C[O] over time (X-axis; culture time [d]); while FIG. 4B represents TNFR-Fc titer (X-axis; product titer [normalized mg/L]), normalized to average harvest day titer, of the same cells over time (culture time [d]). [0016] The effect of varying production temperatures (X-axis; [0C]) in the cell culture of CHO cells transfected with TNFR-Fc on the production of misfolded and/or aggregated TNFR-Fc (Y-axis; % misfolded / aggregated product) is demonstrated in FIG. 5A; while the effect of temperatures on the production of HMWA (Y-axis; % high molecular weight species) is demonstrated in FIG. 5B. [0017] The effect of varying production temperatures (X-axis; temperature [0C]) in the cell culture of CHO cells transfected with TNFR-Fc on the percentage of total sialylation (N- and 0-linked sialylation) of TNFR-Fc (Y-axis; percentage of total sialylation of reference material) is demonstrated in FIG. 6A. The reference material used was a specific aliquot of TNFR-Fc with known and preferable glycosylation pattern to which the assay results may be compared. FIG. 6B demonstrates the effect of varying production temperatures (X-axis; temperature [0C]) in the cell culture of the same cells on the percentage of total sialylated (D) or non-sialylated (•) iV-linked glycans (Y-axis; Percentage of Total iV-linked Glycans).
[0018] FIG. 7 demonstrates the effect of growing cells at different temperatures (27.O0C [♦], 28.O0C [A], 29.O0C [ ■ ], or 30.00C[O]) on the pH of the CHO cell culture (Y-axis) over time (X-axis; culture time [d]). [0019] FIG. 8A represents integrated viable cell numbers (Y-axis; IVC [normalized e9 cells *day/L]), normalized to average harvest day IVC, of the CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]); while FIG. 8B represents cell viability (Y-axis) of the same cells over time (X-axis; culture time [d]).
[0020] FIG. 9A represents glucose profile (Y-axis; glucose [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc, grown at either pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]); while FIG. 9B represents glutamine profile (Y-axis; glutamine [mM]) of the same cells over time (X-axis; culture time [d]). [0021] FIG. 1OA represents lactate concentration in the media (Y-axis; lactate [g/L]) of the cell culture of CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X- axis; culture time [d]); while FIG. 1OB represents ammonium profile (Y-axis; NH4 + [mM]) of the same cells over time (X-axis; culture time [d]). [0022] FIG. HA represents deviations of the pH of the cell culture from the pH set point (X-axis; pH of culture), where the CHO cells transfected with TNFR-Fc were grown at a pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]). FIG. HB represents osmolality (X-axis; osmolality [mOsm/kg]) of the same cells grown at a pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]).
[0023] FIG. 12A represents cell specific productivities, represented by cumulative average Qp (X-axis; Cum. Avg. Qp [normalized mg/e9 cells/day]), normalized to average harvest day cumulative average Qp, of the cell culture of CHO cells transfected with TNFR-Fc grown at pH set point of either 7.20 [O], 7.10 [ ■ ], 7.00 [♦], 6.90 [•], or 6.80 [A] over time (X-axis; culture time [d]).; while FIG. 12B represents TNFR-Fc titer (X-axis; product titer [normalized mg/L]), normalized to average harvest day titer, of the same cells over time (culture time [d]).
[0024] FIG. 13A represents the effect of varying the pH set point of the culture (X-axis; pH) of CHO cells transfected with TNFR-Fc on the production of misfolded / aggregated TNFR-Fc (Y-axis; % misfolded / aggregated product). FIG. 13B represents the effect of varying the pH set point of the cell culture (X- axis; pH) on the production of HMWA (Y-axis; % high molecular weigh species).
[0025] The effect of varying the pH set point (X-axis; pH) in the culture of CHO cells transfected with the TNFR-Fc on the percentage of total sialylation of TNFR-Fc (Y-axis; percentage of total sialylation of reference material) is demonstrated in FIG. 14A. Fig. 14B demonstrates the effect of varying cell culture pH set point (X-axis; pH) in the cell culture of the same cells on the percentage of total sialylated (D) or non-sialylated (•) iV-linked glycans (Y-axis;
Percentage of Total iV-linked Glycans)
[0026] FIG. 15 depicts a typical fluorescence (Y-axis; fluorescence, measured in mV) versus retention time (X-axis; minutes) profile depicting iV-linked glycan sialylation as observed by subjecting hydrazaine released 2-aminobenzamide-
(2-AB)-labeled protein glycoforms to Normal Phase Chromatography.
[0027] FIG. 16 depicts the fluorescence (Y-axis; mV) versus retention time (X- axis; minutes) profiles depicting iV-linked glycan sialylation as observed by subjecting hydrazaine released 2-aminobenzamide- (2-AB)-labeled protein glycoforms to Normal Phase Chromatography of cell culture of CHO cells transfected with TNFR-Fc at varying pH set points of the culture.
[0028] FIG. 17 represents (A) viable cell density (Y-axis; cells/mL), (B) total cell density , which includes both viable and non-viable cells, (Y-axis; cells/mL),
(C) cell viability (Y-axis), and (D) integrated viable cell numbers (IVC) (Y-axis; e9 cells*day/L) of the cell culture of sIL-13R overexpressing cells grown at
37.O0C [♦], 33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X-axis; culture time [d]). .
[0029] FIG. 18 represents sIL-13R titer, both dimer and HMWA, (Y-axis; sIL-
13R [mg/L]) for cell cultures overexpressing sIL-13R, grown at 37.O0C [♦],
33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X-axis; culture time [d]).
[0030] FIG. 19A represents daily specific sIL-13R production rate (Y-axis; Qp
[ug/e9 cells/d]) during various time intervals (X-axis) for cell cultures overexpressing sIL-13R,grown at 37.O0C , 33.O0C , 32.O0C , 31.O0C , 29.O0C , or
RT. FIG. 19B represents cumulative average cell specific productivities (Y- axis; Cum. Avg. Qp [mg/e9 cells/day]) over time (X-axis; culture time [d]) for cell cultures overexpressing sIL-13R, grown at 37.O0C, 33.O0C, 32.O0C, 31.O0C,
29.O0C, or RT.
[0031] FIG. 20 represents daily specific glucose consumption rate (Y-axis; QgIc
[g/e9 cell/d]) for different time intervals (X-axis) for cell cultures overexpressing sIL- 13R, grown at 37.O0C, 33.O0C, 32.O0C, 31.O0C, 29.O0C, or RT [0032] FIG. 21 represents daily specific glutamine consumption rate (Y-axis; QgIn [mmol/e9 cell/d]) for different time intervals (X-axis) for cell cultures overexpressing sIL-13R, grown at 37.O0C, 33.O0C, 32.O0C, 31.O0C, 29.O0C, or RT.
[0033] FIG. 22 represents lactate concentration (Y-axis; lactate[g/L]) in the cell culture medium of the cells overexpressing sIL-13R, grown at 37.O0C [♦], 33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X-axis; culture time [d]).
[0034] FIG. 23 represents ammonium concentration (Y-axis; ammonia [mM]) in the cell culture medium of the cells overexpressing sIL-13R, grown at 37.O0C [♦], 33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X- axis; culture time [d]).
[0035] FIG. 24 represents the effect of cell culture temperature (X-axis; production temperature [0C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 9 of culture of cells overexpressing sIL-13R. [0036] FIG. 25 represents the effect of cell culture temperature (X-axis; production temperature [0C]) on the production of HMWA (Y-axis; % High Molecular Weight Species) on day 18 of culture of cells overexpressing sIL-13R. [0037] FIG. 26A represents percentage of the sIL-13R dimer (Y-axis; % sIL- 13R dimer) recovered from the total sIL-13R protein produced in the conditioned media of sIL-13R overexpressing cells grown at either 37.O0C [♦], 33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X-axis; culture time [d]). FIG. 26B represents the percentage of HMWA (Y-axis; % High Molecular Weight Species) relative to total sIL-13R in the conditioned medium of sIL-13R overexpressing cells over time (X-axis; culture time [d]).
[0038] FIG. 27 represents the sIL-13R dimer titer (Y-axis; sIL-13R dimer only [mg/L]) in the conditioned media of cells grown either 37.O0C [♦], 33.O0C [ ■ ], 32.O0C [•], 31.O0C [O], 29.O0C [Δ], or RT [D] over time (X-axis; culture time [d]). DETAILED DESCRIPTION OF THE INVENTION [0039] The prevailing wisdom for producing therapeutic proteins in a mammalian cell culture is that the temperature during the production phase should be at least 30.00C and the pH should be at least 7.00. However, growing cells in a cell culture during a production phase at temperatures above 30.00C and at a pH above 7.00 can lead to increased protein aggregation (also referred to herein as high molecular weight aggregate, or HMWA, formation) and protein misfolding, and thus production of lower quantity of functional and usable protein.
[0040] The present invention provides a novel method of protein production in a cell culture, wherein the method results in decreased protein misfolding and/or decreased protein aggregation. In another aspect the invention provides for methods of controlling the level of protein glycosylation.
Proteins Produced by Methods of the Invention
[0041] As used herein, the phrases "polypeptide" or "polypeptide product" are synonymous with the terms "protein" and "protein product," respectively, and, as is generally understood in the art, refer to at least one chain of amino acids linked via sequential peptide bonds. In certain embodiments, a "protein of interest" or a "polypeptide of interest" or the like is a protein encoded by an exogenous nucleic acid molecule that has been transfected or transformed into a host cell, e.g., transiently or stably transfected or transformed into a host cell. In certain embodiments, wherein an exogenous DNA with which the host cell has been transfected or transformed codes for the "protein of interest," the nucleic acid sequence of the exogenous DNA determines the sequence of amino acids. This sequence may be a sequence that occurs in nature, or may alternatively be a sequence engineered by man. In certain embodiments, a "protein of interest" is a protein encoded by a nucleic acid molecule that is endogenous to the host cell. The expression of such an endogenous protein of interest may be altered by transfecting a host cell with an exogenous nucleic acid molecule that may, for example, contain one or more regulatory sequences and/or encode a protein that enhances expression of the protein of interest. In the embodiments of the present invention, a polypeptide of interest is produced in the cell culture, e.g., for subsequent purification.
[0042] The term "glycoprotein," "glycosylated protein" and the like refers to proteins that have sugar moieties, e.g., oligosaccharide moieties, attached to either their asparagine side chains (iV-glycosylation) or their serine and/or threonine side chains (0-glycosylation). For instance, one common type of protein iV-glycosylation is protein sialylation (also known as iV-glycan sialylation). The majority of secretory and membrane proteins (e.g., membrane receptors) are glycosylated in the ER and/or the Golgi apparatus. It is known in the art that glycosylation controls protein folding and release in the ER. Additionally, multiple steps of glycosylation / deglycosylation occur in the Golgi. Present understanding of protein glycosylation processes in both ER and Golgi are reviewed in Helenius et al. (2001) Science 291:2364-69; Parodi (2000) Biochem. J. 348:1-13; and Parodi (2000) Annu. Rev. Biochem. 69:69-93. Thus, in certain embodiments of the present invention, the polypeptide of interest is a glycoprotein of interest, and the glycoprotein of interest is produced in the cell culture, e.g., for subsequent purification. In one embodiment of the present invention, the glycoprotein of interest is a receptor, and may be a soluble receptor.
[0043] Methods and compositions of the present invention may be used to produce any protein of interest including, but not limited to, proteins having pharmaceutical, diagnostic, agricultural, and/or any of a variety of other properties that are useful in commercial, experimental and/or other applications. In addition, a protein of interest can be a protein therapeutic. Namely, a protein therapeutic (or a therapeutic protein) is a protein that has a biological effect on a region in the body on which it acts or on a region of the body on which it remotely acts via intermediates. In certain embodiments, proteins produced using methods and/or compositions of the present invention may be processed and/or modified, before administering to a subject as a therapeutic protein. [0044] The present invention may be used to culture cells for the advantageous production of any therapeutic protein, such as pharmaceutically or commercially relevant enzymes, receptors, receptor fusions, soluble receptors, soluble receptor fusions, antibodies (e.g., monoclonal and/or polyclonal antibodies), antigen- binding fragments of an antibody, Fc fusion proteins, SMIPs, cytokines, hormones, regulatory factors, growth factors, coagulation / clotting factors, or antigen-binding agents. The above list of proteins is merely exemplary in nature, and is not intended to be a limiting recitation. One of ordinary skill in the art will know of other proteins that can be produced in accordance with the present invention, and will be able to use methods disclosed herein to produce such proteins.
[0045] The term "antibody" includes a protein comprising at least one, and typically two, VH domains or portions thereof, and/or at least one, and typically two, VL domains or portions thereof. In certain embodiments, the antibody is a tetramer of two heavy immunoglobulin chains and two light immunoglobulin chains, wherein the heavy and light immunoglobulin chains are inter-connected by, e.g., disulfide bonds. The antibodies, or a portion thereof, can be obtained from any origin, including, but not limited to, rodent, primate (e.g., human and non-human primate), camelid, shark as well as recombinantly produced, e.g., chimeric, humanized, and/or in vitro generated, e.g., by methods well known to those of skill in the art.
[0046] This invention also encompasses "antigen-binding fragments of antibodies", which include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHl domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHl domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment, which consists of a VH domain; (vi) a camelid or camelized variable domain, e.g., a VHH domain; (vii) a single chain Fv (scFv); (viii) a bispecific antibody; and (ix) one or more antigen binding fragments of an immunoglobulin fused to an Fc region. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-26; Huston et al. (1988) Proc. Natl. Acad. ScL U.S.A. 85:5879-83). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody. These antibody fragments are obtained using conventional techniques known to those skilled in the art, and the fragments are evaluated for function in the same manner as are intact antibodies. [0047] The invention also encompasses single domain antibodies. Single domain antibodies can include antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, goat, rabbit, cow and shark. According to one aspect of the invention, a single domain antibody as used herein is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 9404678 for example. For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention. Single domain antibodies also include shark IgNARs; see, e.g., Dooley et al., Proc. Natl. Acad. ScL U.S.A., 103:1846-1851 (2006). [0048] Other than "bispecific" or "bifunctional" antibodies, an antibody is understood to have each of its binding sites identical. A "bispecific" or "bifunctional antibody" is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315- 321 (1990); Kostelny et al, J. Immunol. 148, 1547-1553 (1992). [0049] In embodiments where the protein is an antibody or a fragment thereof, it can include at least one, or two full-length heavy chains, and at least one, or two light chains. Alternatively, the antibodies or fragments thereof can include only an antigen-binding fragment (e.g., an Fab, F(ab')2, Fv or a single chain Fv fragment). The antibody or fragment thereof can be a monoclonal or single specificity antibody. The antibody or fragment thereof can also be a human, humanized, chimeric, CDR-grafted, or in vitro generated antibody. In yet other embodiments, the antibody has a heavy chain constant region chosen from, e.g., IgGl, IgG2, IgG3, or IgG4. In another embodiment, the antibody has a light chain chosen from, e.g., kappa or lambda. In one embodiment, the constant region is altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function). Typically, the antibody or fragment thereof specifically binds to a predetermined antigen, e.g., an antigen associated with a disorder, e.g., a neurodegenerative, metabolic, inflammatory, autoimmune and/or a malignant disorder. [0050] Proteins described herein, optionally, further include a moiety that enhances one or more of, e.g., stability, effector cell function or complement fixation. For example, an antibody or antigen-binding protein can further include a pegylated moiety, albumin, or a heavy and/or a light chain constant region. [0051] Antibodies are generally made, for example, via traditional hybridoma techniques (Kohler et al., Nature 256:495 499 (1975)), recombinant DNA methods (U.S. Patent No. 4,816,567), or phage display techniques using antibody libraries (Clackson et al., Nature 352:624 628 (1991); Marks et al., J. MoI. Biol. 222:581 597 (1991)). For various other antibody production techniques, see Antibodies: A Laboratory Manual, eds. Harlow et al., Cold Spring Harbor Laboratory, 1988.
[0052] Further, the antibodies may be tagged with a detectable or functional label. These labels include radiolabels (e.g., 1311 or 99Tc), enzymatic labels (e.g., horseradish peroxidase or alkaline phosphatase), and other chemical moieties (e.g., biotin).
[0053] "Small Modular Immunopharmaceutical" or (SMIP™) drugs (Trubion Pharmaceuticals, Seattle, WA) are single-chain polypeptides composed of a binding domain for a cognate structure such as an antigen, a counterreceptor or the like, a hinge region polypeptide having either one or no cysteine residues, and immunoglobulin CH2 and CH3 domains (see also www.trubion.com). SMIPs and their uses and applications are disclosed in, e.g., U.S. Published Patent Application Nos. 2007/002159, 2003/0118592, 2003/0133939, 2004/0058445, 2005/0136049, 2005/0175614, 2005/0180970, 2005/0186216, 2005/0202012, 2005/0202023, 2005/0202028, 2005/0202534, and 2005/0238646, and related patent family members thereof, all of which are hereby incorporated by reference herein in their entireties.
[0054] In one embodiment of the invention, the protein of interest is a soluble receptor, e.g., a soluble receptor fusion protein. Membrane proteins, e.g., receptors, are usually glycosylated proteins. Therefore, the methods of the present invention are particularly beneficial in producing nonaggregated, properly folded and glycosylated soluble receptor fusion proteins. [0055] Soluble proteins, e.g., soluble receptors, can be produced according to methods well known in the art. In one embodiment of the invention, a soluble receptor comprises an extracellular region of the receptor, or a fragment of the extracellular region of the receptor. In another embodiment of the invention, a soluble receptor comprises two polypeptides. The first polypeptide comprises a full-length receptor; alternatively, the first polypeptide comprises less than the full length of the receptor, e.g., an extracellular portion of the receptor. In one embodiment of the invention, the first polypeptide is a full-length cytokine receptor; alternatively the first polypeptide is less than the full length of the cytokine receptor, e.g., an extracellular portion of the cytokine receptor. Such a soluble receptor can also comprise an additional polypeptide, e.g., a GST, Lex-A, MBP polypeptide sequence or an immunoglobulin chain, including, e.g., an Fc fragment, a heavy chain constant region of the various isotypes, including: IgGl, IgG2, IgG3, IgG4, IgM, IgAl, IgA2, IgD, and IgE. [0056] In one embodiment of the invention, a second polypeptide is preferably soluble. In some embodiments, the second polypeptide enhances the half-life, (e.g., the serum or circulatory half-life) of the linked polypeptide. In preferred embodiments, the second polypeptide includes at least a region of an immunoglobulin polypeptide. Immunoglobulin fusion polypeptides are known in the art and are described in, e.g., U.S. Patent Nos. 5,516,964; 5,225,538; 5,428,130; 5,514,582; 5,714,147; and 5,455,165. Soluble fusion proteins are known to be susceptible to aggregation during production, and therefore the methods according to the invention provide a particular benefit in connection with cell cultures that produce proteins of this type.
[0057] In some embodiments, the second polypeptide comprises a full-length immunoglobulin polypeptide. Alternatively, the second polypeptide comprises less than the full length of immunoglobulin polypeptide, e.g., a heavy chain, light chain, Fab, Fab2, Fv, or Fc. The second polypeptide can include the heavy chain of an immunoglobulin polypeptide. The second polypeptide can include the Fc region of an immunoglobulin polypeptide.
[0058] In one embodiment of the invention, a soluble receptor fusion protein comprises a tumor necrosis factor inhibitor. In certain embodiments, tumor necrosis factor inhibitors, in the form of tumor necrosis factor alpha and beta receptors (TNFR-I; EP 417,563 published Mar. 20, 1991; and TNFR-2, EP 417,014 published Mar. 20, 1991, each of which is incorporated herein by reference in its entirety) are expressed in accordance with systems and methods of the present invention (for review, see Naismith and Sprang, J. Inflamm. 47(1- 2): 1-7, 1995-96, incorporated herein by reference in its entirety). According to some embodiments, a tumor necrosis factor inhibitor comprises a soluble TNF receptor. In certain embodiments, TNF inhibitors of the present invention are soluble forms of TNFRI and TNFRII. In certain embodiments, TNF inhibitors of the present invention are soluble TNF binding proteins. In certain embodiments, TNF inhibitors of the present invention are TNFR fusion proteins, e.g., TNFR-Ig or TNFR-Fc. As used herein, "etanercept," refers to a TNFR-Fc, which is a dimer of two molecules of the extracellular portion of the p75 TNF-alpha receptor, each molecule consisting of a 235 amino acid Fc portion of human IgGl. In accordance with the invention, cells expressing TNFR-Fc are grown in a cell culture at a reduced temperature and/or reduced pH to decrease the amount of misfolded protein and/or high molecular weight aggregates during the production of TNFR-Fc. In certain embodiments, cells expressing TNFR-Fc are grown in a cell culture at a reduced temperature and/or reduced pH to modulate glycosylation during the production of TNFR-Fc.
[0059] In another embodiment of the invention, the soluble receptor fusion protein is the sIL-13R. As used herein, soluble IL- 13 receptor (sIL-13R) refers to a recombinant fusion protein which includes extracellular domain (ECD) of the human interleukin (IL)-13-alpha2 receptor and the Fc region of the human IgGl heavy chain. sIL-13R is composed of two identical polypeptide chains (i.e., dimer of two polypeptide chains) which appear to be linked by intermolecular disulfide bonds. sIL-13R soluble receptor fusion protein and its uses are disclosed in U.S. Patent No. 5,710,023, incorporated herein in its entirety by reference.
[0060] In some embodiments, the second polypeptide has less effector function than the effector function of an Fc region of a wild-type immunoglobulin heavy chain. Fc effector function includes, for example, Fc receptor binding, complement fixation and T cell-depleting activity (see, for example, U.S. Patent No. 6,136,310). Methods for assaying T cell-depleting activity, Fc effector function, and antibody stability are known in the art. In one embodiment, the second polypeptide has low or no affinity for the Fc receptor. In an alternative embodiment, the second polypeptide has low or no affinity for complement protein CIq.
[0061] The fusion proteins, e.g., soluble receptor fusion proteins, may additionally include a linker sequence joining the soluble receptor or a fragment thereof to the second moiety. For example, the fusion protein can include a peptide linker, e.g., a peptide linker of about 2 to 20, more preferably about 5 to 10, amino acids in length.
[0062] In other embodiments, additional amino acid sequences can be added to the N- or C-terminus of the fusion protein to facilitate expression, detection and/or isolation or purification. For example, soluble receptor fusion protein may be linked to one or more additional moieties, e.g., GST, His6 tag, FLAG tag. For example, the fusion protein may additionally be linked to a GST fusion protein in which the fusion protein sequences are fused to the C-terminus of the GST (i.e., glutathione S-transferase) sequences. Such fusion proteins can facilitate the solubility, i.e., increase accurate folding, and thus improve purification of the fusion protein.
Methods of Protein Production in the Cell Culture
[0063] The terms "culture" and "cell culture" as used herein refer to a cell population that is contacted with a cell culture medium under conditions suitable to survival and/or growth of the cell population. As used herein, these terms may refer to the combination comprising the cell population (e.g., the animal cell culture) and the medium with which the population is in contact. [0064] The cells used in the present invention may be recombinant host cells, e.g., eukaryotic host cells, i.e., cells transfected with an expression construct containing a nucleic acid that encodes a polypeptide of interest, including animal cells. The phrase "animal cells" encompasses invertebrate, nonmammalian vertebrate (e.g., avian, reptile and amphibian), and mammalian cells. Nonlimiting examples of invertebrate cells include the following insect cells: Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori (silkworm / silk moth). In preferred embodiments the cell culture is a mammalian cell culture.
[0065] A number of mammalian cell lines are suitable host cells for recombinant expression of polypeptides of interest. Mammalian host cell lines include, for example, COS, PER.C6, TM4, VERO076, MDCK, BRL-3A, W138, Hep G2, MMT, MRC 5, FS4, CHO, 293T, A431, 3T3, CV-I, C3H10T1/2, Colo205, 293, HeLa, L cells, BHK, HL-60, FRhL-2, U937, HaK, Jurkat cells, Rat2, BaF3, 32D, FDCP-I, PC12, Mix, murine myelomas (e.g., SP2/0 and NSO) and C2C12 cells, as well as transformed primate cell lines, hybridomas, normal diploid cells, and cell strains derived from in vitro culture of primary tissue and primary explants. Any eukaryotic cell that is capable of expressing the polypeptide of interest may be used in the disclosed methods. Numerous cell lines are available from commercial sources, such as the American Type Culture Collection (ATCC). In one embodiment of the invention, the cell culture, e.g., the large-scale cell culture, employs CHO cells.
[0066] Although in certain embodiments the cell culture comprises mammalian cells, one skilled in the art will understand that it is possible to recombinantly produce polypeptides of interest in lower eukaryotes such as yeast, or in prokaryotes such as bacteria. One skilled in the art would know that the culture conditions for yeast and bacterial cell cultures will differ from the culture conditions of animals cells, and will understand how these conditions will need to be adjusted in order to optimize cell growth and/or protein production. [0067] Suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium, or any bacterial strain capable of expressing the polypeptide of interest. Expression in bacteria may result in formation of inclusion bodies incorporating the recombinant protein. Thus, refolding of the recombinant protein may be required in order to produce active, or more active, material. Several methods for obtaining correctly folded heterologous proteins from bacterial inclusion bodies are known in the art. These methods generally involve solubilizing the protein from the inclusion bodies, then denaturing the protein completely using a chaotropic agent. When cysteine residues are present in the primary amino acid sequence of the protein, it is often necessary to accomplish the refolding in an environment that allows correct formation of disulfide bonds (a redox system). General methods of refolding are known in the art and disclosed in, e.g., Kohno (1990) Meth. Enzymol. 185:187-95, EP 0433225, and U.S. Patent No. 5,399,677.
[0068] Suitable yeast strains for polypeptide production include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Pichia pastoris, Kluyveromyces strains, Candida, or any yeast strain capable of expressing polypeptide of interest. [0069] The term "bioreactor" as used herein refers to any vessel used for the growth of a eukaryotic cell culture, e.g., an animal cell culture (e.g., a mammalian cell culture). The bioreactor can be of any size as long as it is useful for the culturing of cells, e.g., mammalian cells. Typically, the bioreactor will be at least 30 ml and may be at least 1, 10, 100, 250, 500, 1000, 2500, 5000, 8000, 10,000, 12,000 liters or more, or any intermediate volume. The internal conditions of the bioreactor, including but not limited to pH and temperature, are typically controlled during the culturing period. The term "production bioreactor" as used herein refers to the final bioreactor used in the production of the polypeptide or protein of interest. The volume of a large-scale cell culture production bioreactor is generally greater than about 100 ml, typically at least about 10 liters, and may be 500, 1000, 2500, 5000, 8000, 10,000, 12,000 liters or more, or any intermediate volume. A suitable bioreactor or production bioreactor may be composed of (i.e., constructed of) any material that is suitable for holding cell cultures suspended in media under the culture conditions of the present invention and is conducive to cell growth and viability, including glass, plastic or metal; the material(s) should not interfere with expression or stability of the produced product, e.g., a therapeutic protein product. One of ordinary skill in the art will be aware of, and will be able to choose, suitable bioreactors for use in practicing the present invention.
[0070] The terms "medium," "cell culture medium," and "culture medium" as used herein refer to a solution containing nutrients that nourish growing animal cells, e.g., mammalian cells, and can also refer to medium in combination with cells. The term "inoculation medium" refers to the medium that is used to form a cell culture. Inoculation medium may or may not differ in composition from the medium used during the rest of the cell growth phase. Typically, medium solutions provide, without limitation, essential and nonessential amino acids, vitamins, energy sources, lipids, and trace elements required by the cell for at least minimal growth and/or survival. The solution may also contain components that enhance growth and/or survival above the minimal rate, including hormones and growth factors. The solution is preferably formulated to a pH and salt concentration optimal for cell survival and proliferation. In at least one embodiment, the medium is a defined medium. Defined media are media in which all components have a known chemical structure. In other embodiments of the invention, the medium may contain an amino acid(s) derived from any source or method known in the art, including, but not limited to, an amino acid(s) derived either from single amino acid addition(s) or from a peptone or protein hydrolysate addition(s) (including animal or plant source(s)). In yet other embodiments of the invention, the medium used during the cell growth phase may contain concentrated medium, i.e., medium that contains higher concentration of nutrients than is normally necessary and normally provided to a growing culture. One skilled in the art will recognize which cell media, inoculation media, etc. is appropriate to culture a particular cell, e.g., animal cell (e.g., CHO cells), and the amount of glucose and other nutrients (e.g., glutamine, iron, trace D elements) or agents designed to control other culture variables (e.g., the amount of foaming, osmolality) that the medium should contain (see, e.g., Mather, J.P., et al. (1999) "Culture media, animal cells, large scale production," Encyclopedia of Bioprocess Technology: Fermentation, Biocatalysis, and Bioseparation, Vol. 2:777-85; U.S. Published Patent Application No. 2006/0121568; both of which are hereby incorporated by reference herein in their entireties). The present invention also contemplates variants of such known media, including, e.g., nutrient-enriched variants of such media. The term "cell density" as used herein refers to the number of cells present in a given volume of medium. The term "viable cell density" as used herein refers to the number of live cells present in a given volume of medium under a given set of experimental conditions.
[0071] The term "cell viability" as used herein refers to the ability of cells in culture to survive under a given set of culture conditions or experimental variations. The term as used herein also refers to that portion of cells that are alive at a particular time in relation to the total number of cells, living and dead, in the culture at that time.
[0072] The terms "integrated viable cell density," "integrated viable cell number" or "IVC" as used herein refer to the average density of viable cells over the course of the culture multiplied by the amount of time the culture has run. When the amount of protein produced is proportional to the number of viable cells present over the course of the culture, integrated viable cell density is a useful tool for estimating the amount of protein produced over the course of the culture. [0073] The methods of the present invention are applicable to cells grown in a batch culture, fed batch culture, perfusion culture, modified fed-batch culture (see U.S. Provisional Application Number 60/954,922), batch refeed culture, or any combination thereof.
[0074] The term "batch culture" as used herein refers to a method of culturing cells in which all the components that will ultimately be used in culturing the cells, including the medium as well as the cells themselves, are provided at the beginning of the culturing process. A batch culture is typically stopped at some point and the cells and/or components in the medium are harvested and optionally purified.
[0075] The term "fed-batch culture" as used herein refers to a method of culturing cells in which additional components are provided to the culture at some time subsequent to the beginning of the culture process. The provided components typically comprise nutritional supplements for the cells that have been depleted during the culturing process. A fed-batch culture is typically stopped at some point and the cells and/or components in the medium are harvested and optionally purified.
[0076] The term "perfusion culture" as used herein refers to a method of culturing cells in which additional fresh medium is provided, either continuously over some period of time or intermittently over some period of time, to the culture (subsequent to the beginning of the culture process), and simultaneously spent medium is removed. The fresh medium typically provides nutritional supplements for the cells that have been depleted during the culturing process. Polypeptide product, which may be present in the spent medium, is optionally purified. Perfusion also allows for removal of cellular waste products (flushing) from the cell culture growing in the bioreactor.
[0077] The term "modified fed-batch culture" as used herein refers to a method of culturing cells which combines both the fed-batch culture method and the perfusion culture method. The modified fed-batch culture method is described in U.S. Provisional Application Number 60/954,922, incorporated herein in its entirety by reference. [0078] The invention can also be practiced with batch re-feed processes. It is believed that reduced pH will provide a better handle for reducing protein misfolding and/or aggregation and for modifying glycosylation in this type of cell culture.
[0079] In one embodiment of the present invention, the method comprises first inoculating the cell culture with the inoculation medium during the growth phase of the cell culture, and subsequently switching cells into the production phase, wherein the temperature and/or pH of the cell culture is adjusted to a reduced temperature and/or a reduced pH. Growth phase, as used herein, refers to a stage in the cell culture wherein the cells are grown to achieve the cell density optimal for protein production.
[0080] In another embodiment, following growth phase, the cells may be switched into the production phase, which may occur at different temperature and/or different pH than the growth phase, for example, reduced temperature and/or reduced pH. In embodiments, the cell culture may be switched from growth phase to production phase one day after the inoculation. In other embodiments, the cell culture may be switched from growth phase to production phase five days after the inoculation. When shifting the cell culture from the growth to the production phase, the shift may be relatively gradual. Alternatively, the shift may be abrupt. With gradual change, the temperature and/or pH may be steadily adjusted, e.g., decreased. Alternatively, the temperature and/or pH may be adjusted by discrete intervals. [0081] Production phase of the cell culture is a stage in the cell culture wherein the cells are grown under conditions optimal for producing the polypeptide of interest, e.g., a therapeutic protein. During the production phase, the reduced temperature and/or pH of the cell culture may be selected based on the temperature and/or pH at which the cell culture remains viable, at which high level of protein is produced, at which production and accumulation of metabolic waste products, e.g., lactic acid and ammonia, is minimized, at which the product quality of the protein has been appropriately controlled, and/or any combination of these or other factors deemed important by the practitioner. [0082] In an alternative embodiment of the invention, the method of the invention comprises inoculating the cell culture at a reduced temperature and/or reduced pH, so no temperature or pH shift is necessary at the initiation of the production phase. A skilled artisan would know to monitor the temperature and the pH of the cell culture such that the temperature and the pH do not deviate from the established temperature and pH set points. For instance, a skilled artisan will know to use a base, e.g., sodium carbonate base, to prevent cultures from deviating below the set pH.
[0083] In the examples hereinbelow, the cell culture medium started at a pH above the target pH set point and no deliberate adjustment to pH set point occurred. However, the use of a pH adjustment during the course of the cell culture could also be included as would be understood by those of skill in the art. Similarly, initiating the culture at low temperature without an adjustment is also possible. For example, the process may be comprised of a production phase only. [0084] "Reduced temperature," as used herein, refers to a temperature below the conventional temperature for cell growth (the temperature at which the cell are typically grown) for that type of cell. For example, in embodiments of the present invention, where the cells are mammalian cells, the cell culture in the production phase is preferably in a range of 24.O0C to less than 30.00C, and more preferably in a range of 27.O0C to less than 30.00C. For example, reduced temperature of the cell culture is 24.O0C, 24.50C, 25.O0C, 25.50C, 26.O0C, 26.50C. 27.O0C, 27.50C, 28.O0C, 28.50C, 29.O0C, 29.50C, 29.60C, 29.70C, 29.80C, and 29.90C. In the most preferred embodiment of the invention described herein, reduced temperature of the cell culture is a temperature of about 29.50C. Reduced temperature for cells other than mammalian may be determined on a case by case basis by a person of ordinary skill in the art. [0085] "Reduced pH," as used herein, refers to a pH set point below the conventional pH for cell growth (the pH at which the cells are typically grown) of that particular cell type. In embodiments of the present invention, where the cells are mammalian cells, reduced pH of the cell culture in the production phase is below 7.00. In embodiments of the invention, reduced pH of the cell culture is in a range of 6.50 to less than 7.00, preferably in a range of 6.80 to less than 7.00. For example, reduced pH of the cell culture is 6.80, 6.85, 6.90, 6.95, 6.96, 6.97, 6.98, and 6.99. In the most preferred embodiment of the invention, reduced pH of the cell culture is about 6.95. Reduced pH for cells other than mammalian may be determined on a case by case basis by a person of ordinary skill in the art. [0086] A skilled artisan will understand that depending on the cell type of the cell culture, conventional temperature and pH (as distinguished from the reduced temperature and pH) will differ. For example, conventional temperature and pH for most mammalian cells, e.g., CHO cells, is above 30.0° C (such as, for example, 37.O0C) and above 7.00 (such as, for example, 7.20), respectively. A skilled artisan will also understand that because conventional temperature and pH for other cell types, e.g., insect cells, will differ from the conventional temperature for mammalian cells, the methods of the invention will utilize a different reduced temperature and a different reduced pH for such cells. [0087] In certain embodiments of the present invention, the practitioner may find it beneficial or necessary to periodically monitor particular conditions of the growing cell culture. As nonlimiting examples, it may be beneficial or necessary to monitor, e.g., temperature, pH, dissolved oxygen, cell density, cell viability, integrated viable cell density, lactate levels, ammonium levels, glucose levels, glutamine levels, osmolality, titer of the expressed polypeptide, etc. Numerous techniques are well known to those of skill in the art for measuring such conditions/criteria. For example, cell density may be measured using a hemocytometer, an automated cell-counting device (e.g., a Coulter counter, Beckman Coulter Inc., Fullerton, CA), or cell-density examination (e.g., CEDEX®, Innovatis, Malvern, PA). Viable cell density may be determined by staining a culture sample with Trypan blue. Lactate, ammonium, glucose and glutamine levels, as well as dissolved oxygen and pH may be measured, e.g., with the BioProfile 400 Chemistry Analyzer (Nova Biomedical, Waltham, MA), which takes measurements of key nutrients, metabolites, and gases in cell culture media. Dissolved oxygen and pH may also be measured using, e.g., a blood gas analyzer (e.g., a Bayer Rapidlab 248 pH/blood gas analyzer (Bayer HealthCare LLC, East Walpole, MA)). Temperature, pH, and dissolved oxygen may also be measured by, e.g., various types of in-situ probes. Osmolality of the cell culture may be measured by, e.g., a freezing point osmometer. HPLC can be used to determine, e.g., the levels of lactate, ammonium, or the expressed polypeptide or protein. In one embodiment of the invention, the levels of expressed polypeptide can be determined by using, e.g., protein A HPLC. Alternatively, the level of the expressed polypeptide or protein can be determined by standard techniques such as Coomassie staining of SDS-PAGE gels, Western blotting, Bradford assays, Lowry assays, biuret assays, and UV absorbance. It may be necessary to monitor the posttranslational modifications of the expressed polypeptide or protein, e.g., glycosylation. It also may be beneficial to monitor other posttranslational modifications of the protein, e.g., phosphorylation, etc. In order to monitor certain cell culture conditions, it may be necessary to remove small aliquots of the culture for analysis. One of ordinary skill in the art will understand that such removal may potentially introduce contamination into the cell culture, and will take appropriate care to minimize the risk of such contamination. [0088] In one embodiment of the invention, a practitioner will monitor integrated viable cell number at the reduced cell culture temperature and/or reduced pH. Preferably, growing cells at the reduced temperature and/or reduced pH reduces integrated viable cell density by 20%, more preferably less than 20% (e.g., 15%). In another embodiment of the invention, a practitioner will monitor cell viability at the reduced cell culture temperature and/or reduced pH. Preferably, growing cells at the reduced temperature and/or reduced pH reduces cell viabilities by less than 15%, more preferably by less than 5%.
[0089] Glucose is a primary source of energy for the cell culture. Significant deviations in glucose consumption in the cell culture could indicate a negative effect of cell culture conditions on the health of the cell culture. Thus, in a preferred embodiment of the invention, growing cells at the reduced temperature and/or reduced pH results in minimal change, e.g., reduction, in glucose consumption of the cell culture.
[0090] Glutamine is an alternative source of energy for the cell culture, and is an important source of nitrogen for various molecules, e.g., amino acids, in the cell culture. Thus, in a preferred embodiment of the invention, growing cells at the reduced temperature and/or reduced pH results in a minimal change, e.g., reduction, in the glutamine consumption of the cell culture. [0091] In another embodiment of the invention, a practitioner will monitor waste product production, e.g., lactate and ammonia production, in the cell culture. Thus, in a preferred embodiment of the invention, growing cells at the reduced temperature and/or pH results in a minimal change, e.g., increase, in production of lactate and ammonia. In some embodiments of the invention, growing cells at a reduced temperature an/or pH can minimize lactate and ammonia production. [0092] Additionally, in a preferred embodiment of the present invention, growing cells at a reduced temperature and/or pH results in minimal reduction in cumulative average productivity and product titer. The term "titer" as used herein refers to the total amount of polypeptide of interest, e.g., a glycoprotein of interest, produced by a cell culture (e.g., an animal cell culture), divided by a given amount of medium volume; thus "titer" refers to a concentration. Titer is typically expressed in units of milligrams of polypeptide per liter of medium. Preferably, any reduction in cell productivity and product titer experienced in the cell culture is offset by decreased production of aggregated or misfolded protein product, e.g., decreased production of HMWA.
[0093] As used herein, the term "aggregated protein" refers to the protein groupings, i.e., high molecular weight aggregates, that produce nonfunctional, sub-optimal or undesired protein product. The term "misfolded protein" refers to an improperly folded protein, often a protein that can no longer display normal biological activity, e.g., normal enzymatic activity. One skilled in the art will know that protein aggregates may comprise either or both correctly folded and/or misfolded proteins. Aggregated or misfolded proteins are commonly formed in overexpressing cell culture, e.g., a cell culture overexpressing a protein of interest, e.g., a glycoprotein of interest. For instance, aggregation can be caused either by nonspecific hydrophobic interactions of unfolded polypeptide chains or by interaction of folding intermediates. In one embodiment of the invention, growing cells at the reduced temperature and/or reduced pH results in at least 50% reduction in protein misfolding/aggregation, preferably about 60% reduction in protein misfolding/aggregation. One skilled in the art will know the techniques required to monitor protein aggregation/misfolding, e.g., hydrophobic interaction HPLC (HIC-HPLC).
[0094] The term "high molecular weight aggregate" (HMWA) refers to undesirable byproduct of protein production which results from association between at least two proteins. "High molecular weight aggregate" may be an association between at least two of the same proteins and/or association between the protein of interest and other proteins found in the cell culture, e.g., host cell proteins. The association may arise by any method including, but not limited to, covalent, noncovalent, disulfide, and/or nonreducible crosslinking. One skilled in the art will understand that when a protein is active in a multimer form (e.g., a dimer form), i.e., when more than one polypeptide chain is required for protein activity, the term "high molecular weight aggregates" will refer to an association between two or more of such multimer forms. In one embodiment of the invention, a protein that is active in a multimer form is a receptor, e.g., a cytokine receptor (e.g., sIL-13R). In one embodiment, growing cells at a reduced temperature and/or pH results in at least about a 10% reduction, preferably about a 40% reduction, more preferably about a 50% reduction, and even more preferably about an 80% or more reduction in high molecular weight aggregates, or any intermediate values. One skilled in the art will know the techniques required to monitor production of high molecular weight aggregates, e.g., size- exclusion high performance liquid chromatography (SEC-HPLC). [0095] In another aspect of the invention, temperature and/or pH in a cell culture are used to change protein glycosylation, e.g., protein sialylation, to a predetermined level. For example, decreasing pH and/or temperature of the cell culture may lead to the decrease in both N- and 0-linked glycosylation, e.g., decrease in iV-glycan sialylation). Change in protein glycosylation (e.g., sialylation) can affect stability, enzymatic activity, circulation lifetime, and immunogenicity of the therapeutic protein. One skilled in the art may monitor the degree of glycosylation (e.g., sialylation) to ensure that the changing temperature and/or pH achieves a desired type and level of protein glycosylation. Chromatographic techniques, e.g., Normal Phase Chromatography (NPC), may be used to monitor protein glycosylation of the protein product. [0096] At the end of the production phase, the cells are harvested and the polypeptide of interest is collected and purified. Preferably, at the end of the production phase, the polypeptide of interest displays reduced misfolding and/or aggregation, while retaining an acceptable glycosylation pattern. In one embodiment of the invention, the polypeptide of interest at the end of the production process is in a soluble form (e.g., the polypeptide of interest is a soluble receptor, e.g., a soluble cytokine receptor). Such soluble forms of the polypeptide can be purified from conditioned media.
[0097] Membrane-bound forms of the polypeptide can be purified by preparing a total membrane fraction from the expressing cells and extracting the membranes with a nonionic detergent such as TRITON® X-IOO (EMD Biosciences, San Diego, CA). Cytosolic or nuclear proteins may be prepared by lysing the host cells (via mechanical force, Parr-bomb, sonication, detergent, etc.), removing the cell membrane fraction by centrifugation, and retaining the supernatant. [0098] The polypeptide can be purified using other methods known to those skilled in the art. For example, a polypeptide produced by the disclosed methods can be concentrated using a commercially available protein concentration filter, for example, an AMICON® or PELLICON® ultrafiltration unit (Millipore, Billerica, MA). Following the concentration step, the concentrate can be applied to a purification matrix such as a gel filtration medium. Alternatively, an anion exchange resin (e.g., a MonoQ column, Amersham Biosciences, Piscataway, NJ) may be employed; such resin contains a matrix or substrate having pendant diethylaminoethyl (DEAE) or polyethylenimine (PEI) groups. The matrices used for purification can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step may be used for purification of proteins. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups (e.g., S-SEPHAROSE® columns, Sigma-Aldrich, St. Louis, MO). [0099] The purification of the polypeptide from the culture supernatant may also include one or more column steps over affinity resins, such as concanavalin A- agarose, AF-HEPARIN650, heparin-TOYOPEARL® or Cibacron blue 3GA SEPHAROSE® (Tosoh Biosciences, San Francisco, CA); hydrophobic interaction chromatography columns using such resins as phenyl ether, butyl ether, or propyl ether; or immunoaffinity columns using antibodies to the labeled protein. Finally, one or more HPLC steps employing hydrophobic HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups (e.g., Ni-NTA columns), can be employed to further purify the protein. Alternatively, the polypeptides may be recombinantly expressed in a form that facilitates purification. For example, the polypeptides may be expressed as a fusion with proteins such as maltose-binding protein (MBP), glutathione-5-transferase (GST), or thioredoxin (TRX); kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ), and Invitrogen (Carlsbad, CA), respectively. The proteins can also be tagged with a small epitope (e.g., His, myc or Flag tags) and subsequently identified or purified using a specific antibody to the chosen epitope. Antibodies to common epitopes are available from numerous commercial sources. Some or all of the foregoing purification steps in various combinations or with other known methods, can be employed to purify a polypeptide of interest, e.g., a therapeutic protein, produced by the methods of the present invention.
Pharmaceutical Compositions
[0100] In certain embodiments of the invention, proteins produced according to one or more methods of the present invention can be useful in the preparation of pharmaceuticals. Proteins produced according to one or more methods of the present invention may be administered to a subject or may first be formulated for delivery by any available route including, but not limited to, e.g., parenteral (e.g., intravenous), intradermal, subcutaneous, oral, nasal, bronchial, ophthalmic, transdermal (topical), transmucosal, rectal, and vaginal routes. Inventive pharmaceutical compositions typically include a purified protein expressed from a mammalian cell line, a delivery agent (e.g., a cationic polymer, peptide molecular transporter, surfactant, etc., as described above) in combination with a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" includes nontoxic materials that do not interfere with the effectiveness of the biological activity of the active ingredient(s), e.g., solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The characteristics of the carrier will depend on the route of administration. Supplementary active compounds can also be incorporated into the compositions.
[0101] A pharmaceutical composition is formulated to be compatible with its intended route of administration. When the therapeutic protein produced according to one or more methods of the present invention is administered in an oral form, the pharmaceutical composition will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder will contain from about 5 to 95% binding agent, and preferably from about 25 to 90% binding agent. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin, such as sesame oil, peanut oil (taking into consideration the occurrence of allergic reactions in the population), mineral oil, or soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of the binding agent, and preferably from about 1 to 50% by weight of the binding agent.
[0102] When the therapeutic protein produced according to one or more methods of the present invention is administered by intravenous, cutaneous or subcutaneous injection, the therapeutic protein will be in the form of a pyrogen- free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable protein solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill of those in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to the therapeutic protein, an isotonic vehicle such as sodium chloride injection, Ringer's injection, dextrose injection, dextrose and sodium chloride injection, lactated Ringer's injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additive known to those of skill in the art.
[0103] Additional formulation of the pharmaceutical compositions comprising the therapeutic proteins produced by one or more methods of the present invention will be known to those skilled in the art. One of ordinary skill in the art will also be aware of unit dosage formulations appropriate for proteins produced according to the present invention.
[0104] The entire contents of all references, patents, and patent applications cited throughout this application are hereby incorporated by reference herein.
EXAMPLES
[0105] The Examples which follow are set forth to aid in the understanding of the invention but are not intended to, and should not be construed to, limit the scope of the invention in any way. The Examples do not include detailed descriptions of conventional methods, e.g., cloning, transfection, and basic aspects of methods for overexpressing proteins in cell lines. Such methods are well known to those of ordinary skill in the art.
Example 1
TNFR-Fc Protein Production
Example 1.1: Effects of Reduced Temperature on Cell Culture Performance of Recombinant CHO Cells and on Product Quality of TNFR-Fc Fusion Protein
Example 1.1.1: Materials and Methods
[0106] Chinese Hamster Ovary (CHO) cells overexpressing recombinant glycoprotein TNFR-Fc were inoculated at identical concentrations and conditions in four separate bench-scale bioreactors at 37.O0C. Cells were grown in a fed- batch culture for one day, followed by a temperature- shift to either 30.00C, 29.O0C, 28.O0C, or 27.O0C. The initial pH set point lower limit of the culture was 7.00. A cell sample was removed from the culture each day to measure various conditions of the cells culture, e.g., integrated viable cell numbers, cell viabilities, residual glucose profile, residual glutamine profile, lactate and ammonia concentrations, cumulative average cell productivity (Qp), product titer, levels of misfolded / aggregated product, levels of high molecular weight aggregates, levels of sialylation, and pH.
[0107] Integrated viable cell numbers (IVC) were calculated by measuring cell density using cell-density examination (e.g., CEDEX®, Innovatis, Malvern, PA) and were normalized to the average harvest day IVC. The average harvest day IVC was determined by calculating the arithmetic mean of the harvest day (e.g., day 10) integrated viable cell density for the all experimental conditions tested. All individual IVC values were then scaled to the average harvest IVC to generate normalized values.
[0108] Cell viabilities were measured by staining with Trypan blue using cell- density examination (e.g., CEDEX®, Innovatis, Malvern, PA). Additionally, the residual glucose profile and the residual glutamine profile in the cell culture were measured using the BioProfile Chemistry Analyzer (Nova Biomedical, Waltham, MA). Concentrations of the waste products, lactate and ammonia, were measured using the BioProfile Chemistry Analyzer (Nova Biomedical, Waltham, MA).
[0109] Cumulative average cell productivity (Qp) and product titer were measured using protein A HPLC and were normalized to the average harvest day cumulative average Qp and titer. The average harvest day titer was determined by calculating the arithmetic mean of the harvest day (e.g., day 10) titer for all experimental conditions tested. All individual titer values were then scaled to the average harvest day titer to generate normalized values. The average harvest day cumulative average Qp was determined by calculating the arithmetic mean of the harvest day cumulative average Qp for all experimental conditions tested. All individual values were then scaled to the average harvest day titer to generate normalized values.
[0110] Levels of misfolded / aggregated product were measured using HIC- HPLC, and levels of high molecular weight aggregates were measured using SEC-HPLC. Levels of total and iV-linked glycan sialylation were measured by subjecting 2-aminobenzamide- (2-AB)-labeled protein glycoforms to Normal Phase Chromatography (NPC). The amount of total sialylation (N- and O- linked) was defined as the percentage relative to the amount of total sialylation of reference material, i.e., TNFR-Fc aliquot with known and preferable sialylation pattern. pH of the cell culture was determined by an off-line measurement via blood gas analyzer (Bayer Rapidlab 248 pH/blood gas analyzer (Bayer Healthcare LLC, East Walpole, MA)).
Example 1.1.2: Results
[0111] Growing cells at a reduced temperature had minimal effects on IVC number. Specifically, lower operating temperatures resulted in a lower final IVC number; however, the effect of reducing cell culture temperature to 270C was minor - only about a 20% decrease in the IVC number was observed (FIG. IA) Cell viabilities were not greatly affected by the decrease in temperature; the lowest temperature condition (i.e., 270C) had 5% lower harvest day viability (FIG. IB).
[0112] Additionally, reduced operating temperatures resulted in a higher residual glucose profile, which indicated lower glucose consumption of the cell culture (FIG. 2A); however glutamine profiles were not significantly altered by reduced temperatures (FIG. 2B).
[0113] In some cases, late in the cell culture, lactic acid and ammonia may also be consumed by the cell culture. The cessation of the production of lactic acid and ammonia or the consumption of lactic acid and ammonia promote cell viability, cell productivity, and can have an effect of increasing polypeptide product titer. Growing cells at the reduced temperature had an effect of altering the net consumption of lactate in the later half of the fed-batch culture (FIG. 3A). Reduced temperatures also resulted in higher ammonia production (FIG. 3B). Reduced temperatures resulted in lower cell-specific productivities (lower Qp) (FIG. 4A) and lower product titer (FIG. 4B). Lower product titer is the result of lower cell specific productivity and lower integrated viable cell number. However, lower product titer and cell productivity was offset by a significantly decreased proportion of misfolded and aggregated product (FIG. 5A). Moreover, reducing temperature of the cell culture significantly decreased proportion of the high molecular weight aggregates in the cell culture (FIG. 5B). Therefore, reducing temperature resulted in improved TNFR-Fc protein product. [0114] This decrease in temperature correlated with the decrease in total product sialylation (both N- and 0-linked sialylation) (FIG. 6A). In fact, there was a direct relationship between reducing cell culture temperature and reducing the proportion of sialylated iV-linked glycans (FIG. 6B), indicating that the production temperature must be chosen to balance the beneficial effect of reducing HMWA and protein misfolding and the unfavorable effect of reducing glycosylation. For instance, growing cells at a reduced temperature of 29.50C significantly reduced concentration of both misfolded and aggregated TNFR-Fc, while having minimal effects on TNFR-Fc glycosylation. [0115] Differences in the net consumption of lactate between the cell cultures grown at different temperatures led to a difference in pH of the cell culture at the time of harvest (FIG. 7). This is due to the fact that larger net consumption of lactate (lactic acid) led to a larger rise in culture pH since acid was being removed from the environment.
Example 1.2: Effects of Reduced pH on Cell Culture Performance of Recombinant CHO Cells and on Product Quality of TNFR-Fc Fusion Protein
Example 1.2.1: Materials and Methods.
[0116] CHO cells overexpressing recombinant glycoprotein TNFR-Fc were inoculated at identical concentrations at 37.O0C and at pH set point of either 7.20, 7.10, 7.00, 6.90 or 6.80. Addition of sodium carbonate base was utilized to prevent cultures from deviating below the set point pH. No pH control was utilized above the set point pH. Bioreactors were temperature- shifted to 30.00C on day 1 (one day post inoculation, i.e., one day after the start of the experiment). Various conditions of the cell culture were measured as described in Example 1.1.1.
Example 1.2.2: Results
[0117] Growing cells at pH set points of 0.2 pH units away from pH 7.00 led to slightly decreased IVC; specifically, growing cells at pH of 6.80 led to about a 15% decrease in IVC compared to cells grown at pH of 7.00 (FIG. 8A). Additionally, growing cells at pH of 7.20 led to lower cell viability (FIG. 8B). [0118] Additionally, lower operating pH set points resulted in lower glucose consumption (FIG. 9A). Glutamine profiles were not significantly altered by growing cells at pH set points of 6.80 to 7.10 (FIG. 9B).
[0119] Not surprisingly, higher operating pH set points led to higher production of lactate (FIG. 10A). At pH set point of 6.80, net consumption of lactate in the later half of the fed-batch did not occur. Lower operating pH set points resulted in higher ammonia production (FIG. 10B).
[0120] Differences in the net consumption of lactate in the later half of the experiment led to deviation of the pH of cultures from the operating pH set point, as only base, and not acid, addition was utilized to maintain pH (FIG. HA). As a result of the differences in the pH set points, lactate profiles, and titrant additions, osmolality also differed between the pH conditions (FIG. HB). [0121] Operating cultures at pH set points of 0.2 pH units away from pH 7.00 led to slightly lower cell-specific productivities (i.e., Qp) (FIG. 12A). Utilizing a pH set point of 6.80 or 7.20 resulted in lower product titers due to both lower cell-specific productivity and lower integrated viable cell numbers (FIG. 12B). [0122] However, lower operating pH set points resulted in decreased product misfolding and aggregation (FIG. 13A). Additionally, significant reduction in the proportion of high molecular weight aggregates was observed at lower pH set points (FIG. 13B).
[0123] Lower operating pH set points resulted in lower total sialylation (both N- and 0-linked glycans) (FIG. 14A), as well as a lower proportion of iV-linked glycan sialylation (FIG. 14B). Because the amount of total sialylation (N- and 0-linked) was defined as the percentage relative to the amount of total sialylation of reference material, any values below 100% represented reduction in total sialylation and values above 100% represented increase in total sialylation. The pattern of glycosylation can be depicted as the profile of glycans of the glycoprotein (FIG. 15). Growing cells at the reduced pH resulted in a significant alteration of the overall glycosylation profile (FIG.16). While no new glycoforms were detected at the reduced pH, ratios of glycoforms present were significantly altered; growing cells at the reduced pH resulted in reduction of complex di- and mono-sialylated glycoforms. Because of the potentially unfavorable effect of the reduced pH on protein glycosylation, pH of the cell culture must be chosen in such as way that the detrimental effects on protein glycosylation are balanced by the beneficial effects on reduction of misfolded and/or aggregated proteins. For instance, growing cells at a reduced pH of 6.95 has significantly reduced the proportion of misfolded and aggregated TNFR-Fc protein, while having minimal effects on glycosylation of TNFR-Fc.
Example 1.3: Discussion
[0124] These studies demonstrate that within a temperature range of 27.O0C to 30.00C or within a pH set point range of 6.80 to 7.20, cell growth and specific productivity of the cell culture can be significantly affected. However, while altering the temperature by as little as 1-20C and the pH set points by as little as 0.1-0.2, does not significantly affect cell growth and specific productivity, a significant (favorable) difference can be observed in protein folding and protein aggregation. Thus, small differences in cell culture conditions that do not significantly affect cell growth, viability or productivity, may significantly alter product quality, e.g., reduce protein folding and protein aggregation or affect glycosylation.
Example 2
Effects of Temperature on sIL-13R Production and Evaluation of a New sIL-13R Producing Cell Line
Example 2.1 : Materials and Methods
[0125] Stably transfected Chinese Hamster Ovary (CHO) cells overexpressing recombinant glycoprotein sIL-13Rwere seeded in Applikon bioreactors at 3x105 cells/mL in an inoculation medium with Antifoam (Dow Corning Corporation, Midland, MI). Additional Antifoam was added as required. A concentrated nutrient medium was used as a feed medium. The pH set point lower limit was 6.80. The dθ2 set point was 23% utilizing 7% CO2/93% air sparge gas, while agitation was 200 rpm. Bioreactors were temperature- shifted from 37.O0C on day 5. The production phase temperature was either 37.O0C, 33.O0C, 32.O0C, 28.O0C, or room temperature (RT; about 24.O0C). Feeding schedules for experimental conditions are summarized in Table 1. Feed volumes are listed as percentage of culture volume in bioreactor. The temperature shift for cell cultures occurred at about 6x106 cells/mL.
Table 1. Bioreactor Feed Schedules
Figure imgf000038_0001
Example 2.2. Results
[0126] The viable cell densities achieved after the temperature shift were slightly lower at RT (about 6xl06cells/mL) and 29.O0C (about 7xl06cells/mL) than at higher temperatures (about 8xl06cells/mL) (FIG. 17A). Similarly, the total cell densities achieved after the temperature shift were slightly lower at RT (about 6.5xl06cells/mL) and 29.O0C (about 7.5xlO6 cells/mL) than at higher temperatures (about 8 to 9xl06 cells/mL) (FIG. 17B). Viabilities were better maintained throughout the fed-batch at lower temperatures (FIG. 17C) The rate of decline in viability varied with the temperature; the viability profiles of the RT and 29.O0C cultures were significantly higher than the remaining cultures at 31.O0C, 32.O0C, 33.O0C and 37.O0C (FIG. 17C).
[0127] At the end of the 18-day fed-batch culture, the highest titer (combined sIL-13R dimer and HMWA titer) was achieved by the 31.O0C culture followed by the 32.O0C culture (188 mg/L), 29.O0C culture (178 mg/L), and 33.O0C culture (151 mg/L) (FIG. 18). The RT and 37.O0C cultures had significantly lower titers. The cell specific productivity, or daily specific sIL-13R production rate (Qp), of the sIL-13R-producing cell line was significantly diminished at RT and 37.O0C (FIG. 19A). Additionally, the cumulative average specific productivities, or cumulative average Qp, were also lower at 37.O0C and RT (FIG. 19B).
Interestingly, the large increases in specific productivities seen toward the end of the 37.O0C, 33.O0C, 32.O0C, and 31.O0C cultures corresponded with significant decreases in the culture viabilities (data not shown). [0128] Specific glucose consumption (QgIc) declined with decreasing temperature (FIG. 20). Specific glutamine consumption (QgIn) also appeared to decline with decreasing temperature (FIG. 21). However, it should be noted that the data on specific glutamine consumption did not account for glutamine degradation in the media, which would occur to a greater extent with increasing temperature. Therefore, the impact of temperature on specific glutamine consumption may be skewed. Lactate concentration profiles for the RT, 29.O0C, 31.O0C, and 32.O0C cultures were similar with lactate concentration peaking on or around the day of the temperature shift, then decreasing to between 0.7 and 2.0 g/L by day 18 (FIG. 22). For the 33.O0C and 37.O0C cultures, the lactate profiles were similar to the other cultures during the growth phase, but were significantly different during the production phase. For the 33.O0C culture, lactate concentration did not decrease after the temperature shift, and instead remained essentially constant. For the 37.O0C culture, lactate increased throughout the entire duration of the fed-batch.
[0129] Ammonia concentration varied during the production phase according to temperature (FIG. 23). For the 29.O0C culture, the ammonia concentration peaked on or around the day of the temperature shift, decreased during the middle stage of the fed-batch, and then increased during the late stage of the fed-batch. With increasing temperature above 29.O0C, the increase in ammonia concentration in the late stage of the fed-batch was greater and occurred sooner. For the 37.O0C culture, ammonia concentration increased throughout the entire course of the fed-batch. For the RT culture, the ammonia levels remained approximately constant after the temperature shift.
[0130] Samples of conditioned media from these bioreactor runs were analyzed for high molecular weight aggregates (HMWA) by size-exclusion chromatography (SEC) of batch-bound protein A eluates. A trend evident from the results of the cell line was the relative increase in the percentage of HMWA present in the conditioned medium with increasing temperature (FIG. 24 and FIG. 25). Thus, operating the cell culture at lower temperatures resulted in decreased sIL-13R product aggregation.
[0131] The decrease in percentage of sIL-13R dimer correlated with the increase in percentage of the HMWA. (FIGS. 26A and 26B). The RT culture had lower %HMWA than other cultures (FIG. 26B), as well as higher percentage of dimer formation. (FIG. 26A). The RT culture and the 31.O0C culture yielded similar dimer only titer, while the 29.O0C culture yielded the highest dimer only titer (FIG.27).
[0132] In order to corroborate the conclusions from the SEC analysis, conditioned media samples were analyzed using Western blots. The qualitative differences in the amount of sIL-13R present as HMWA species versus dimer were examined. The Western blots demonstrate the trend that the amount HMWA aggregates present in the conditioned medium relative to the amount of dimer present increased with increasing temperature (data not shown). [0133] These experiments demonstrate the importance of the interplay between the temperature-dependent variables of viability and viable cell density, specific productivity, and HMWA formation on volumetric productivity of sIL-13R dimer. Although the 31.O0C culture achieved the highest titer (measurements included HMWA and dimer), cultures at lower temperatures (e.g., 29.O0C ) may yield comparable or improved volumetric productivity when measured in terms of dimer only.

Claims

WHAT IS CLAIMED IS:
1. A method of producing a protein in a cell culture comprising at least one of:
(a) growing cells in the cell culture at a reduced temperature; and
(b) growing cells in the cell culture at a reduced pH; to reduce production of misfolded proteins and/or aggregated proteins.
2. The method of claim 1, comprising growing cells in the cell culture at a reduced temperature and at a reduced pH.
3. The method of claim 1, wherein the cell culture is a large-scale cell culture.
4. The method of claim 1, wherein the cell culture is a fed-batch cell culture.
5. The method of claim 1, wherein the cell culture is a mammalian cell culture.
6. The method of claim 5, wherein the cell culture is a CHO cell culture.
7. The method of claim 5, wherein the reduced temperature is in a range of 27.O0C to less than 30.00C.
8. The method of claim 5, wherein the reduced pH is in a range of 6.80 to less than 7.00.
9. The method of claim 5, wherein the protein produced is a soluble receptor.
10. The method of claim 9, wherein the soluble receptor is TNFR-Fc.
11. The method of claim 10 wherein the reduced temperature is 29.50C.
12. The method of claim 10, wherein the reduced pH is 6.95.
13. The method of claim 9, wherein the soluble receptor is sIL-13R.
14. A method of producing a protein in a cell culture comprising at least one of:
(a) modifying the level of protein glycosylation of the produced protein to a predetermined level by modifying the temperature of the cell culture; and
(b) modifying the level of protein glycosylation of the produced protein to a predetermined level by modifying the pH of the cell culture.
15. The method of claim 14, wherein the protein glycosylation is iV-glycan sialylation.
16. The method of claim 14, wherein the cell culture is a large-scale cell culture.
17. The method of claim 14, wherein the cell culture is a fed-batch cell culture.
18. The method of claim 14, wherein the cell culture is a mammalian cell culture.
19. The method of claim 18, wherein the cell culture is a CHO cell culture.
20. The method of claim 18, wherein the method includes growing the cell culture at a temperature in a range of 27.O0C to less than 30.00C.
21. The method of claim 18, wherein the method includes growing the cell culture at a pH in a range of 6.80 to less than 7.00.
22. The method of claim 18, wherein the protein produced is a soluble receptor.
23. The method of claim 22, wherein the soluble receptor is TNFR-Fc.
24. The method of claim 22, wherein the soluble receptor is sIL-13R.
25. The method of claim 1, wherein the produced protein is a therapeutic protein.
26. A pharmaceutical composition comprising the therapeutic protein made by the method of claim 25, and a pharmaceutically acceptable carrier.
27. The method of claim 14, wherein the produced protein is a therapeutic protein.
28. A pharmaceutical composition comprising the therapeutic protein made by the method of claim 27, and a pharmaceutically acceptable carrier.
29. The method as in any of claims 1-25 or 27, further comprising a step of isolating the protein from the cell culture.
30. The method according to claim 29, wherein the protein is further purified or processed for formulation.
31. The method according to claim 30, wherein the protein is formulated into a pharmaceutical composition.
PCT/US2008/061123 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture WO2008131374A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
RU2009139054/10A RU2478702C2 (en) 2007-04-23 2008-04-22 USE OF LOW TEMPERATURE AND/OR LOW pH IN CELL CULTURE
KR20147030274A KR20140132016A (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
BRPI0810511-1A2A BRPI0810511A2 (en) 2007-04-23 2008-04-22 LOW TEMPERATURE AND / OR LOW PH USE IN CELL CULTURE
AU2008242632A AU2008242632B2 (en) 2007-04-23 2008-04-22 Use of low temperature and/or low pH in cell culture
DK08746526.6T DK2139986T3 (en) 2007-04-23 2008-04-22 Use of low temperature and / or low pH in cell culture
CN200880013275A CN101679943A (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
EP08746526.6A EP2139986B1 (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
JP2010506424A JP5557736B2 (en) 2007-04-23 2008-04-22 Use of low temperature and low pH in cell culture
PL08746526T PL2139986T3 (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
CA2684727A CA2684727C (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
MX2009011362A MX2009011362A (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture.
SI200831866T SI2139986T1 (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
ES08746526.6T ES2646090T3 (en) 2007-04-23 2008-04-22 Use of low temperature and / or low pH in cell culture
IL201720A IL201720A (en) 2007-04-23 2009-10-22 Method of producing a protein at reduced temperature and reduced ph in a mammalian cell culture

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91338207P 2007-04-23 2007-04-23
US60/913,382 2007-04-23

Publications (1)

Publication Number Publication Date
WO2008131374A1 true WO2008131374A1 (en) 2008-10-30

Family

ID=39493848

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/061123 WO2008131374A1 (en) 2007-04-23 2008-04-22 Use of low temperature and/or low ph in cell culture
PCT/US2008/061125 WO2008131375A1 (en) 2007-04-23 2008-04-22 Methods of protein production using anti-senescence compounds

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2008/061125 WO2008131375A1 (en) 2007-04-23 2008-04-22 Methods of protein production using anti-senescence compounds

Country Status (26)

Country Link
US (2) US12054762B2 (en)
EP (2) EP2139986B1 (en)
JP (3) JP2010524504A (en)
KR (3) KR20140132016A (en)
CN (3) CN105039473A (en)
AR (2) AR066239A1 (en)
AU (2) AU2008242633B2 (en)
BR (2) BRPI0810511A2 (en)
CA (2) CA2685552C (en)
CL (2) CL2008001159A1 (en)
CO (2) CO6241165A2 (en)
DK (2) DK2139986T3 (en)
ES (2) ES2646090T3 (en)
HK (2) HK1142095A1 (en)
HU (1) HUE034776T2 (en)
IL (2) IL201720A (en)
MX (2) MX2009011363A (en)
PA (2) PA8777701A1 (en)
PE (2) PE20090731A1 (en)
PL (2) PL2139986T3 (en)
PT (2) PT2139987E (en)
RU (2) RU2009138226A (en)
SI (2) SI2139986T1 (en)
TW (2) TW200902708A (en)
WO (2) WO2008131374A1 (en)
ZA (1) ZA200907426B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012091124A1 (en) 2010-12-28 2012-07-05 中外製薬株式会社 Animal cell culturing method
EP2563906B1 (en) 2010-04-26 2017-11-08 Novartis AG Process for cultivation of cho cells
EP2188371B1 (en) 2007-08-09 2017-12-20 Wyeth LLC Use of perfusion to enhance production of fed-batch cell culture in bioreactors
WO2018073365A1 (en) 2016-10-19 2018-04-26 F. Hoffmann-La Roche Ag Method for producing an immunoconjugate
WO2018151819A1 (en) * 2017-02-17 2018-08-23 Lonza Ltd A method for producing biologic product variants
WO2020180967A1 (en) * 2019-03-04 2020-09-10 Amgen Inc. In vivo reversibility of high molecular weight species
US11046772B2 (en) 2015-03-26 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Process of production with controlled copper ions
EP3363811B1 (en) 2015-10-15 2023-04-19 Alteogen, Inc. Method for producing fusion protein having igg fc domain
US12018070B2 (en) 2014-10-15 2024-06-25 Alexion Pharmaceuticals, Inc. Methods of shifting an isoelectric profile of a protein product and uses thereof

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
TWI456062B (en) 2006-09-13 2014-10-11 Abbvie Inc Cell culture improvements
MX2009009240A (en) 2007-03-02 2009-09-08 Wyeth Corp Use of copper and glutamate in cell culture for production of polypeptides.
WO2010034015A2 (en) 2008-09-22 2010-03-25 The Regents Of The University Of Colorado, A Body Corporate Modulating the alternative complement pathway
CN102257006A (en) 2008-10-20 2011-11-23 雅培制药有限公司 Isolation and purification of antibodies using protein a affinity chromatography
MX2011004201A (en) 2008-10-20 2011-05-24 Abbott Lab Viral inactivation during purification of antibodies.
BR112012013330A2 (en) * 2009-12-02 2017-03-28 Acceleron Pharma Inc compositions and methods for increasing fc fusion protein serum half life
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2012170938A1 (en) 2011-06-08 2012-12-13 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
SG11201504249XA (en) 2012-09-02 2015-07-30 Abbvie Inc Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
WO2014143205A1 (en) 2013-03-12 2014-09-18 Abbvie Inc. Human antibodies that bind human tnf-alpha and methods of preparing the same
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
WO2014145098A1 (en) * 2013-03-15 2014-09-18 Genentech, Inc. Cell culture compositions with antioxidants and methods for polypeptide production
KR20150133833A (en) 2013-03-26 2015-11-30 코히러스 바이오사이언시즈, 인코포레이티드 Protein Production Method
US9416181B2 (en) 2013-05-06 2016-08-16 Abbvie Inc. Compositions for cell culture and methods of using the same
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
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
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
USD759075S1 (en) * 2014-04-11 2016-06-14 Nutonian, Inc. Display screen with graphical user interface
USD759076S1 (en) * 2014-04-18 2016-06-14 Nutonian, Inc. Display screen with graphical user interface
TWI743024B (en) 2014-06-06 2021-10-21 美商健臻公司 Perfusion culturing methods and uses thereof
TW202330904A (en) 2015-08-04 2023-08-01 美商再生元醫藥公司 Taurine supplemented cell culture medium and methods of use
WO2017065559A1 (en) * 2015-10-15 2017-04-20 (주)알테오젠 Method for producing fusion protein having igg fc domain
CN107460221B (en) * 2016-06-02 2021-01-15 正大天晴药业集团股份有限公司 Cell culture method for reducing protein polymer in anti-PD-L1 antibody
BR122023022045A2 (en) * 2017-10-16 2024-01-16 Regeneron Pharmaceuticals, Inc. METHODS AND SYSTEMS FOR CONTROLLING CELL CULTURE MEDIA CONDITIONS AND FOR REDUCING POST-TRANSLATION MODIFICATIONS OF A SECRETED PROTEIN
WO2020088180A1 (en) * 2018-11-02 2020-05-07 Wuxi Biologics (Shanghai) Co., Ltd. Cell culture process by intensified perfusion with continuous harvest and without cell bleeding
IL294321A (en) 2020-01-09 2022-08-01 Mersana Therapeutics Inc Site specific antibody-drug conjugates with peptide-containing linkers
WO2022254319A1 (en) * 2021-06-01 2022-12-08 Pfizer Inc. Cell culture method for producing sfgfr3 polypeptide
KR102682066B1 (en) * 2021-10-12 2024-07-05 프레스티지바이오로직스 주식회사 Method for manufacturing a population of antibodies
CN118006716A (en) * 2024-03-15 2024-05-10 通化安睿特生物制药股份有限公司 Method for preparing recombinant human albumin with high expression and low O-glycosylation level

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
WO2000036092A2 (en) 1998-12-17 2000-06-22 Biogen, Inc. METHOD FOR THE HIGH LEVEL EXPRESSION OF ACTIVE LYMPHOTOXIN-β RECEPTOR IMMUNOGLOBULIN CHIMERIC PROTEINS AND THEIR PURIFICATION
WO2003083066A2 (en) * 2002-03-27 2003-10-09 Immunex Corporation Methods for increasing polypeptide production

Family Cites Families (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) * 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5223409A (en) * 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
CA1340288C (en) 1988-09-02 1998-12-29 Robert Charles Ladner Generation and selection of novel binding proteins
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
JPH0396383A (en) 1989-09-08 1991-04-22 Riso Kagaku Corp Image forming device
ATE236249T1 (en) * 1989-09-12 2003-04-15 Hoffmann La Roche TFN-BINDING PROTEINS
GB8927546D0 (en) 1989-12-06 1990-02-07 Ciba Geigy Process for the production of biologically active tgf-beta
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5610033A (en) 1990-04-25 1997-03-11 Novo Nordisk A/S Method of producing proteins with FVIII activity and/or FVIII derivatives
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
EP0585287B1 (en) 1990-07-10 1999-10-13 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
GB9021679D0 (en) 1990-10-05 1990-11-21 Gorman Scott David Antibody preparation
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
WO1992009298A1 (en) * 1990-11-23 1992-06-11 Peptide Technology Ltd. The delay, prevention and/or reversal of cell senescence
DE69129154T2 (en) 1990-12-03 1998-08-20 Genentech, Inc., South San Francisco, Calif. METHOD FOR ENRICHING PROTEIN VARIANTS WITH CHANGED BINDING PROPERTIES
WO1992015677A1 (en) 1991-03-01 1992-09-17 Protein Engineering Corporation Process for the development of binding mini-proteins
ES2223040T3 (en) 1991-04-10 2005-02-16 The Scripps Research Institute HETERODIMERILE RECEIVER LIBRARIES USING PHAGEMIDS.
EP0940468A1 (en) * 1991-06-14 1999-09-08 Genentech, Inc. Humanized antibody variable domain
DE4122599C2 (en) 1991-07-08 1993-11-11 Deutsches Krebsforsch Phagemid for screening antibodies
US6136310A (en) 1991-07-25 2000-10-24 Idec Pharmaceuticals Corporation Recombinant anti-CD4 antibodies for human therapy
ES2322324T3 (en) 1992-08-21 2009-06-19 Vrije Universiteit Brussel IMMUMOGLOBULINS DESPROVISTAS OF LIGHT CHAINS.
US5399677A (en) 1993-12-07 1995-03-21 Genetics Institute, Inc. Mutants of bone morphogenetic proteins
GB2285446B (en) 1994-01-03 1999-07-28 Genentech Inc Thrombopoietin
US5516964A (en) 1994-01-21 1996-05-14 Sun Company, Inc. (R&M) Hydrocarbon isomerization using solid superacid catalysts comprising platinum metal
CA2761116A1 (en) 1995-04-27 1996-10-31 Amgen Fremont Inc. Human antibodies derived from immunized xenomice
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5721121A (en) * 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
US5830761A (en) * 1995-06-07 1998-11-03 Genetics Institute, Inc. Medium and methods for culturing mammalian cho cells
EP0859841B1 (en) * 1995-08-18 2002-06-19 MorphoSys AG Protein/(poly)peptide libraries
JP4306813B2 (en) 1995-09-19 2009-08-05 アスビオファーマ株式会社 New method for culturing animal cells
US5710023A (en) 1996-03-01 1998-01-20 Genetics Institute, Inc. IL-13 cytokine receptor chain
US6146847A (en) * 1996-11-01 2000-11-14 Genespan Corporation Stabilized transient gene expression
KR20080059467A (en) * 1996-12-03 2008-06-27 아브게닉스, 인크. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
EP0983303B1 (en) 1997-05-21 2006-03-08 Biovation Limited Method for the production of non-immunogenic proteins
AU2113199A (en) 1998-01-12 1999-07-26 Betagene, Inc. Media for neuroendocrine cells
ES2278463T3 (en) 1998-12-08 2007-08-01 Biovation Limited METHOD TO REDUCE THE IMMUNOGENICITY OF PROTEINS.
US7294481B1 (en) 1999-01-05 2007-11-13 Immunex Corporation Method for producing recombinant proteins
EP1185691B1 (en) 1999-04-30 2009-04-29 Novartis Vaccines and Diagnostics, Inc. Neisseria genomic sequences and methods of their use
JP3090657B1 (en) 1999-08-09 2000-09-25 伸子 蓮山 Zeolite manufacturing method and zeolite manufacturing apparatus
EP1103615A1 (en) 1999-11-25 2001-05-30 Universite De Geneve Vectors capable of immortalizing non-dividing cells and cells immortalized with said vectors
AUPR038200A0 (en) * 2000-09-26 2000-10-19 Beta Peptide Foundation Pty Ltd, The Compositions and methods for delaying, preventing, rejuvenating or reversing senescence
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US7829084B2 (en) 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US20030040095A1 (en) * 2001-03-16 2003-02-27 Achille Arini Method for the production of pharmaceutically active recombinant proteins
US20040058445A1 (en) 2001-04-26 2004-03-25 Ledbetter Jeffrey Alan Activation of tumor-reactive lymphocytes via antibodies or genes recognizing CD3 or 4-1BB
US6544076B2 (en) 2001-07-10 2003-04-08 Alan L. Pocrass Dual function RJ connector
JP4404631B2 (en) * 2001-11-19 2010-01-27 ノボ ノルディスク アクティーゼルスカブ Method for producing insulin compound
CA2417689C (en) 2002-03-05 2006-05-09 F. Hoffmann-La Roche Ag Improved methods for growing mammalian cells in vitro
GB0208041D0 (en) 2002-04-08 2002-05-22 Lonza Biologics Plc Method of culturing animal cells
CN100591759C (en) 2002-07-09 2010-02-24 巴克斯特国际有限公司 Animal protein free media for cultivation of cells
JP4510938B2 (en) 2002-07-15 2010-07-28 イミュネックス・コーポレーション Method and medium for regulating sialic acid addition of proteins produced by mammalian cells
US6924124B1 (en) 2002-08-23 2005-08-02 Immunex Corporation Feeding strategies for cell culture
US7261893B2 (en) 2002-10-22 2007-08-28 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
MXPA05006523A (en) 2002-12-23 2005-08-26 Squibb Bristol Myers Co Mammalian cell culture processes for protein production.
WO2004096833A2 (en) 2003-04-25 2004-11-11 Immunex Corporation Inducers of recombinant protein expression
CN1565631A (en) * 2003-06-27 2005-01-19 上海中信国健药业有限公司 Usage of fused protein of TNF receptor and globin in acute lung injury treating medicine
MXPA06004472A (en) 2003-10-27 2006-06-20 Wyeth Corp Removal of high molecular weight aggregates using hydroxyapatite chromatography.
WO2005064013A1 (en) 2003-12-23 2005-07-14 Bart Janssen The use of carnosine metabolism-associated genes to determine genetic predisposition/susceptibility to diabetic nephropathy
US7722915B2 (en) * 2004-07-07 2010-05-25 Coloplast A/S Preparation of hydrophillic coatings utilizing a 1,3-dioxolane compound
CN103172731A (en) 2004-07-15 2013-06-26 赞科股份有限公司 Optimized Fc variants
EP1781684A4 (en) 2004-08-25 2008-05-21 Amprotein Corp A novel chimeric polypeptide and use thereof
US7300773B2 (en) 2004-08-27 2007-11-27 Wyeth Research Ireland Limited Production of TNFR-Ig
US7335491B2 (en) * 2004-08-27 2008-02-26 Wyeth Research Ireland Limited Production of anti-abeta
TWI384069B (en) * 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
US7426440B2 (en) * 2004-09-24 2008-09-16 Nutritional Bioscience Ltd. Repair and protection factor scoring method for bioactive agents
US20070102622A1 (en) 2005-07-01 2007-05-10 Olsen Richard I Apparatus for multiple camera devices and method of operating same
WO2007011595A2 (en) 2005-07-15 2007-01-25 Neuren Pharmaceuticals Limited Neural regeneration peptides and antioxidants protect neurons from degeneration
DE102005034616B4 (en) * 2005-07-18 2008-07-03 Elringklinger Ag Fuel cell unit and fuel cell stack
AR058140A1 (en) 2005-10-24 2008-01-23 Wyeth Corp PROTEIN PRODUCTION METHOD USING ANTI-SENESCENCE COMPOUNDS
EP2495307B9 (en) * 2006-07-13 2018-05-02 Wyeth LLC Production of coagulation factor IX with improved glycosylation pattern
MX2009009240A (en) 2007-03-02 2009-09-08 Wyeth Corp Use of copper and glutamate in cell culture for production of polypeptides.
EP2188371B1 (en) 2007-08-09 2017-12-20 Wyeth LLC Use of perfusion to enhance production of fed-batch cell culture in bioreactors
JP2023544949A (en) 2021-09-15 2023-10-26 寧徳時代新能源科技股▲分▼有限公司 Battery cells, batteries, power consumption devices, manufacturing methods and equipment for battery cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
WO2000036092A2 (en) 1998-12-17 2000-06-22 Biogen, Inc. METHOD FOR THE HIGH LEVEL EXPRESSION OF ACTIVE LYMPHOTOXIN-β RECEPTOR IMMUNOGLOBULIN CHIMERIC PROTEINS AND THEIR PURIFICATION
WO2003083066A2 (en) * 2002-03-27 2003-10-09 Immunex Corporation Methods for increasing polypeptide production

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BORYS M C ET AL: "Culture pH affects expression rates and glycosylation of recombinant mouse placental lactogen proteins by Chinese hamster ovary (CHO) cells.", BIO/TECHNOLOGY (NATURE PUBLISHING COMPANY) JUN 1993, vol. 11, no. 6, June 1993 (1993-06-01), pages 720 - 724, XP002486307, ISSN: 0733-222X *
See also references of EP2139986A1
TRUMMER EVELYN ET AL: "Process parameter shifting: Part I. Effect of DOT, pH, and temperature on the performance of Epo-Fc expressing CHO cells cultivated in controlled batch bioreactors.", BIOTECHNOLOGY AND BIOENGINEERING 20 AUG 2006, vol. 94, no. 6, 20 August 2006 (2006-08-20), pages 1033 - 1044, XP002486305, ISSN: 0006-3592 *
YOON SUNG KWAN ET AL: "Effect of culture pH on erythropoietin production by chinese hamster ovary cells grown in suspension at 32.5 and 37.0 °C", BIOTECHNOLOGY AND BIOENGINEERING, vol. 89, no. 3, 5 February 2005 (2005-02-05), pages 345 - 356, XP002486306, ISSN: 0006-3592 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2188371B1 (en) 2007-08-09 2017-12-20 Wyeth LLC Use of perfusion to enhance production of fed-batch cell culture in bioreactors
EP3330370B1 (en) 2010-04-26 2021-02-24 Novartis AG Process for cultivation of cho cells
EP2563906B1 (en) 2010-04-26 2017-11-08 Novartis AG Process for cultivation of cho cells
AU2011350456B2 (en) * 2010-12-28 2016-05-26 Chugai Seiyaku Kabushiki Kaisha Animal cell culturing method
WO2012091124A1 (en) 2010-12-28 2012-07-05 中外製薬株式会社 Animal cell culturing method
US12018070B2 (en) 2014-10-15 2024-06-25 Alexion Pharmaceuticals, Inc. Methods of shifting an isoelectric profile of a protein product and uses thereof
US11046772B2 (en) 2015-03-26 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Process of production with controlled copper ions
EP3363811B1 (en) 2015-10-15 2023-04-19 Alteogen, Inc. Method for producing fusion protein having igg fc domain
WO2018073365A1 (en) 2016-10-19 2018-04-26 F. Hoffmann-La Roche Ag Method for producing an immunoconjugate
JP2020507336A (en) * 2017-02-17 2020-03-12 ロンザ リミテッドLonza Limited Methods for producing biological product variants
WO2018151819A1 (en) * 2017-02-17 2018-08-23 Lonza Ltd A method for producing biologic product variants
JP7252896B2 (en) 2017-02-17 2023-04-05 ロンザ リミテッド Methods of Producing Biological Product Variants
WO2020180967A1 (en) * 2019-03-04 2020-09-10 Amgen Inc. In vivo reversibility of high molecular weight species

Also Published As

Publication number Publication date
CL2008001159A1 (en) 2008-11-03
JP2010524504A (en) 2010-07-22
DK2139987T3 (en) 2013-09-02
CN101668845B (en) 2015-11-25
HUE034776T2 (en) 2018-02-28
CA2684727C (en) 2018-09-04
JP5557736B2 (en) 2014-07-23
US20080269132A1 (en) 2008-10-30
IL201720A0 (en) 2011-08-01
DK2139987T5 (en) 2014-01-27
MX2009011362A (en) 2009-11-05
ES2424217T9 (en) 2014-01-27
PE20090731A1 (en) 2009-07-15
AU2008242633A1 (en) 2008-10-30
PT2139986T (en) 2017-11-14
RU2009139054A (en) 2011-05-27
US9988662B2 (en) 2018-06-05
PT2139987E (en) 2013-08-22
KR20100016271A (en) 2010-02-12
BRPI0810482A2 (en) 2014-10-07
CN101668845A (en) 2010-03-10
IL201719A (en) 2015-09-24
CO6241166A2 (en) 2011-01-20
EP2139986B1 (en) 2017-09-06
KR20100017211A (en) 2010-02-16
HK1142095A1 (en) 2010-11-26
WO2008131375A1 (en) 2008-10-30
IL201719A0 (en) 2011-08-01
PL2139986T3 (en) 2018-01-31
EP2139987A1 (en) 2010-01-06
DK2139986T3 (en) 2017-10-02
PE20090153A1 (en) 2009-04-23
JP2010524503A (en) 2010-07-22
AU2008242633B2 (en) 2014-09-18
KR101540124B1 (en) 2015-07-28
JP5881755B2 (en) 2016-03-09
RU2478702C2 (en) 2013-04-10
CA2685552A1 (en) 2008-10-30
EP2139986A1 (en) 2010-01-06
CA2684727A1 (en) 2008-10-30
CA2685552C (en) 2016-02-23
AR066239A1 (en) 2009-08-05
ES2646090T3 (en) 2017-12-12
BRPI0810511A2 (en) 2014-10-07
EP2139987B9 (en) 2013-11-13
AU2008242632B2 (en) 2013-11-28
JP2014113161A (en) 2014-06-26
ES2424217T3 (en) 2013-09-30
PA8778001A1 (en) 2008-11-19
MX2009011363A (en) 2009-11-05
KR101560421B1 (en) 2015-10-15
CO6241165A2 (en) 2011-01-20
HK1216261A1 (en) 2016-10-28
AU2008242632A1 (en) 2008-10-30
ZA200907426B (en) 2010-08-25
US12054762B2 (en) 2024-08-06
IL201720A (en) 2016-03-31
EP2139987B1 (en) 2013-07-03
RU2009138226A (en) 2011-05-27
CN105039473A (en) 2015-11-11
TW200902708A (en) 2009-01-16
SI2139987T1 (en) 2013-09-30
CN101679943A (en) 2010-03-24
SI2139986T1 (en) 2017-11-30
KR20140132016A (en) 2014-11-14
PL2139987T3 (en) 2013-11-29
CL2008001156A1 (en) 2009-01-16
AR066238A1 (en) 2009-08-05
US20080274507A1 (en) 2008-11-06
PA8777701A1 (en) 2008-11-19
TW200902709A (en) 2009-01-16

Similar Documents

Publication Publication Date Title
EP2139986B1 (en) Use of low temperature and/or low ph in cell culture
US20230365920A1 (en) Use Of Perfusion To Enhance Production Of Fed-batch Cell Culture In Bioreactors
AU717847B2 (en) Process for controlling sialylation of proteins produced by mammalian cell culture
US9447188B2 (en) Method for controlling the main complex N-glycan structures and the acidic variants and variability in bioprocesses producing recombinant proteins
US6528286B1 (en) Mammalian cell culture process for producing glycoproteins
US9096879B2 (en) Method of supplementing culture media to prevent undesirable amino acid substitutions
CN104928336A (en) Method for the production of a glycosylated immunoglobulin
US20120100553A1 (en) Cho/cert cell lines
US8822198B2 (en) Method for optimizing a biopharmaceutical production process
KR20230045615A (en) how to make protein
WO2023079058A1 (en) Cell culture with reduced production of lactate
WO2023235762A2 (en) Commercial-scale recombinant protein production in rat hybridoma cells

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880013275.1

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08746526

Country of ref document: EP

Kind code of ref document: A1

REEP Request for entry into the european phase

Ref document number: 2008746526

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2008746526

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2684727

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 09117978

Country of ref document: CO

Ref document number: MX/A/2009/011362

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2010506424

Country of ref document: JP

Ref document number: 201720

Country of ref document: IL

Ref document number: 6775/DELNP/2009

Country of ref document: IN

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008242632

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 20097024259

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2009139054

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2008242632

Country of ref document: AU

Date of ref document: 20080422

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0810511

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20091023