US20170145465A1 - Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins - Google Patents

Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins Download PDF

Info

Publication number
US20170145465A1
US20170145465A1 US15/425,003 US201715425003A US2017145465A1 US 20170145465 A1 US20170145465 A1 US 20170145465A1 US 201715425003 A US201715425003 A US 201715425003A US 2017145465 A1 US2017145465 A1 US 2017145465A1
Authority
US
United States
Prior art keywords
antibody
recombinant protein
modulating
culture medium
glycosylation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/425,003
Inventor
Alane E. Wentz
Don Hemmavanh
Joseph G. Matuck
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
Original Assignee
AbbVie Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by AbbVie Inc filed Critical AbbVie Inc
Priority to US15/425,003 priority Critical patent/US20170145465A1/en
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HEMMAVANH, Don, MATUCK, JOSEPH G., WENTZ, ALANE E.
Publication of US20170145465A1 publication Critical patent/US20170145465A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • C12N5/005Protein-free medium
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the present invention is directed to methods for adjusting the glycosylation profile of recombinantly-expressed proteins by supplementing the culture media metal ions.
  • Protein glycosylation is a well-established post translation modification that influences the folding (Walsh et al. (1990) Biochemistry 29(26):6250-7), solubility (Leavitt et al. (1977) J. Biol. Chem. 252(24):9018-23), immunogenicity (Shields et al., (2002) J. Biol. Chem. 277(30):26733-40; Rudd et al., (2001) Science 291(5512):2370-6; Lowe and Marth, (2003) Annu. Rev. Biochem. 72:643-91, binding (Wallick et al., (1988) J. Exp. Med.
  • oligosaccharides are added onto asparagine residues of protein therapeutics in a step-wise manner followed by the step-wise removal and addition of individual monosaccharides in the endoplasmic reticulum (ER), and Golgi apparatus.
  • ER endoplasmic reticulum
  • FIG. 1 This progression of potential glycoforms is summarized in FIG. 1 . Briefly, mannose is removed to allow the addition of N-acetylglucosamine (GlcNAc) followed by the addition of galactose.
  • GlcNAc N-acetylglucosamine
  • the glycosylation profile for a given therapeutic glycoprotein often includes a mixture of these different glycoforms.
  • the impact of protein glycosylation on the physiochemical properties of recombinant proteins is well-documented.
  • the protein glycosylation profile is therefore a critical process parameter in the manufacturing of recombinant protein therapeutics.
  • the present invention is directed to methods for modulating the glycosylation profile of proteins recombinantly expressed in cultured eukaryotic cells by supplementing a chemically defined cell culture media with millimolar amounts of a metal ion.
  • the metal ion is iron
  • the N-glycan oligosaccharide profile of recombinant proteins are increased by supplementing the chemically defined cell culture media with ferrous sulfate, ferric citrate, or ferric nitrate.
  • ferrous sulfate ferric citrate
  • ferric nitrate ferric nitrate
  • the present invention provides for a method for modulating the glycosylation profile of a recombinant protein expressed in a eukaryotic cell comprising the culturing of a eukaryotic cell in a protein-free, chemically defined culture medium supplemented with an effective amount of a metal salt and expressing a recombinant protein in the eukaryotic cell, wherein the amount of the metal salt is effective at modulating the glycosylation profile of the recombinant protein.
  • the metal salt can be an iron salt such as ferric nitrate, ferric citrate or ferrous sulfate.
  • the metal ion further includes zinc, copper, chromium and/or manganese.
  • the effective amount of a metal salt in the culture medium can be less than about 1 ⁇ M.
  • the effective amount of zinc, chromium or copper ions in the culture medium is less than about 1 ⁇ M.
  • the effective amount of a metal salt in the culture medium can be from about 1 ⁇ M to about 100 ⁇ M.
  • the effective amount of ferrous sulfate in the culture media can be about 20 ⁇ M.
  • the effective amount of a metal salt in the culture medium can be from about 0.1 mM to about 10 mM of metal ions.
  • the effective amount of magnesium salt in the culture medium can be about 1 to about 10 mM.
  • the eukaryotic cell can be a mammalian cell including Chinese hamster ovary (CHO) cells, Chinese hamster ovary (CHO) cells, CHO DUX-B 11 cells, mouse myeloma SP2/0, human HT-1080, NSO myeloma cell line, mouse L cells, mouse A9 cells, baby hamster kidney cells, C127 cells, COS cells and PC8 cells.
  • CHO Chinese hamster ovary
  • CHO Chinese hamster ovary
  • CHO DUX-B 11 cells mouse myeloma SP2/0
  • human HT-1080 human HT-1080
  • NSO myeloma cell line mouse L cells
  • mouse A9 cells mouse A9 cells
  • baby hamster kidney cells C127 cells
  • COS cells COS cells and PC8 cells.
  • the recombinantly expressed protein can be any of a number of protein binders, such as an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
  • the recombinantly expressed protein binder can be humanized.
  • the addition of the metal ion to the chemically defined cell culture medium reduces NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein.
  • the addition of the metal ion to the chemically defined cell culture medium can reduce NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein by at least 15%.
  • the addition of the metal ion to the chemically defined cell culture medium increases galactosylaton of the expressed recombinant glycoprotein.
  • the addition of the metal ion to the chemically defined cell culture medium increases NA1F and NA2F glycosylation of the expressed recombinant protein.
  • the addition of the metal ion to the chemically defined cell culture medium can increase NA1F glycosylation of the expressed recombinant protein by at least about 4 fold.
  • the addition of the metal ion to the chemically defined cell culture medium can increase NA2F glycosylation of the expressed recombinant protein by at least about 7 fold.
  • the present invention is directed to a cell culture medium for modulating the glycosylation profile of a recombinantly-expressed protein, comprising a protein-free chemically defined culture medium supplemented with an effective amount of a metal salt effective at modulating the glycosylation profile of a recombinantly-expressed protein, wherein the metal salt is not ferric citrate.
  • the metal salt can be an iron salt such as ferric nitrate.
  • the effective amount of the metal salt is about 0.2 mM to about 1 mM.
  • the recombinantly-expressed protein is a protein binder, such as an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
  • FIG. 1 depicts a simplified linear reaction view of the N-glycan biosynthetic pathway in mammalian cells.
  • FIG. 2 shows an exemplary depiction of the cell culture performance of Antibody 1 in ferric nitrate supplemented media.
  • FIG. 3 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in ferric nitrate supplemented media.
  • FIG. 4 shows an exemplary depiction of the cell culture performance of Antibody 1 in various iron supplemented media.
  • FIG. 5 shows an exemplary depiction of N-glycan oligosaccharide results of Antibody 1 in various iron supplemented media.
  • FIG. 6 shows an exemplary depiction of the cell culture performance of Antibody 2 in various iron supplemented media.
  • FIG. 7 shows an exemplary depiction of the titer and N-glycan oligosaccharide results of Antibody 2 in various iron supplemented media.
  • FIG. 8 shows an exemplary depiction of the cell culture performance of DVD-Ig in various iron supplemented media.
  • FIG. 9 shows an exemplary depiction of the titer and N-glycan oligosaccharide results of DVD-Ig in various iron supplemented media.
  • FIG. 10 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 for various iron supplemented feed media.
  • FIG. 11 shows an exemplary depiction of the N-glycan oligosaccharide results in bioreactors for Antibody 1 in various iron supplemented media.
  • FIG. 12 shows an exemplary depiction of the N-glycan oligosaccharide results in bioreactors for DVD-Ig 2 in various iron supplemented media.
  • FIG. 13 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various metal ion supplemented media.
  • FIG. 14 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various iron, transferrin, and galactose supplemented media.
  • FIG. 15 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various iron, manganese, and galactose supplemented media.
  • chemically defined medium refers to a synthetic growth medium in which the identity and concentration of all the components are known. Chemically defined mediums do not contain bacterial, yeast, animal, or plant extracts, animal serum or plasma although they may or may not include individual plant or animal-derived components (e.g., proteins, polypeptides, etc). Chemically defined media may contain inorganic salts such as phosphates, sulfates, and the like needed to support growth.
  • the carbon source is defined, and is usually a sugar such as glucose, lactose, galactose, and the like, or other compounds such as glycerol, lactate, acetate, and the like.
  • a chemically defined cell culture medium contains no more than micromolar amounts of any metal ion.
  • Examples of commercially available chemically defined mediums include, but are not limited to, various Dulbecco's Modified Eagle's (DME) mediums (Sigma-Aldrich Co; SAFC Biosciences, Inc), Ham's Nutrient Mixture (Sigma-Aldrich Co; SAFC Biosciences, Inc), combinations thereof, and the like.
  • DME Dulbecco's Modified Eagle's
  • SAFC Biosciences, Inc Ham's Nutrient Mixture
  • combinations thereof and the like.
  • the chemically defined medium does not contain non-ionic detergents such as Pluronic® F-68.
  • serum-free culture medium or “serum-free culture media” refers to a culture medium that is not supplemented with an animal serum. Although oftentimes serum-free mediums are chemically defined mediums, serum-free mediums may be supplemented with discrete animal or plant proteins or protein fractions. Methods of preparing serum-free culture medium are known in the art, for example in U.S. Pat. Nos. 6,171,825; 6,936,441 and EP 1849862.
  • an effective amount means a sufficient amount of a metal ion to modulate the glycosylation profile of a recombinantly-expressed protein.
  • a metal ion may be any positively charged metal ion selected from, but not limited to, the following Al +3 , Cr +2 , Cr +3 , Cd +1 , Cd +2 , Co +2 , Co +3 , Ca +2 , Mg +2 , Ni +2 , Ti +2 , Ti +3 , Ti +4 , V +2 , V +3 , V +5 , Sr +2 , Fe +2 , Fe +3 , Au +2 , Au +3 , Au +1 , Ag +2 , Ag +1 , Pd +2 , Pd +4 , Pt +2 , Pt +4 , Cu +1 , Cu +2 , Mn +2 , Mn +3 , Mn +4 , Zn +2 , Se +4+2 and mixtures thereof.
  • the ions are generally added to the medium in the form of salts such as chlorides and sulphates.
  • the metal ion may not include ferr
  • a mammalian cell can be a mammalian cell line including, but not limited to, CHO cells, human embryonic kidney cells (HEK), baby hamster kidney (BHK), mouse myeloma (NSO), and human retinal cells (PERC.6).
  • CHO cells human embryonic kidney cells (HEK), baby hamster kidney (BHK), mouse myeloma (NSO), and human retinal cells (PERC.6).
  • antibody as referred to herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chains thereof.
  • An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • antibody portion refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., TNF ⁇ , IL-12). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • an antigen e.g., TNF ⁇ , IL-12
  • binding fragments encompassed within the term “antigen-binding portion” of an antibody include: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 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 CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH or VL domain; and (vi) an isolated complementarity determining region (CDR).
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH1 domains
  • a F(ab′)2 fragment a bivalent fragment comprising two Fab fragments
  • 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 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody.
  • the antibody fragment is selected from the group consisting of an Fab, an Fd, an Fd', a single chain Fv (scFv), an scFva, and a domain antibody (dAb).
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • these other proteins or peptides can have functionalities that allow for the purification of antibodies or antigen-binding portions thereof or allow for their association with each other or other molecules.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric single chain variable fragment (scFv) molecules (Kipriyanov et al.
  • Antibody portions such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques.
  • Two antibody domains are “complementary” where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature.
  • a VH domain and a VL domain of an antibody are complementary; two VH domains are not complementary, and two VL domains are not complementary.
  • Complementary domains may be found in other members of the immunoglobulin superfamily, such as the Va and VP (or gamma and delta) domains of the T-cell receptor.
  • domain refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain.
  • single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains.
  • variable domains and modified variable domains, for example, in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.
  • Variable domains of the present disclosure may be combined to form a group of domains; for example, complementary domains may be combined, such as VL domains being combined with VH domains. Non-complementary domains may also be combined. Domains may be combined in a number of ways, involving linkage of the domains by covalent or non-covalent means.
  • a “dAb” or “domain antibody” refers to a single antibody variable domain (VH or VL) polypeptide that specifically binds antigen.
  • antigen binding region or “antigen binding site” refers to the portion(s) of an antibody molecule, or antigen binding portion thereof, which contains the amino acid residues that interact with an antigen and confers on the antibody its specificity and affinity for the antigen.
  • epitope is meant to refer to that portion of any molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen binding regions.
  • first and second “epitopes” are understood to be epitopes which are not the same and are not bound by a single monospecific antibody, or antigen-binding portion thereof.
  • DVD-Ig or “dual-variable-domain-immunoglobulin” refers to a protein containing two or more antigen binding sites and is a tetravalent or multivalent binding protein.
  • the DVD-Ig comprises two heavy chain DVD polypeptides and two light chain DVD polypeptides; each half of a DVD-Ig comprises a heavy chain DVD polypeptide, and a light chain DVD polypeptide, and two antigen binding sites.
  • Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of six CDRs involved in antigen binding per antigen binding site.
  • the description and generation of DVD-Ig molecules are detailed in the U.S. Pat. No. 7,612,181 B2.
  • recombinant antibody refers to antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et al. (1992) Nucl. Acids Res. 20:6287-6295) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of particular immunoglobulin gene sequences (such as human immunoglobulin gene sequences) to other DNA sequences.
  • recombinant antibodies include chimeric, CDR-grafted and humanized antibodies.
  • human antibody refers to antibodies having variable and constant regions corresponding to, or derived from, human germline immunoglobulin sequences as described by, for example, Kabat et al. (See Kabat, et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • the human antibodies of the present disclosure may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • Recombinant human antibodies of the present disclosure have variable regions, and may also include constant regions, derived from human germline immunoglobulin sequences (See Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242).
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • such recombinant antibodies are the result of selective mutagenesis or backmutation or both.
  • backmutation refers to a process in which some or all of the somatically mutated amino acids of a human antibody are replaced with the corresponding germline residues from a homologous germline antibody sequence.
  • the heavy and light chain sequences of a human antibody of the present disclosure are aligned separately with the germline sequences in the VBASE database to identify the sequences with the highest homology. Differences in the human antibody of the present disclosure are returned to the germline sequence by mutating defined nucleotide positions encoding such different amino acid.
  • the role of each amino acid thus identified as candidate for backmutation should be investigated for a direct or indirect role in antigen binding and any amino acid found after mutation to affect any desirable characteristic of the human antibody should not be included in the final human antibody.
  • chimeric antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.
  • CDR-grafted antibody refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
  • humanized antibody refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences.
  • a non-human species e.g., a mouse
  • human CDR-grafted antibody in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences.
  • the oligosaccharide structure attached to the peptide chain is known as a “glycan” molecule.
  • N-linked glycans or N-linked oligosaccharides
  • 0-linked glycans or O-linked oligosaccharides
  • Peptides comprising “0-linked glycans” have a saccharide attached to the hydroxy oxygen of serine, threonine, tyrosine, hydroxylysine, and or hydroxyproline residue in the primary protein.
  • N-glycans are N-glycosylated at an amide nitrogen of an asparagine or an arginine residue in a protein via an N-acetylglucosamine residue.
  • N-linked glycosylation sites occur in the peptide primary structure containing, for example, the amino acid sequence asparagine-X-serine/threonine, where X is any amino acid residue except proline and aspartic acid.
  • All N-linked oligosaccharides have a common “pentasaccharide core” of Man 3 GlcNAc 2 .
  • Man refers to mannose
  • Glc refers to glucose
  • NAc refers to N-acetyl
  • GlcNAc refers to N-acetylglucosamine
  • the pentasaccharide core is also referred to as the “trimannose core” or the “paucimannose core.” (see FIG. 1 )
  • N-glycans differ with respect to the presence of, and/or in the number of branches (also called “antennae”) comprising peripheral sugars such as N-acetylglucosamine, galactose, N-acetylgalactosamine, N-acetylneuraminic acid, fucose and sialic acid that are added to the Man 3 GlcNAc 2 core structure.
  • this structure may also contain a core fucose molecule and/or a xylose molecule.
  • N-glycans are classified according to their branched constituents (e.g., oligomannose-type, complex, or hybrid).
  • An “oligomannose-type” or “high mannose-type” N-glycan has five or more mannose residues.
  • a “complex-type” N-glycan typically has at least one GlcNAc attached to the 1,3 mannose arm and at least one GlcNAc attached to the 1,6 mannose arm of a pentasaccharide core.
  • Complex-type N-glycans may also have galactose (“Gal”) or N-acetylgalactosamine residues that are optionally modified with sialic acid or derivatives, e.g., N-acetyl neuraminic acid.
  • Complex-type N-glycans may also have intrachain substitutions comprising “bisecting” GlcNAc, and core fucose (“Fuc”).
  • Complex N-glycans may also have multiple antennae on the pentasaccharide core and are, therefore, also referred to as “multiple antennary-type glycans.”
  • a “hybrid-type” N-glycan comprises at least one GlcNAc on the terminal of the 1,3 mannose arm of the pentasaccharide core and zero or more mannoses on the 1,6 mannose arm of the trimannose core.
  • NGA2F +NGA2F-GlcNAc refers to the agalactosyl fucosylated biantennary oligosaccharides.
  • NGA2F has the structure:
  • NA1F and NA2F refer to the fraction of galactose-containing fucosylated biantennary oligosaccharides.
  • NGA2F glycan is found on many mammalian glycoproteins including human IgG and is a substructure of bi-antennary N-linked oligosaccharides such as A2F, A1F, and NA2F.
  • glycoform denotes a type of polypeptide with a specific type and distribution of polysaccharides attached to, i.e. two polypeptides would be of the same glycoform if they comprise glycans with the same number, kind, and sequence of monosaccharides, i.e. have the same “glycosylation profile.” Glycoproteins often consist of a number of different glycoforms.
  • glycosylation profile refers to the properties of the glycans of a glycosylated polypeptide. These properties include, but are not limited to, the glycosylation sites, or the glycosylation site occupancy, or the identity, structure, composition or quantity of the glycan attached to a recombinantly expressed polypeptide.
  • CHO cell lines CHO DUX-B 11 origin based on a DHFR (dihydrofolate reductase) expression system
  • DHFR dihydrofolate reductase
  • DVD-Ig dual variable domain immunoglobulin
  • CDBM chemically defined basal medium
  • CDFM chemically defined feed medium
  • the cell line producing Antibody 1 (also called ABBV-696) was cultured in shake flasks in fedbatch mode after an abbreviated seed train in two independent experiments.
  • Ferric nitrate was supplemented into chemically-defined basal and feed media at concentrations of 0.1, 1.0, and 5.0 mM (Experiment 1) and 0.1, 0.5, 1.0, and 2.0 mM (Experiment 2), and compared to an unsupplemented control condition.
  • Viable cell density (VCD) and cell viability values were measured through trypan blue exclusion via ViCell automated cell counters (Beckman Coulter, Fullerton, Calif.), glucose and lactate values were measured with a ABL-805 (Radiometer Medical, Denmark) blood gas analyzer.
  • Antibody titers were measured from crude cell culture harvests on a Poros ATM (Life Technologies, Carlsbad, Calif.) Protein A affinity column using an Agilent (Santa Clara, Calif.) 1200 Series HPLC, or equivalent, operating with a low pH, step elution gradient with detection at 280 nm. Absolute concentrations were assigned with respect to reference standard calibration curves. Purified antibodies subjected to additional analytical characterization were purified using MabSelectTM Protein A (GE Healthcare, Piscataway, N.J.) using a low pH, step elution gradient, followed by buffer exchange using Corning Life Sciences (Tewksbury, Mass.) Spin Concentrator X UF columns according to the manufacturers' recommended procedures.
  • the viable cell density, viability, and harvest titer results are shown in FIG. 2 .
  • ferric nitrate generally facilitated an increase in VCD, with the exception of the highest concentration (5.0 mM) supporting the lowest peak VCD.
  • Viability results were comparable to the control, i.e. unsupplemented conditions, with the exception of the 5.0 mM condition, which facilitated a drop in cell viability that was larger, and earlier in the culture, compared to control conditions.
  • the glycosylation profile of the recombinantly expressed proteins was then determined using standard procedures. Approximately 200 ⁇ g of Protein A purified samples from Cell Lines 1 and 2 were then treated with N-glycanase at 37° C. overnight to remove the N-glycans from the protein. The protein was precipitated and the supernatant was taken for subsequent chemical derivatization of the reducing end of the released glycans with 2-aminobenzamide (2-AB) dye. Following the derivatization step, the excess labeled was removed using clean up cartridges and the samples were analyzed using normal phase HPLC with fluorescent detection. Mobile phase A was 100% acetonitrile and mobile phase B was 50 mM ammonium formate pH 4.4.
  • the glycan were eluted from a polyamide column (Prozyme, Hayward, Calif.) using a shallow gradient.
  • the labeled glycans were detected using a fluorescence detector with an excitation wavelength of 330 nm and an emission wavelength of 420 nm.
  • N-glycan oligosaccharide results were quite different between the variously supplemented cultures and the unsupplemented control as shown in FIG. 3 .
  • Data in each experiment for each oligosaccharide was normalized to the level observed in the 1.0 mM condition for that particular experiment.
  • the addition of iron shifted the distribution toward increased galactosylation, reducing the total NGA2F (NGA2F and NGA2F-GlcNAc) species thereby increasing the total NA1F (NA1F plus NA1F-GlcNAc) and NA2F species.
  • a concentration-dependent response was observed below 1.0 mM ferric nitrate.
  • FIG. 4 highlights the cell culture results observed through the use of iron supplementation into the basal medium. All iron supplemented cultures showed a higher viable cell density compared to the unsupplemented control. The results indicate a significant increase in peak VCD from 9 ⁇ 10 6 cells/mL for the control versus 12-14 ⁇ 10 6 cells/mL for the iron supplemented cultures. At the end of the culture period, the final titer ratio was similar to the control suggesting a slight drop in overall antibody productivity upon inclusion of these particular concentrations of iron, consistent with the results observed from the previous shake flask cultures.
  • the N-glycan glycoform profile was measured with the results highlighted in FIG. 5 . Similar to the initial ferric nitrate shake flask results, there was a significant increase in overall galactosylation. The total NGA2F species was reduced 15-30% with the least impact from ferrous sulfate and the most significant impact from the ferric nitrate or ferric citrate at higher concentrations. Corresponding increases in the total NA1F and NA2F species were observed as well. Ferrous sulfate supplementation had modest 3-fold increases in both total NA1F and NA2F species. Ferric nitrate supplementation provided 5-fold increases in total NA1F with 7- to 9-fold increases in NA2F.
  • Ferric citrate supplementation provided very similar results to ferric nitrate with a concentration dependence observed below 1.0 mM as previously observed with ferric nitrate.
  • the absolute value of the fold increase in NA2F varied between experiments and relies heavily on quantification of the very small amount of this species present in the control culture. Nonetheless, significant increases were observed in NA2F with the addition of iron to the basal medium.
  • the cell line expressing Antibody 2 (also called ABT-806) was evaluated in shake flask culture in fedbatch mode. Ferric citrate or ferric nitrate was supplemented into the basal medium at various concentrations to evaluate the resulting impact on cell culture performance, as well as the protein glycosylation profile. The unsupplemented control was performed in duplicate. The viable cell density, viability, and harvest titer results are shown in FIGS. 6 and 7 . Ferric citrate supplementation at either 0.1 or 1.0 mM improved overall cell growth with higher peak viable cell density (increased by 4 ⁇ 10 6 cells/mL) while ferric nitrate at 0.1 mM had very little impact when compared to the unsupplemented control. Contrary to Antibody 1, all iron-supplemented conditions had a positive impact to the titer for Antibody 2.
  • FIG. 8 shows the cell culture viable cell density and viability performance for one representative from each experimental condition. The average harvest titer for the triplicates and the relative oligosaccharide profile results are in FIG. 9 .
  • Iron supplementation had minimal impact to the cell culture growth performance and harvest titer. However, the impact to the galactosylated species is similar to that observed for both Antibody 1 and 2 with iron supplementation. Increased concentration of both ferric nitrate and ferric citrate supplemented to the culture provided increased galactosylation as observed previously with both antibody-expressing cell lines. The total NGA2F species declined by approximately 20%, while the total NA1F species increased 3- to 9-fold, and the NA2F species increased 50- to 100-fold compared to the unsupplemented control. Sodium citrate had no effect on the oligosaccharide profile compared to the unsupplemented control.
  • FIG. 10 shows the impact to the relative oligosaccharide profile and that regardless of where the iron is introduced to the cell culture (basal versus feed medium), the galactosylation of the antibody increases as observed previously.
  • FIG. 12 shows the similar effect with the cell line expressing a DVD-Ig molecule, however the impact to this DVD-Ig molecule is limited to the total NA1F species as the NA2F species is not observed when this cell line is cultured in chemically defined medium.
  • the same cell line expressing this DVD-Ig molecule grown in a complex cell culture medium contains a significantly different oligosaccharide profile and iron supplementation has little impact on altering this profile.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Protein glycosylation greatly influences the structure, function, and pharmacokinetics of recombinant proteins. Here, growth media supplemented with metal ions is shown to modulate the protein glycosylation profile of recombinant proteins expressed in a variety of eukaryotic cell lines. In particular, millimolar amounts of ferric salts (e.g. ferric nitrate, ferric citrate, etc.) significantly increased the percentage of galactosylated N-glycan species. The effect on protein glycosylation was concentration-dependent manner up to 1.0 mM across multiple cell lines without adverse effects on cell viability. The use of iron supplements in cell culture media provides an efficient and effective approach towards the specific re-targeting of N-glycan glycoform profiles of recombinantly expressed proteins.

Description

    RELATED APPLICATIONS
  • The present application is a continuation of U.S. patent application Ser. No. 14/506,372, filed on Oct. 3, 2014, which in turn claims priority from U.S. Provisional Patent Application No. 61/886,855, filed on Oct. 4, 2013. The entire contents of each of the foregoing applications are incorporated herein by reference.
  • FIELD
  • The present invention is directed to methods for adjusting the glycosylation profile of recombinantly-expressed proteins by supplementing the culture media metal ions.
  • BACKGROUND
  • Protein glycosylation is a well-established post translation modification that influences the folding (Walsh et al. (1990) Biochemistry 29(26):6250-7), solubility (Leavitt et al. (1977) J. Biol. Chem. 252(24):9018-23), immunogenicity (Shields et al., (2002) J. Biol. Chem. 277(30):26733-40; Rudd et al., (2001) Science 291(5512):2370-6; Lowe and Marth, (2003) Annu. Rev. Biochem. 72:643-91, binding (Wallick et al., (1988) J. Exp. Med. 168(3):1099-109), stability (Wyss and Wagner, (1996) Curr. Opin. Biotechnol. 7(4):409-16), and pharmacokinetics (PK) (Millward, T. A., et al. (2008) Biologicals 36(1):41-7; Gross et al., (1988) Eur. J. Biochem. 173(3):653-9; Clincke et al., (2011) BMC Proceedings 5 (Suppl 8): p114-115) of recombinant protein therapeutics (reviewed in Hossler et al. Glycobiology (2009) vol. 19 pp. 936-949).
  • During the N-glycosylation biosynthetic pathway, oligosaccharides are added onto asparagine residues of protein therapeutics in a step-wise manner followed by the step-wise removal and addition of individual monosaccharides in the endoplasmic reticulum (ER), and Golgi apparatus. This progression of potential glycoforms is summarized in FIG. 1. Briefly, mannose is removed to allow the addition of N-acetylglucosamine (GlcNAc) followed by the addition of galactose. The glycosylation profile for a given therapeutic glycoprotein often includes a mixture of these different glycoforms.
  • The impact of protein glycosylation on the physiochemical properties of recombinant proteins is well-documented. The protein glycosylation profile is therefore a critical process parameter in the manufacturing of recombinant protein therapeutics.
  • For the foregoing reasons, there is an unmet need in the art for the consistent and reliable glycosylation of recombinant biologics during manufacture.
  • SUMMARY
  • The present invention is directed to methods for modulating the glycosylation profile of proteins recombinantly expressed in cultured eukaryotic cells by supplementing a chemically defined cell culture media with millimolar amounts of a metal ion.
  • In one embodiment, the metal ion is iron.
  • In other embodiments, the N-glycan oligosaccharide profile of recombinant proteins are increased by supplementing the chemically defined cell culture media with ferrous sulfate, ferric citrate, or ferric nitrate. The observed effect of these metal ions on glycosylation is scale or vessel independent and occurs even if the metal irons are introduced to the culture via the feed medium.
  • In another embodiment, the present invention provides for a method for modulating the glycosylation profile of a recombinant protein expressed in a eukaryotic cell comprising the culturing of a eukaryotic cell in a protein-free, chemically defined culture medium supplemented with an effective amount of a metal salt and expressing a recombinant protein in the eukaryotic cell, wherein the amount of the metal salt is effective at modulating the glycosylation profile of the recombinant protein.
  • The metal salt can be an iron salt such as ferric nitrate, ferric citrate or ferrous sulfate. In certain embodiments, the metal ion further includes zinc, copper, chromium and/or manganese.
  • In certain embodiments, the effective amount of a metal salt in the culture medium can be less than about 1 μM.
  • For example, in one embodiment, the effective amount of zinc, chromium or copper ions in the culture medium is less than about 1 μM.
  • In other embodiments, the effective amount of a metal salt in the culture medium can be from about 1 μM to about 100 μM.
  • For example, in one embodiment, the effective amount of ferrous sulfate in the culture media can be about 20 μM.
  • In other embodiments, the effective amount of a metal salt in the culture medium can be from about 0.1 mM to about 10 mM of metal ions.
  • For example, in one embodiment, the effective amount of magnesium salt in the culture medium can be about 1 to about 10 mM.
  • The eukaryotic cell can be a mammalian cell including Chinese hamster ovary (CHO) cells, Chinese hamster ovary (CHO) cells, CHO DUX-B 11 cells, mouse myeloma SP2/0, human HT-1080, NSO myeloma cell line, mouse L cells, mouse A9 cells, baby hamster kidney cells, C127 cells, COS cells and PC8 cells.
  • In another embodiment, the recombinantly expressed protein can be any of a number of protein binders, such as an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
  • In another embodiment, the recombinantly expressed protein binder can be humanized.
  • In one embodiment, the addition of the metal ion to the chemically defined cell culture medium reduces NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein. For example, the addition of the metal ion to the chemically defined cell culture medium can reduce NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein by at least 15%.
  • In another embodiment, the addition of the metal ion to the chemically defined cell culture medium increases galactosylaton of the expressed recombinant glycoprotein.
  • In yet another embodiment, the addition of the metal ion to the chemically defined cell culture medium increases NA1F and NA2F glycosylation of the expressed recombinant protein. For example, the addition of the metal ion to the chemically defined cell culture medium can increase NA1F glycosylation of the expressed recombinant protein by at least about 4 fold. In another example, the addition of the metal ion to the chemically defined cell culture medium can increase NA2F glycosylation of the expressed recombinant protein by at least about 7 fold.
  • In another embodiment, the present invention is directed to a cell culture medium for modulating the glycosylation profile of a recombinantly-expressed protein, comprising a protein-free chemically defined culture medium supplemented with an effective amount of a metal salt effective at modulating the glycosylation profile of a recombinantly-expressed protein, wherein the metal salt is not ferric citrate.
  • The metal salt can be an iron salt such as ferric nitrate. In one embodiment, the effective amount of the metal salt is about 0.2 mM to about 1 mM.
  • In another embodiment, the recombinantly-expressed protein is a protein binder, such as an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
  • The previously described embodiments have many advantages, including the ability to modulate the glycosylation of recombinantly expressed proteins by supplementing the culture media with millimolar amounts of a metal ion.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The skilled artisan will understand that the drawings, described below, are for illustration purposes only. The figures are not intended to limit the scope of the teachings in any way.
  • FIG. 1 depicts a simplified linear reaction view of the N-glycan biosynthetic pathway in mammalian cells.
  • FIG. 2 shows an exemplary depiction of the cell culture performance of Antibody 1 in ferric nitrate supplemented media.
  • FIG. 3 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in ferric nitrate supplemented media.
  • FIG. 4 shows an exemplary depiction of the cell culture performance of Antibody 1 in various iron supplemented media.
  • FIG. 5 shows an exemplary depiction of N-glycan oligosaccharide results of Antibody 1 in various iron supplemented media.
  • FIG. 6 shows an exemplary depiction of the cell culture performance of Antibody 2 in various iron supplemented media.
  • FIG. 7 shows an exemplary depiction of the titer and N-glycan oligosaccharide results of Antibody 2 in various iron supplemented media.
  • FIG. 8 shows an exemplary depiction of the cell culture performance of DVD-Ig in various iron supplemented media.
  • FIG. 9 shows an exemplary depiction of the titer and N-glycan oligosaccharide results of DVD-Ig in various iron supplemented media.
  • FIG. 10 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 for various iron supplemented feed media.
  • FIG. 11 shows an exemplary depiction of the N-glycan oligosaccharide results in bioreactors for Antibody 1 in various iron supplemented media.
  • FIG. 12 shows an exemplary depiction of the N-glycan oligosaccharide results in bioreactors for DVD-Ig 2 in various iron supplemented media.
  • FIG. 13 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various metal ion supplemented media.
  • FIG. 14 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various iron, transferrin, and galactose supplemented media.
  • FIG. 15 shows an exemplary depiction of the N-glycan oligosaccharide results of Antibody 1 in various iron, manganese, and galactose supplemented media.
  • DETAILED DESCRIPTION
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. The following definitions are provided to help interpret the disclosure and the claims of this application.
  • As used herein, the term “chemically defined medium” or “chemically defined media” refers to a synthetic growth medium in which the identity and concentration of all the components are known. Chemically defined mediums do not contain bacterial, yeast, animal, or plant extracts, animal serum or plasma although they may or may not include individual plant or animal-derived components (e.g., proteins, polypeptides, etc). Chemically defined media may contain inorganic salts such as phosphates, sulfates, and the like needed to support growth. The carbon source is defined, and is usually a sugar such as glucose, lactose, galactose, and the like, or other compounds such as glycerol, lactate, acetate, and the like. While certain chemically defined culture media also use phosphate salts as a buffer, other buffers may be employed such as citrate, triethanolamine, and the like. As used herein, a chemically defined cell culture medium contains no more than micromolar amounts of any metal ion. Examples of commercially available chemically defined mediums include, but are not limited to, various Dulbecco's Modified Eagle's (DME) mediums (Sigma-Aldrich Co; SAFC Biosciences, Inc), Ham's Nutrient Mixture (Sigma-Aldrich Co; SAFC Biosciences, Inc), combinations thereof, and the like. Methods of preparing chemically defined culture mediums are known in the art, for example in U.S. Pat. Nos. 6,171,825 and 6,936,441, WO 2007/077217, and U.S. Patent Application Publication Nos. 2008/0009040 and 2007/0212770. In certain embodiments, the chemically defined medium does not contain non-ionic detergents such as Pluronic® F-68.
  • As used herein, the term “serum-free culture medium” or “serum-free culture media” refers to a culture medium that is not supplemented with an animal serum. Although oftentimes serum-free mediums are chemically defined mediums, serum-free mediums may be supplemented with discrete animal or plant proteins or protein fractions. Methods of preparing serum-free culture medium are known in the art, for example in U.S. Pat. Nos. 6,171,825; 6,936,441 and EP 1849862.
  • As used herein, the term “an effective amount” means a sufficient amount of a metal ion to modulate the glycosylation profile of a recombinantly-expressed protein.
  • A metal ion may be any positively charged metal ion selected from, but not limited to, the following Al+3, Cr+2, Cr+3, Cd+1, Cd+2, Co+2, Co+3, Ca+2, Mg+2, Ni+2, Ti+2, Ti+3, Ti+4, V+2, V+3, V+5, Sr+2, Fe+2, Fe+3, Au+2, Au+3, Au+1, Ag+2, Ag+1, Pd+2, Pd+4, Pt+2, Pt+4, Cu+1, Cu+2, Mn+2, Mn+3, Mn+4, Zn+2, Se+4+2 and mixtures thereof. The ions are generally added to the medium in the form of salts such as chlorides and sulphates. In certain embodiments, the metal ion may not include ferric citrate.
  • A mammalian cell can be a mammalian cell line including, but not limited to, CHO cells, human embryonic kidney cells (HEK), baby hamster kidney (BHK), mouse myeloma (NSO), and human retinal cells (PERC.6).
  • The term “antibody” as referred to herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chains thereof. An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • The term “antigen-binding portion” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., TNFα, IL-12). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 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 CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; (v) a dAb fragment (Ward et al, (1989) Nature 341:544-546), which consists of a VH or VL domain; and (vi) an isolated complementarity determining region (CDR). 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 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. In one embodiment of the present disclosure, the antibody fragment is selected from the group consisting of an Fab, an Fd, an Fd', a single chain Fv (scFv), an scFva, and a domain antibody (dAb).
  • Still further, an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. These other proteins or peptides can have functionalities that allow for the purification of antibodies or antigen-binding portions thereof or allow for their association with each other or other molecules. Thus, examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric single chain variable fragment (scFv) molecules (Kipriyanov et al. (1995) Human Antibodies and Hybridomas 6:93-101) and the use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov et al. (1994) Mol. Immunol. 31:1047-1058). Antibody portions, such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as papain or pepsin digestion, respectively, of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques.
  • Two antibody domains are “complementary” where they belong to families of structures which form cognate pairs or groups or are derived from such families and retain this feature. For example, a VH domain and a VL domain of an antibody are complementary; two VH domains are not complementary, and two VL domains are not complementary. Complementary domains may be found in other members of the immunoglobulin superfamily, such as the Va and VP (or gamma and delta) domains of the T-cell receptor.
  • The term “domain” refers to a folded protein structure which retains its tertiary structure independently of the rest of the protein. Generally, domains are responsible for discrete functional properties of proteins, and in many cases may be added, removed or transferred to other proteins without loss of function of the remainder of the protein and/or of the domain. By single antibody variable domain is meant a folded polypeptide domain comprising sequences characteristic of antibody variable domains. It therefore includes complete antibody variable domains and modified variable domains, for example, in which one or more loops have been replaced by sequences which are not characteristic of antibody variable domains, or antibody variable domains which have been truncated or comprise N- or C-terminal extensions, as well as folded fragments of variable domains which retain at least in part the binding activity and specificity of the full-length domain.
  • Variable domains of the present disclosure may be combined to form a group of domains; for example, complementary domains may be combined, such as VL domains being combined with VH domains. Non-complementary domains may also be combined. Domains may be combined in a number of ways, involving linkage of the domains by covalent or non-covalent means.
  • A “dAb” or “domain antibody” refers to a single antibody variable domain (VH or VL) polypeptide that specifically binds antigen.
  • As used herein, the term “antigen binding region” or “antigen binding site” refers to the portion(s) of an antibody molecule, or antigen binding portion thereof, which contains the amino acid residues that interact with an antigen and confers on the antibody its specificity and affinity for the antigen.
  • The term “epitope” is meant to refer to that portion of any molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen binding regions. In the context of the present disclosure, first and second “epitopes” are understood to be epitopes which are not the same and are not bound by a single monospecific antibody, or antigen-binding portion thereof.
  • The term “DVD-Ig” or “dual-variable-domain-immunoglobulin” refers to a protein containing two or more antigen binding sites and is a tetravalent or multivalent binding protein. The DVD-Ig comprises two heavy chain DVD polypeptides and two light chain DVD polypeptides; each half of a DVD-Ig comprises a heavy chain DVD polypeptide, and a light chain DVD polypeptide, and two antigen binding sites. Each binding site comprises a heavy chain variable domain and a light chain variable domain with a total of six CDRs involved in antigen binding per antigen binding site. The description and generation of DVD-Ig molecules are detailed in the U.S. Pat. No. 7,612,181 B2.
  • The phrase “recombinant antibody” refers to antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et al. (1992) Nucl. Acids Res. 20:6287-6295) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of particular immunoglobulin gene sequences (such as human immunoglobulin gene sequences) to other DNA sequences. Examples of recombinant antibodies include chimeric, CDR-grafted and humanized antibodies.
  • The term “human antibody” refers to antibodies having variable and constant regions corresponding to, or derived from, human germline immunoglobulin sequences as described by, for example, Kabat et al. (See Kabat, et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). The human antibodies of the present disclosure, however, may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • Recombinant human antibodies of the present disclosure have variable regions, and may also include constant regions, derived from human germline immunoglobulin sequences (See Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242). In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. In certain embodiments, however, such recombinant antibodies are the result of selective mutagenesis or backmutation or both.
  • The term “backmutation” refers to a process in which some or all of the somatically mutated amino acids of a human antibody are replaced with the corresponding germline residues from a homologous germline antibody sequence. The heavy and light chain sequences of a human antibody of the present disclosure are aligned separately with the germline sequences in the VBASE database to identify the sequences with the highest homology. Differences in the human antibody of the present disclosure are returned to the germline sequence by mutating defined nucleotide positions encoding such different amino acid. The role of each amino acid thus identified as candidate for backmutation should be investigated for a direct or indirect role in antigen binding and any amino acid found after mutation to affect any desirable characteristic of the human antibody should not be included in the final human antibody. To minimize the number of amino acids subject to backmutation those amino acid positions found to be different from the closest germline sequence but identical to the corresponding amino acid in a second germline sequence can remain, provided that the second germline sequence is identical and colinear to the sequence of the human antibody of the present disclosure for at least 10, up to about 12 amino acids, on both sides of the amino acid in question. Backmuation may occur at any stage of antibody optimization.
  • The term “chimeric antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species and constant region sequences from another species, such as antibodies having murine heavy and light chain variable regions linked to human constant regions.
  • The term “CDR-grafted antibody” refers to antibodies which comprise heavy and light chain variable region sequences from one species but in which the sequences of one or more of the CDR regions of VH and/or VL are replaced with CDR sequences of another species, such as antibodies having murine heavy and light chain variable regions in which one or more of the murine CDRs (e.g., CDR3) has been replaced with human CDR sequences.
  • The term “humanized antibody” refers to antibodies which comprise heavy and light chain variable region sequences from a non-human species (e.g., a mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more “human-like”, i.e., more similar to human germline variable sequences. One type of humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced into non-human VH and VL sequences to replace the corresponding nonhuman CDR sequences. The oligosaccharide structure attached to the peptide chain is known as a “glycan” molecule. The glycan structures found in naturally occurring glycopeptides are typically divided into two classes, “N-linked glycans” or N-linked oligosaccharides” and “0-linked glycans” or O-linked oligosaccharides”.
  • Peptides comprising “0-linked glycans” have a saccharide attached to the hydroxy oxygen of serine, threonine, tyrosine, hydroxylysine, and or hydroxyproline residue in the primary protein.
  • Peptides expressed in eukaryotic cells typically comprise N-glycans. “N-glycans” are N-glycosylated at an amide nitrogen of an asparagine or an arginine residue in a protein via an N-acetylglucosamine residue. These “N-linked glycosylation sites” occur in the peptide primary structure containing, for example, the amino acid sequence asparagine-X-serine/threonine, where X is any amino acid residue except proline and aspartic acid.
  • All N-linked oligosaccharides have a common “pentasaccharide core” of Man3GlcNAc2. (“Man” refers to mannose; “Glc” refers to glucose; “NAc” refers to N-acetyl; and “GlcNAc” refers to N-acetylglucosamine). The pentasaccharide core is also referred to as the “trimannose core” or the “paucimannose core.” (see FIG. 1)
  • N-glycans differ with respect to the presence of, and/or in the number of branches (also called “antennae”) comprising peripheral sugars such as N-acetylglucosamine, galactose, N-acetylgalactosamine, N-acetylneuraminic acid, fucose and sialic acid that are added to the Man3GlcNAc2 core structure. Optionally, this structure may also contain a core fucose molecule and/or a xylose molecule. For a review of standard glycobiology nomenclature see, Essentials of Glycobiology Varki et al. eds., 1999, CSHL Press, the contents of which are incorporated herein by reference. N-glycans are classified according to their branched constituents (e.g., oligomannose-type, complex, or hybrid). An “oligomannose-type” or “high mannose-type” N-glycan has five or more mannose residues.
  • A “complex-type” N-glycan typically has at least one GlcNAc attached to the 1,3 mannose arm and at least one GlcNAc attached to the 1,6 mannose arm of a pentasaccharide core. Complex-type N-glycans may also have galactose (“Gal”) or N-acetylgalactosamine residues that are optionally modified with sialic acid or derivatives, e.g., N-acetyl neuraminic acid. Complex-type N-glycans may also have intrachain substitutions comprising “bisecting” GlcNAc, and core fucose (“Fuc”). Complex N-glycans may also have multiple antennae on the pentasaccharide core and are, therefore, also referred to as “multiple antennary-type glycans.”
  • A “hybrid-type” N-glycan comprises at least one GlcNAc on the terminal of the 1,3 mannose arm of the pentasaccharide core and zero or more mannoses on the 1,6 mannose arm of the trimannose core.
  • NGA2F +NGA2F-GlcNAc refers to the agalactosyl fucosylated biantennary oligosaccharides.
  • NGA2F has the structure:
  • Figure US20170145465A1-20170525-C00001
  • NA1F and NA2F refer to the fraction of galactose-containing fucosylated biantennary oligosaccharides.
  • NGA2F glycan is found on many mammalian glycoproteins including human IgG and is a substructure of bi-antennary N-linked oligosaccharides such as A2F, A1F, and NA2F.
  • The term “glycoform” denotes a type of polypeptide with a specific type and distribution of polysaccharides attached to, i.e. two polypeptides would be of the same glycoform if they comprise glycans with the same number, kind, and sequence of monosaccharides, i.e. have the same “glycosylation profile.” Glycoproteins often consist of a number of different glycoforms.
  • The “glycosylation profile” refers to the properties of the glycans of a glycosylated polypeptide. These properties include, but are not limited to, the glycosylation sites, or the glycosylation site occupancy, or the identity, structure, composition or quantity of the glycan attached to a recombinantly expressed polypeptide.
  • EXAMPLES
  • The following examples describe experiments relating to methods disclosed herein.
  • Example 1 Shake Flask Culture of CHO cell Lines Expressing Humanized Monoclonal Antibodies in the Presence of Different Amounts of Ferric Nitrate
  • To test the effect of metal ions of protein glycosylation, recombinant Chinese Hamster Ovary (CHO) cell lines (CHO DUX-B 11 origin based on a DHFR (dihydrofolate reductase) expression system) expressing two different humanized monoclonal antibodies (Antibody 1, Antibody 2) or a dual variable domain immunoglobulin (DVD-Ig) were shake flask cultured in chemically defined basal medium (CDBM) and chemically defined feed medium (CDFM) supplemented with selected iron compounds. The cell lines were serially expanded through separate seed train inoculums to generate enough cells for inoculation. All supplemented cultures were performed in triplicate unless otherwise indicated. Supplements and concentrations utilized during the cultures were slightly different between the 2 different cell lines as shown in Table 1.
  • TABLE 1
    Summary of cell culture process conditions & iron supplementation details
    Antibody 1 Antibody 2 DVD-Ig
    Culture Vessel
    500 mL 500 mL 500 mL 500 mL 500 mL 500 mL 500 mL 500 mL
    shake flask shake flask shake flask shake flask shake flask shake flask shake flask shake flask
    Culture Mode Fedbatch Fedbatch Fedbatch Fedbatch Fedbatch Fedbatch Fedbatch Fedbatch
    Initial Culture 36 36 36 36 36 36 36 36
    Temperature (° C.)
    Iron Supplements Ferric Ferric Ferrous Ferric Ferric Ferric Ferric Sodium
    Evaluated Nitrate Citrate Sulfate Nitrate Citrate Nitrate Citrate Citrate
    Supplement 0.1, 0.5, 1, 0.1, 0.5, 1 1 0.1 0.1, 1 0.1, 1 0.1, 1 0.5
    Concentrations 2, 5
    (mM)a
    aHigher concentrations were evaluated with Antibody 1, but resulted in significantly detrimental cell culture performance: Ferric Nitrate (10 mM), Ferrous Sulfate (3, 5, 10 mM).
  • The cell line producing Antibody 1 (also called ABBV-696) was cultured in shake flasks in fedbatch mode after an abbreviated seed train in two independent experiments. Ferric nitrate was supplemented into chemically-defined basal and feed media at concentrations of 0.1, 1.0, and 5.0 mM (Experiment 1) and 0.1, 0.5, 1.0, and 2.0 mM (Experiment 2), and compared to an unsupplemented control condition.
  • Viable cell density (VCD) and cell viability values were measured through trypan blue exclusion via ViCell automated cell counters (Beckman Coulter, Fullerton, Calif.), glucose and lactate values were measured with a ABL-805 (Radiometer Medical, Denmark) blood gas analyzer.
  • Antibody titers were measured from crude cell culture harvests on a Poros A™ (Life Technologies, Carlsbad, Calif.) Protein A affinity column using an Agilent (Santa Clara, Calif.) 1200 Series HPLC, or equivalent, operating with a low pH, step elution gradient with detection at 280 nm. Absolute concentrations were assigned with respect to reference standard calibration curves. Purified antibodies subjected to additional analytical characterization were purified using MabSelect™ Protein A (GE Healthcare, Piscataway, N.J.) using a low pH, step elution gradient, followed by buffer exchange using Corning Life Sciences (Tewksbury, Mass.) Spin Concentrator X UF columns according to the manufacturers' recommended procedures.
  • The viable cell density, viability, and harvest titer results are shown in FIG. 2. At these particular concentrations, ferric nitrate generally facilitated an increase in VCD, with the exception of the highest concentration (5.0 mM) supporting the lowest peak VCD. Viability results were comparable to the control, i.e. unsupplemented conditions, with the exception of the 5.0 mM condition, which facilitated a drop in cell viability that was larger, and earlier in the culture, compared to control conditions.
  • Harvest titer results demonstrated that below 2.0 mM ferric nitrate did not adversely impact titer, but a concentration of ferric nitrate of 2.0 and 5.0 mM lowered the harvest titer significantly. Collectively, these results suggest that there are concentrations of ferric nitrate which minimize the drop in viable cell density, while simultaneously ensuring a comparable cell viability and harvest titer profile.
  • The glycosylation profile of the recombinantly expressed proteins was then determined using standard procedures. Approximately 200 μg of Protein A purified samples from Cell Lines 1 and 2 were then treated with N-glycanase at 37° C. overnight to remove the N-glycans from the protein. The protein was precipitated and the supernatant was taken for subsequent chemical derivatization of the reducing end of the released glycans with 2-aminobenzamide (2-AB) dye. Following the derivatization step, the excess labeled was removed using clean up cartridges and the samples were analyzed using normal phase HPLC with fluorescent detection. Mobile phase A was 100% acetonitrile and mobile phase B was 50 mM ammonium formate pH 4.4. The glycan were eluted from a polyamide column (Prozyme, Hayward, Calif.) using a shallow gradient. The labeled glycans were detected using a fluorescence detector with an excitation wavelength of 330 nm and an emission wavelength of 420 nm.
  • The N-glycan oligosaccharide results were quite different between the variously supplemented cultures and the unsupplemented control as shown in FIG. 3. Data in each experiment for each oligosaccharide was normalized to the level observed in the 1.0 mM condition for that particular experiment. The addition of iron shifted the distribution toward increased galactosylation, reducing the total NGA2F (NGA2F and NGA2F-GlcNAc) species thereby increasing the total NA1F (NA1F plus NA1F-GlcNAc) and NA2F species. A concentration-dependent response was observed below 1.0 mM ferric nitrate. The impact of iron on galactosylation was still effective but seemed to reach a plateau at 2.0 mM ferric nitrate. The total NGA2F species was reduced by as much as 25% with 7-fold increases in total NA1F and 50-fold increases in NA2F species. The supplementation of ferric nitrate into the cell culture media is capable of significantly influencing the overall protein galactosylation pathway, and for some commercial protein therapeutics this is the desired product quality distribution.
  • Example 2 Shake Flask Cell Culture in the Presence of Additional Iron Supplements
  • Additional shake flask experiments were performed with Cell Line 1 to evaluate the impact of other iron compounds, namely ferric citrate and ferrous sulfate. Concentrations of the respective iron compounds were chosen so as to minimize any potential adverse impacts on cell growth and productivity, but still facilitate a measurable impact on the glycoform profile. Both an unsupplemented control and cultures supplemented with ferric nitrate were included in this study. Cell culture process performance indicators were monitored and measured throughout the respective cultures. Viable cell density, cell viability, harvest titer, and harvest N-glycan oligosaccharide data was measured and reported.
  • FIG. 4 highlights the cell culture results observed through the use of iron supplementation into the basal medium. All iron supplemented cultures showed a higher viable cell density compared to the unsupplemented control. The results indicate a significant increase in peak VCD from 9×106 cells/mL for the control versus 12-14×106 cells/mL for the iron supplemented cultures. At the end of the culture period, the final titer ratio was similar to the control suggesting a slight drop in overall antibody productivity upon inclusion of these particular concentrations of iron, consistent with the results observed from the previous shake flask cultures.
  • After Protein A purification, the N-glycan glycoform profile was measured with the results highlighted in FIG. 5. Similar to the initial ferric nitrate shake flask results, there was a significant increase in overall galactosylation. The total NGA2F species was reduced 15-30% with the least impact from ferrous sulfate and the most significant impact from the ferric nitrate or ferric citrate at higher concentrations. Corresponding increases in the total NA1F and NA2F species were observed as well. Ferrous sulfate supplementation had modest 3-fold increases in both total NA1F and NA2F species. Ferric nitrate supplementation provided 5-fold increases in total NA1F with 7- to 9-fold increases in NA2F. Ferric citrate supplementation provided very similar results to ferric nitrate with a concentration dependence observed below 1.0 mM as previously observed with ferric nitrate. The absolute value of the fold increase in NA2F varied between experiments and relies heavily on quantification of the very small amount of this species present in the control culture. Nonetheless, significant increases were observed in NA2F with the addition of iron to the basal medium.
  • Example 3 Evaluation of Alternative Cell Lines for the Impact of Iron Supplementation on the Resulting Protein Glycosylation Profiles
  • The cell line expressing Antibody 2 (also called ABT-806) was evaluated in shake flask culture in fedbatch mode. Ferric citrate or ferric nitrate was supplemented into the basal medium at various concentrations to evaluate the resulting impact on cell culture performance, as well as the protein glycosylation profile. The unsupplemented control was performed in duplicate. The viable cell density, viability, and harvest titer results are shown in FIGS. 6 and 7. Ferric citrate supplementation at either 0.1 or 1.0 mM improved overall cell growth with higher peak viable cell density (increased by 4×106 cells/mL) while ferric nitrate at 0.1 mM had very little impact when compared to the unsupplemented control. Contrary to Antibody 1, all iron-supplemented conditions had a positive impact to the titer for Antibody 2.
  • Similar to Antibody 1, there was a very significant impact on the protein glycosylation profiles of harvest samples across all iron concentrations tested (FIG. 7) and the modulation of the protein glycosylation was concentration-dependent. The total NGA2F species declined by approximately 20%, similar to Antibody 1. The total NA1F species increased 4- to 6-fold compared to the unsupplemented control while the NA2F species increased 7- to 10-fold. Hence, as with Antibody 1, there was a very significant impact on the N-glycan glycoform profile, and the individual N-glycans which increased or decreased upon iron supplementation was consistent between both cell lines.
  • To confirm that the observed effect on protein glycosylation was not unique to recombinantly expressed monoclonal antibodies, the impact of iron supplementation was evaluated with a cell line expressing a DVD-Ig molecule (denoted DVD 1, also called ABBV-257). The DVD-Ig cell line was cultured in shake flasks with ferric nitrate, ferric citrate, or sodium citrate supplementation at various concentrations. Sodium citrate was included to provide additional evidence that the impact to the oligosaccharide profile with the prior experiments was a result of the iron addition. All conditions were performed in triplicate. FIG. 8 shows the cell culture viable cell density and viability performance for one representative from each experimental condition. The average harvest titer for the triplicates and the relative oligosaccharide profile results are in FIG. 9.
  • Iron supplementation had minimal impact to the cell culture growth performance and harvest titer. However, the impact to the galactosylated species is similar to that observed for both Antibody 1 and 2 with iron supplementation. Increased concentration of both ferric nitrate and ferric citrate supplemented to the culture provided increased galactosylation as observed previously with both antibody-expressing cell lines. The total NGA2F species declined by approximately 20%, while the total NA1F species increased 3- to 9-fold, and the NA2F species increased 50- to 100-fold compared to the unsupplemented control. Sodium citrate had no effect on the oligosaccharide profile compared to the unsupplemented control.
  • Example 4 Evaluation of Impact of Iron Supplementation in Feed Medium
  • Prior examples included iron supplementation via the culture basal medium, thus introducing iron to the cell culture throughout the duration of the shake flask experiment. By introducing iron to the culture via the feed medium, exposure to iron would be more gradual over the length of the culture. The impact to Antibody 1 was evaluated in triplicate shake flasks as described previously. Similar to prior results with Antibody 1, cell growth was enhanced with no improvement to titer. FIG. 10 shows the impact to the relative oligosaccharide profile and that regardless of where the iron is introduced to the cell culture (basal versus feed medium), the galactosylation of the antibody increases as observed previously.
  • Example 5 Evaluation of Impact of Iron Supplementation in Bioreactors
  • Prior experiments included iron supplementation in shake flask cultures. Experiments with the cell line expressing Antibody 1 and with a cell line expressing a different DVD-Ig (denoted DVD 2, also called ABT-122) than discussed previously were carried out to confirm the impact from iron supplements would be observed in bioreactors. Operating conditions were similar to shake flask experiments, but now included pH control at pH 6.90 and dissolved oxygen control at a setpoint of 40% air saturation. Antibody 1 bioreactors were run in triplicate with the exception of the 0.08 mM FC condition. DVD 2 bioreactors were run in duplicate. In FIG. 11, two independent experiments with the cell line expressing Antibody 1 show the dose-dependence effect of ferric citrate on the increase in total NA1F and NA2F species in bioreactors. FIG. 12 shows the similar effect with the cell line expressing a DVD-Ig molecule, however the impact to this DVD-Ig molecule is limited to the total NA1F species as the NA2F species is not observed when this cell line is cultured in chemically defined medium. The same cell line expressing this DVD-Ig molecule grown in a complex cell culture medium (containing plant-derived hydrolysates) contains a significantly different oligosaccharide profile and iron supplementation has little impact on altering this profile.
  • Example 6 Evaluation of Impact of Alternative Metal Ions on Oligosaccharide Profile
  • Given the observations presented here with both ferric and ferrous forms of iron, and studies performed previously with manganese, additional metal ions listed in the table below were explored for their impact on the glycosylation pathway. All evaluations were performed in triplicate in shake flasks with the cell line expressing Antibody 1.
  • Metal Salt Concentration Evaluated
    Cupric Chloride 0.01, 0.2 mM
    Cupric Sulfate 0.01, 0.2 mM
    Zinc Chloride 0.02, 0.5 mM
    Zinc Sulfate 0.05, 0.5 mM
    Chromium Chloride 0.001, 0.01 mM 
  • As seen in FIG. 13, these additional metal ions were unsuccessful at increasing the galactosylation for the oligosaccharide profile for Antibody 1. In fact, copper appeared to have a slightly inhibitory effect on the galactosylation. This figure only depicts the lower concentrations evaluated, but similar results were observed at the higher concentrations listed in the table above.
  • Example 7 Evaluation of Iron Transport Limitation
  • In all experiments with all cell lines evaluated, the galactosylation reaches a plateau between 0.5 and 1.0 mM iron supplemented. Despite the increase in iron with minimal impact to culture performance, the total NA1F and NA2F levels do not increase. One potential explanation is the inability for iron to effectively enter the cell. Another possible explanation is the lack of galactose to incorporate into the oligosaccharide chain. The next set of experiments explored if iron transport into the cell or the galactose availability were the limiting factors. Recombinant transferrin (rTf) was added to triplicate shake flask cultures with the cell line expressing Antibody 1 with varying concentrations of ferric citrate. An independent experiment was performed with the same cell line to also determine if galactose supplementation in addition to iron and transferrin would enhance the galactosylation with triplicate shake flasks for each condition. As seen in FIG. 14, no impact was observed to the galactosylation with the addition of transferrin and without galactose, the typical profile for the oligosaccharide species is similar to results shown previously without transferrin. However, when galactose is supplemented with iron, increased galactosylation is observed overall, even though the plateau between 0.5 and 1.0 mM ferric citrate continues to persist.
  • Example 8 Evaluation of Iron Synergy with Manganese and Galactose on the Oligosaccharide Profile
  • Manganese has been reported in several studies to increase the galactosylation of recombinant proteins with synergistic effects observed when combined with galactose supplementation. Shake flask experiments were conducted in triplicate with the cell line expressing Antibody 1 to determine if the plateau observed between 0.5 and 1.0 mM ferric citrate could be overcome by including these additional factors known to improve galactosylation. Ferric citrate was supplemented to the cultures via the CDBM as described previously. Manganese and galactose were supplemented into both the CDBM and CDFM at the concentrations listed in FIG. 15. There is no synergistic effect with ferric citrate and manganese observed. Manganese in combination with galactose is effective at altering the galactosylation for Antibody 1, however ferric citrate with galactose is more effective, and a dose-dependence is observed with galactose supplementation.
  • Any patent, patent application, publication, or other disclosure material identified in the specification is hereby incorporated by reference herein in its entirety. Any material, or portion thereof, that is said to be incorporated by reference herein, but which conflicts with existing definitions, statements, or other disclosure material set forth herein is only incorporated to the extent that no conflict arises between that incorporated material and the present disclosure material.
  • Abbreviations
    • DVD-Ig Dual variable domain immunoglobulin
    • EDTA Ethylenediaminetetraacetic acid
    • FC Ferric citrate
    • FN Ferric nitrate
    • FS Ferrous sulfate
    • Fuc Fucose
    • Gal Galactose
    • GlcNAc N-acetylglucosamine
    • Man Mannose
    • PK Pharmacokinetics
    • SC Sodium citrate
    • VCD Viable cell density

Claims (25)

1. A method for modulating the glycosylation profile of a recombinant protein expressed in a eukaryotic cell, comprising:
culturing a eukaryotic cell in a protein-free, chemically defined culture medium supplemented with an effective amount of a metal salt; and
expressing a recombinant protein in the eukaryotic cell,
wherein the amount of the metal salt is effective at modulating the glycosylation profile of the recombinant protein.
2. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the metal salt is an iron salt.
3. The method for modulating the glycosylation profile of a recombinant protein according to claim 2, wherein the iron salt is selected from the group consisting of ferric nitrate, ferric citrate and ferrous sulfate.
4. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the effective amount of the metal salt is about 0.1 mM to about 1 mM.
5. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the chemically defined culture medium contains less than about 1 μM of metal ions.
6. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the eukaryotic cell is a mammalian cell.
7. The method for modulating the glycosylation profile of a recombinant protein according to claim 6, wherein the mammalian cell is selected from the group consisting of Chinese hamster ovary (CHO) cells, CHO DUX-B 11 cells, mouse myeloma SP2/0, human HT-1080, NSO myeloma cell line, mouse L cells, mouse A9 cells, baby hamster kidney cells, C127 cells, COS cells and PC8 cells.
8. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the recombinant protein is an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
9. The method for modulating the glycosylation profile of a recombinant protein according to claim 8, wherein the recombinant antibody is humanized.
10. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium reduces NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein.
11. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium reduces NGA2F and NGA2F-GlcNAc glycosylation of the expressed recombinant protein by at least 15%.
12. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium increases galactosylaton of the expressed recombinant glycoprotein.
13. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium increases NA1F and NA2F glycosylation of the expressed recombinant protein.
14. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium increases NA1F glycosylation of the expressed recombinant protein by at least about 4 fold.
15. The method for modulating the glycosylation profile of a recombinant protein according to claim 1, wherein the addition of the metal ion to the chemically defined cell culture medium increases NA2F glycosylation of the expressed recombinant protein by at least about 7 fold.
16. A cell culture medium for modulating the glycosylation profile of a recombinantly-expressed protein, comprising:
a protein-free chemically defined culture medium supplemented with an effective amount of a metal salt effective at modulating the glycosylation profile of a recombinantly-expressed protein,
wherein the chemically defined culture medium does not contain ferric citrate.
17. The cell culture medium according to claim 16, wherein the metal salt is an iron salt.
18. The cell culture medium according to claim 17, wherein the iron salt is ferric nitrate.
19. The cell culture medium according to claim 16, wherein the effective amount of the metal salt is about 0.1 mM to about 1 mM.
20. The cell culture medium according to claim 16, wherein the recombinantly-expressed protein is an antibody, a fragment or variant thereof, or a fusion protein comprising an antibody, a fragment or variant thereof.
21. An antibody or dual-variable-domain-immunoglobulin (DVD-Ig) produced by the process for modulating a glycosylation profile of claim 1.
22. The antibody or dual-variable-domain-immunoglobulin (DVD-Ig) of claim 21, wherein the addition of the metal ion to the chemically defined cell culture medium reduces NGA2F and NGA2F-GlcNAc glycosylation of the expressed antibody or dual-variable-domain-immunoglobulin (DVD-Ig).
23. The antibody or dual-variable-domain-immunoglobulin (DVD-Ig) of claim 21, wherein the addition of the metal ion to the chemically defined cell culture medium increases NA1F and NA2F glycosylation of the expressed antibody or dual-variable-domain-immunoglobulin (DVD-Ig).
24. The antibody or dual-variable-domain-immunoglobulin (DVD-Ig) of claim 21, wherein the addition of the metal ion to the chemically defined cell culture medium increases galactosylaton of the expressed antibody or dual-variable-domain-immunoglobulin (DVD-Ig).
25. An antibody or DVD-Ig molecule having a glycoprofile as disclosed herein.
US15/425,003 2013-10-04 2017-02-06 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins Abandoned US20170145465A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/425,003 US20170145465A1 (en) 2013-10-04 2017-02-06 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361886855P 2013-10-04 2013-10-04
US14/506,372 US9598667B2 (en) 2013-10-04 2014-10-03 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US15/425,003 US20170145465A1 (en) 2013-10-04 2017-02-06 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/506,372 Continuation US9598667B2 (en) 2013-10-04 2014-10-03 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins

Publications (1)

Publication Number Publication Date
US20170145465A1 true US20170145465A1 (en) 2017-05-25

Family

ID=51844844

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/506,372 Active 2035-06-13 US9598667B2 (en) 2013-10-04 2014-10-03 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US15/425,003 Abandoned US20170145465A1 (en) 2013-10-04 2017-02-06 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/506,372 Active 2035-06-13 US9598667B2 (en) 2013-10-04 2014-10-03 Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins

Country Status (3)

Country Link
US (2) US9598667B2 (en)
EP (1) EP3052640A2 (en)
WO (1) WO2015051293A2 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
EP2830651A4 (en) 2013-03-12 2015-09-02 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
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
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
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
WO2015128314A1 (en) 2014-02-27 2015-09-03 F. Hoffmann-La Roche Ag Modulation of cell growth and glycosylation in recombinant glycoprotein production

Family Cites Families (521)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE30985E (en) 1978-01-01 1982-06-29 Serum-free cell culture media
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4634665A (en) 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US5179017A (en) 1980-02-25 1993-01-12 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
AU560087B2 (en) 1981-09-08 1987-03-26 Rockefeller University, The Lipoprotein lipase suppression by endotoxin-induced mediator (shock assay)
US4510245A (en) 1982-11-18 1985-04-09 Chiron Corporation Adenovirus promoter system
US4560655A (en) 1982-12-16 1985-12-24 Immunex Corporation Serum-free cell culture medium and process for making same
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
US4704366A (en) 1984-06-22 1987-11-03 Bio-Rad Laboratories, Inc. Process for binding IgG to protein A
US5672347A (en) 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
DE3431140A1 (en) 1984-08-24 1986-03-06 Behringwerke Ag, 3550 Marburg ENHANCER FOR EUKARYOTIC EXPRESSION SYSTEMS
IL73883A (en) 1984-12-20 1990-12-23 Yeda Res & Dev Monoclonal antibodies against tnf-alpha,hybridomas producing them and method for the purification of tnf-alpha
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5672502A (en) 1985-06-28 1997-09-30 Celltech Therapeutics Limited Animal cell culture
GB8516415D0 (en) 1985-06-28 1985-07-31 Celltech Ltd Culture of animal cells
EP0212489B1 (en) 1985-08-16 1994-11-30 The Rockefeller University Anabolic activity modulator and uses thereof
IT206190Z2 (en) 1985-12-17 1987-07-13 Iveco Fiat IMPROVING THE BODYWORK OF A HEAVY VEHICLE
US4968615A (en) 1985-12-18 1990-11-06 Ciba-Geigy Corporation Deoxyribonucleic acid segment from a virus
US4925796A (en) 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US5681718A (en) 1986-03-14 1997-10-28 Celltech Limited Methods for enhanced production of tissue plasminogen activator in cell culture using alkanoic acids or salts thereof
US4927762A (en) 1986-04-01 1990-05-22 Cell Enterprises, Inc. Cell culture medium with antioxidant
DE3631229A1 (en) 1986-09-13 1988-03-24 Basf Ag MONOCLONAL ANTIBODIES AGAINST HUMAN TUMORNESCROSE FACTOR (TNF) AND THEIR USE
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
US4877608A (en) 1987-11-09 1989-10-31 Rorer Pharmaceutical Corporation Pharmaceutical plasma protein formulations in low ionic strength media
US6238891B1 (en) 1987-11-18 2001-05-29 Cetus Oncology Corporation Method of increasing product expression through solute stress
US5118796A (en) 1987-12-09 1992-06-02 Centocor, Incorporated Efficient large-scale purification of immunoglobulins and derivatives
JP2638652B2 (en) 1988-07-18 1997-08-06 カイロン・コーポレーション Monoclonal antibody reacting with cachectin
DE68925971T2 (en) 1988-09-23 1996-09-05 Cetus Oncology Corp CELL GROWING MEDIUM FOR INCREASED CELL GROWTH, FOR INCREASING LONGEVITY AND EXPRESSION OF PRODUCTS
US6048728A (en) 1988-09-23 2000-04-11 Chiron Corporation Cell culture medium for enhanced cell growth, culture longevity, and product expression
US5126250A (en) 1988-09-28 1992-06-30 Eli Lilly And Company Method for the reduction of heterogeneity of monoclonal antibodies
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
DE68914244T2 (en) 1988-10-24 1994-10-27 Otsuka Pharma Co Ltd Monoclonal antibody.
KR0184860B1 (en) 1988-11-11 1999-04-01 메디칼 리써어치 카운실 Single domain ligands receptors comprising said ligands methods for their production and use of said ligands
DE68927671T2 (en) 1988-12-19 1997-05-07 American Cyanamid Co Products to treat endotoxin shock in a mammal
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE4009603A1 (en) 1989-03-30 1990-10-04 Leybold Ag Lock chamber for substrate
US4933435A (en) 1989-04-05 1990-06-12 Bioprobe International Antibody purification process
US5169936A (en) 1989-04-14 1992-12-08 Biogen, Inc. Protein purification on immobilized metal affinity resins effected by elution using a weak ligand
US6448380B2 (en) 1989-08-07 2002-09-10 Peptech Limited Tumor necrosis factor antibodies
EP0486526B2 (en) 1989-08-07 2001-03-07 Peptech Limited Tumour necrosis factor binding ligands
US5959087A (en) 1989-08-07 1999-09-28 Peptide Technology, Ltd. Tumour necrosis factor binding ligands
GB8921123D0 (en) 1989-09-19 1989-11-08 Millar Ann B Treatment of ards
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5945098A (en) 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
DE4037604A1 (en) 1990-04-25 1991-10-31 Bayer Ag Use of anti-TNF antibodies to treat ischaemia and its sequelae - esp. to increase survival rate after myocardial infarct and transplants
US5378612A (en) 1990-05-11 1995-01-03 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Culture medium for production of recombinant protein
US5110913A (en) 1990-05-25 1992-05-05 Miles Inc. Antibody purification method
US5096816A (en) 1990-06-05 1992-03-17 Cetus Corporation In vitro management of ammonia's effect on glycosylation of cell products through pH control
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022547D0 (en) 1990-10-17 1990-11-28 Wellcome Found Purified immunoglobulin
GB9022543D0 (en) 1990-10-17 1990-11-28 Wellcome Found Antibody production
GB2279077B (en) 1990-12-21 1995-06-14 Celltech Ltd Therapeutic compositions comprising recombinant antibodies specific for the TNFalpha
GB9109645D0 (en) 1991-05-03 1991-06-26 Celltech Ltd Recombinant antibodies
US5994510A (en) 1990-12-21 1999-11-30 Celltech Therapeutics Limited Recombinant antibodies specific for TNFα
GB9028123D0 (en) 1990-12-28 1991-02-13 Erba Carlo Spa Monoclonal antibodies against human tumor necrosis factor alpha
SG47099A1 (en) 1991-03-15 1998-03-20 Amgen Boulder Inc Pegylation of polypeptides
US5698195A (en) 1991-03-18 1997-12-16 New York University Medical Center Methods of treating rheumatoid arthritis using chimeric anti-TNF antibodies
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
US20070298040A1 (en) 1991-03-18 2007-12-27 Centocor, Inc. Methods of treating seronegative arthropathy with anti-TNF antibodies
DK0610201T4 (en) 1991-03-18 2008-02-04 Centocor Inc Monoclonal and chimeric antibodies specific for human tumor necrosis factor
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
US20040120952A1 (en) 2000-08-07 2004-06-24 Centocor, Inc Anti-TNF antibodies and peptides of human tumor necrosis factor
US20060246073A1 (en) 1991-03-18 2006-11-02 Knight David M Anti-TNF antibodies and peptides of human tumor necrosis factor
US7192584B2 (en) 1991-03-18 2007-03-20 Centocor, Inc. Methods of treating psoriasis with anti-TNF antibodies
DK0604418T3 (en) 1991-03-29 1999-06-14 Immunex Corp Isolated viral protein cytokine antagonists
US5328985A (en) 1991-07-12 1994-07-12 The Regents Of The University Of California Recombinant streptavidin-protein chimeras useful for conjugation of molecules in the immune system
ATE243747T1 (en) 1991-07-15 2003-07-15 Wellcome Found PRODUCTION OF ANTIBODIES
DK0605522T3 (en) 1991-09-23 2000-01-17 Medical Res Council Process for producing humanized antibodies
GB9122820D0 (en) 1991-10-28 1991-12-11 Wellcome Found Stabilised antibodies
AU3244693A (en) 1991-12-17 1993-07-19 Schering Corporation Use of the combination of anti-tumor necrosis factor plus interleukin-6 to treat septic shock
US5605923A (en) 1992-04-02 1997-02-25 Smithkline Beecham Corporation Compounds useful for treating inflammatory diseases and inhibiting production of tumor necrosis factor
ATE381614T1 (en) 1992-07-24 2008-01-15 Amgen Fremont Inc FORMATION OF XENOGENE ANTIBODIES
ATE172880T1 (en) 1992-08-28 1998-11-15 Bayer Ag USE OF MONOCLONAL ANTI-TNF ANTIBODIES FOR THE TREATMENT OF BACTERIAL MENINGITIS
WO1994008619A1 (en) 1992-10-08 1994-04-28 The Kennedy Institute Of Rheumatology Treatment of autoimmune and inflammatory disorders
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
AU682156B2 (en) 1992-10-15 1997-09-25 Dana-Farber Cancer Institute, Inc. Treatment of insulin resistance in obesity linked type II diabetes using antagonists to TNF-alpha function
JPH06292592A (en) 1993-02-09 1994-10-21 Snow Brand Milk Prod Co Ltd Production of glycoprotein
DK0614984T4 (en) 1993-03-05 2010-12-20 Bayer Healthcare Llc Human monoclonal anti-TNF alpha antibodies
DE4307508A1 (en) 1993-03-10 1994-09-15 Knoll Ag Use of anti-TNF antibodies as a medicine in the treatment of heart failure (heart muscle weakness)
CA2161351C (en) 1993-04-26 2010-12-21 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
JPH08510642A (en) 1993-05-12 1996-11-12 ゾマ コーポレイション Immunotoxin consisting of gelonin and antibody
GB2294267B (en) 1993-06-03 1996-11-20 Therapeutic Antibodies Inc Anti-TNFalpha Fab fragments derived from polyclonal IgG antibodies
AU8083594A (en) 1993-10-19 1995-05-08 Scripps Research Institute, The Synthetic human neutralizing monoclonal antibodies to human immunodeficiency virus
EP0659766A1 (en) 1993-11-23 1995-06-28 Schering-Plough Human monoclonal antibodies against human cytokines and methods of making and using such antibodies
NZ278607A (en) 1994-02-07 1999-05-28 Knoll Ag Use of tnf antagonists for treating disorders involving elevated serum levels of il-6 wherein the serum levels are 500pg/ml or above
EP0666312A1 (en) 1994-02-08 1995-08-09 Wolfgang A. Renner Process for the improvement of mammalian cell growth
US5429746A (en) 1994-02-22 1995-07-04 Smith Kline Beecham Corporation Antibody purification
WO1995023813A1 (en) 1994-03-04 1995-09-08 Merck & Co., Inc. In vitro antibody affinity maturation using alanine scanning mutagenesis
US5856179A (en) 1994-03-10 1999-01-05 Genentech, Inc. Polypeptide production in animal cell culture
US5945311A (en) 1994-06-03 1999-08-31 GSF--Forschungszentrumfur Umweltund Gesundheit Method for producing heterologous bi-specific antibodies
NZ288997A (en) 1994-06-24 1999-01-28 Immunex Corp Controlled release pharmaceutical formulation comprising polypeptide encapsulated in alginate
ZA955642B (en) 1994-07-07 1997-05-06 Ortho Pharma Corp Lyophilized imaging agent formulation
US5561053A (en) 1994-08-05 1996-10-01 Genentech, Inc. Method for selecting high-expressing host cells
SE503424C2 (en) 1994-11-14 1996-06-10 Pharmacia Ab Process for purification of recombinant coagulation factor VIII
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
ES2304786T3 (en) 1995-04-27 2008-10-16 Amgen Fremont Inc. ANTI-IL-8 HUMAN ANTIBODIES, DERIVED FROM IMMUNIZED XENORATONES.
EP0823941A4 (en) 1995-04-28 2001-09-19 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
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
US6656466B1 (en) 1995-06-06 2003-12-02 Genetech, Inc. Human tumor necrosis factor—immunoglobulin(TNFR1-IgG1) chimera composition
US6113898A (en) 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
JPH11510170A (en) 1995-07-27 1999-09-07 ジェネンテック インコーポレーテッド Protein Formula
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
WO1997014719A1 (en) 1995-10-16 1997-04-24 Unilever N.V. A bifunctional or bivalent antibody fragment analogue
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
AR005035A1 (en) 1995-12-11 1999-04-07 Merck Patent Ges Mit Beschränkter Haftung PROCEDURE TO PREPARE RECOMBINANT PROTEINS IN E. COLI, BY FERMENTATION WITH GREAT CONCENTRATION OF CELLS.
RU2270030C2 (en) 1996-02-09 2006-02-20 Абботт Байотекнолоджи эЛтиди. METHOD AND OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT FOR INHIBITING HUMAN TNFα ACTIVITY, APPLYING THE OBTAINED HUMAN ANTIBODY OR ITS ANTIGEN-BINDING FRAGMENT AS INGREDIENT FOR PRODUCING MEDICAMENT
PT802257E (en) 1996-04-19 2003-01-31 Nestle Sa IMMORTALIZED LINE OF HUMAN COLON EPITHELIAL CELLS
TW491855B (en) 1996-08-07 2002-06-21 Csl Ltd Purification of immunoglobulins
EP1482031B1 (en) * 1996-08-30 2015-10-28 Life Technologies Corporation Serum-free mammalian cell culture medium, and uses thereof
US20040171152A1 (en) 1996-10-10 2004-09-02 Invitrogen Corporation Animal cell culture media comprising non-animal or plant-derived nutrients
EP1864999B1 (en) 1996-11-27 2009-03-18 Genentech, Inc. Affinity purification of polypeptide on protein a matrix
DK1500329T3 (en) 1996-12-03 2012-07-09 Amgen Fremont Inc Human antibodies that specifically bind TNF-alpha
GB9625175D0 (en) 1996-12-04 1997-01-22 Medi Cult As Serum-free cell culture media
US5804420A (en) 1997-04-18 1998-09-08 Bayer Corporation Preparation of recombinant Factor VIII in a protein free medium
NZ337828A (en) 1997-04-28 2001-06-29 Lilly Co Eli Activated protein C formulations having a bulking agent and a salt
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
EP0999853B1 (en) 1997-06-13 2003-01-02 Genentech, Inc. Stabilized antibody formulation
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6475725B1 (en) 1997-06-20 2002-11-05 Baxter Aktiengesellschaft Recombinant cell clones having increased stability and methods of making and using the same
US20040191256A1 (en) 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
DE69830315T2 (en) 1997-06-24 2006-02-02 Genentech Inc., San Francisco GALACTOSYLATED GLYCOPROTEIN CONTAINING COMPOSITIONS AND METHOD FOR THE PRODUCTION THEREOF
US20020045207A1 (en) 1997-10-31 2002-04-18 Lynne A. Krummen Glycoprotein production process
ATE419009T1 (en) 1997-10-31 2009-01-15 Genentech Inc METHODS AND COMPOSITIONS CONSISTING OF GLYCOPROTEIN GLYCOFORMS
US20040136986A1 (en) 1997-10-31 2004-07-15 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
ES2252876T5 (en) 1997-12-03 2014-04-03 Roche Diagnostics Gmbh Process to prepare polypeptides with adequate glycolization
WO1999032605A1 (en) 1997-12-19 1999-07-01 Novo Nordisk A/S Method for producing heterologous proteins in eukaryotic cells on an industrial scale using nucleotide-manipulating agents
AU3657899A (en) 1998-04-20 1999-11-08 James E. Bailey Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO1999057246A1 (en) 1998-05-01 1999-11-11 Life Technologies, Inc. Animal cell culture media comprising non-animal or plant-derived nutrients
ATE321066T1 (en) 1998-05-06 2006-04-15 Genentech Inc ANTI-HER2 ANTIBODY COMPOSITION
EP1308455B9 (en) 1998-05-06 2006-06-14 Genentech, Inc. A composition comprising anti-HER2 antibodies
DE19821933C1 (en) 1998-05-15 1999-11-11 Disetronic Licensing Ag Device for administering an injectable product
DE19822031C2 (en) 1998-05-15 2000-03-23 Disetronic Licensing Ag Auto injection device
US6528286B1 (en) 1998-05-29 2003-03-04 Genentech, Inc. Mammalian cell culture process for producing glycoproteins
US6406909B1 (en) 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
US7883704B2 (en) 1999-03-25 2011-02-08 Abbott Gmbh & Co. Kg Methods for inhibiting the activity of the P40 subunit of human IL-12
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
AU3633000A (en) 1999-03-26 2000-10-16 Human Genome Sciences, Inc. Neutrokine-alpha binding proteins and methods based thereon
EP2275540B1 (en) 1999-04-09 2016-03-23 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
EP1171615B1 (en) 1999-04-26 2006-12-13 Genentech, Inc. Cell culture process for glycoproteins
WO2000066160A1 (en) 1999-04-28 2000-11-09 Yamanouchi Pharmaceutical Co., Ltd. Parenteral medicinal composition containing humanized monoclonal antibody fragment and method for stabilizing the same
AT409379B (en) 1999-06-02 2002-07-25 Baxter Ag MEDIUM FOR PROTEIN- AND SERUM-FREE CELL CULTURE
AU778022B2 (en) 1999-07-30 2004-11-11 Genentech Inc. Charged filtration membranes and uses therefor
JP2003510087A (en) 1999-09-27 2003-03-18 ジェネンテック・インコーポレーテッド Method for producing recombinant protein using apoptosis inhibitor
AR026743A1 (en) 1999-12-09 2003-02-26 Pharmacia Ab PRODUCTION OF PEPTIDES
KR20010056451A (en) 1999-12-15 2001-07-04 윤재승 Arginine-enriched medium used for mass-producing recombinant protein in animal cell culture
AU2228901A (en) 1999-12-28 2001-07-09 Chugai Seiyaku Kabushiki Kaisha Stable antibody compositions and injection preparations
CA2399266A1 (en) 2000-02-08 2001-08-16 Genentech, Inc. Improved galactosylation of recombinant glycoproteins
TWI373343B (en) 2000-02-10 2012-10-01 Abbott Gmbh & Co Kg Antibodies that bind human interleukin-18 and methods of making and using
GB0003231D0 (en) 2000-02-11 2000-04-05 Medi Cult As Cell culture media
JP2001218840A (en) 2000-02-14 2001-08-14 Kanegafuchi Chem Ind Co Ltd Adsorbent for transforming growth factor. beta, adsorbing/removing method, and adsorber
AU2001246846A1 (en) 2000-04-06 2001-10-23 Chugai Seiyaku Kabushiki Kaisha Immunoassay of anti-hm1.24 antibody
EP1276849A4 (en) 2000-04-12 2004-06-09 Human Genome Sciences Inc Albumin fusion proteins
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
KR100787073B1 (en) 2000-06-28 2007-12-21 글리코파이, 인크. Methods for producing modified glycoproteins
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US20030170813A1 (en) 2000-07-05 2003-09-11 Kenichi Suga Process for producing glycoprotein
JP5490972B2 (en) 2000-08-04 2014-05-14 中外製薬株式会社 Protein injection formulation
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
US20060018907A1 (en) 2000-08-07 2006-01-26 Centocor, Inc. Anti-TNF antibodies and peptides of human tumor necrosis factor
US20050249735A1 (en) 2000-08-07 2005-11-10 Centocor, Inc. Methods of treating ankylosing spondylitis using anti-TNF antibodies and peptides of human tumor necrosis factor
US6635448B2 (en) 2000-08-21 2003-10-21 Clonexdevelopment, Inc. Methods and compositions for increasing protein yield from a cell culture
BR0114374A (en) 2000-10-02 2003-12-30 Novo Nordisk As Preparation comprising a plurality of factor vii polypeptides, methods for determining the pattern of factor vii glycoform and factor vii-related polypeptides, and for producing said preparation, pharmaceutical formulation, methods for treating a factor responsive syndrome. vii, for the prevention of unwanted bleeding, for the prevention of unwanted blood clotting, and for the prevention of tissue factor mediated reactions, and the use of the preparation.
US20090151023A1 (en) 2000-11-13 2009-06-11 Viktor Kuvshinov Transformation system for Camelina sativa
WO2002066603A2 (en) 2001-02-15 2002-08-29 Centocor, Inc. Chemically defined medium for cultured mammalian cells
US20030096414A1 (en) 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
EP1373547A4 (en) 2001-03-27 2006-01-18 Smithkline Beecham Corp Control of glycoforms in igg
CA2446734A1 (en) 2001-05-24 2002-11-28 Human Genome Sciences, Inc. Antibodies against tumor necrosis factor delta (april)
US20030012786A1 (en) 2001-05-25 2003-01-16 Teoh Leah S. Use of anti-TNF antibodies as drugs in treating septic disorders of anemic patients
CA2868614A1 (en) 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
AU2002316230A1 (en) 2001-06-13 2002-12-23 Genentech, Inc. Methods of culturing animal cells and polypeptide production in animal cells
PL222211B1 (en) 2001-06-26 2016-07-29 Amgen Fremont Inc Antibodies to opgl
NZ592087A (en) 2001-08-03 2012-11-30 Roche Glycart Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
GB0119520D0 (en) 2001-08-10 2001-10-03 Owen Mumford Ltd Improvements relating to injection devices
CA2460206C (en) 2001-10-02 2012-01-24 Novo Nordisk Health Care Ag Method for production of recombinant proteins in eukaryote cells
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
ES2326964T3 (en) 2001-10-25 2009-10-22 Genentech, Inc. GLICOPROTEIN COMPOSITIONS.
AU2002340787B2 (en) 2001-11-12 2009-09-17 Monash University Peptide purification by means of metal ion affinity chromatography
AU2002356749A1 (en) 2001-11-28 2003-06-10 Hermann Katinger Process for the production of polypeptides in mammalian cell cultures
PL368749A1 (en) 2001-11-28 2005-04-04 Sandoz Ag Cell culture process
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
JP4723185B2 (en) 2001-11-28 2011-07-13 サンド・アクチエンゲゼルシヤフト Method for producing recombinant polypeptide
WO2003059935A2 (en) 2001-12-21 2003-07-24 Immunex Corporation Methods for purifying protein
AU2003210802B2 (en) 2002-02-05 2009-09-10 Genentech Inc. Protein purification
US20030161828A1 (en) 2002-02-19 2003-08-28 Abbott Gmbh & Co. Kg Use of TNF antagonists as drugs for the treatment of patients with an inflammatory reaction and without suffering from total organ failure
CA2417689C (en) 2002-03-05 2006-05-09 F. Hoffmann-La Roche Ag Improved methods for growing mammalian cells in vitro
JP2005521401A (en) 2002-03-27 2005-07-21 イミュネックス・コーポレーション Methods for increasing polypeptide production
US20030190710A1 (en) 2002-03-28 2003-10-09 Devries Ruth L. Control of glycoforms in IgG
CA2481837A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
CA2478925C (en) 2002-04-26 2016-06-07 Robert Lee Fahrner Non-affinity purification of proteins
US20040009172A1 (en) 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20040029229A1 (en) 2002-05-20 2004-02-12 Reeves Philip J. High level protein expression system
MXPA05000552A (en) 2002-07-15 2005-04-28 Immunex Corp Methods and media for controlling sialylation of proteins produced by mammalian cells.
AU2003267999B2 (en) 2002-07-19 2010-03-11 Abbvie Biotechnology Ltd Treatment of TNFalpha related disorders
US20090280065A1 (en) 2006-04-10 2009-11-12 Willian Mary K Uses and Compositions for Treatment of Psoriasis
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US7067279B1 (en) 2002-08-23 2006-06-27 Immunex Corporation Cell culture performance with betaine
US6924124B1 (en) 2002-08-23 2005-08-02 Immunex Corporation Feeding strategies for cell culture
US6974681B1 (en) 2002-08-23 2005-12-13 Immunex Corporation Cell culture performance with vanadate
WO2004026891A2 (en) 2002-09-18 2004-04-01 Genencor International, Inc. Protein purification
US6890736B1 (en) 2002-09-20 2005-05-10 Immunex Corporation Methods for producing proteins in cultured cells
MY150740A (en) 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
US7674885B2 (en) 2002-11-01 2010-03-09 Bayer Healthcare Llc Process for concentration of macromolecules
US20040086532A1 (en) 2002-11-05 2004-05-06 Allergan, Inc., Botulinum toxin formulations for oral administration
US20040162414A1 (en) 2002-11-22 2004-08-19 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
US20040101939A1 (en) 2002-11-22 2004-05-27 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
AU2003293543A1 (en) 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
CA2508925C (en) 2002-12-23 2012-08-28 Bristol-Myers Squibb Company Product quality enhancement in mammalian cell culture processes for protein production
CA2511520A1 (en) 2002-12-23 2004-07-15 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
EP1601697B1 (en) 2003-02-28 2007-05-30 Lonza Biologics plc Antibody purification by Protein A and ion exchange chromatography
DK1620549T3 (en) 2003-05-01 2007-11-05 Dsm Ip Assets Bv Process for the preparation of biological substances by perfusion culture of suspended animal cells
ATE472597T2 (en) 2003-05-15 2010-07-15 Wyeth Llc CONTROLLED GLUCOSE SUPPLY FOR ANIMAL CELL CULTURE
GB2404665B (en) 2003-08-08 2005-07-06 Cambridge Antibody Tech Cell culture
WO2005035748A1 (en) 2003-10-10 2005-04-21 Novo Nordisk Health Care Ag Method for large-scale production of a polypeptide in eukaryote cells and a culture vessel suitable therefor
FR2861080B1 (en) 2003-10-20 2006-02-17 Lab Francais Du Fractionnement ANTIBODIES HAVING AN OPTIMIZED FUCOSE AND GALACTOSE RATE
ES2342539T3 (en) 2003-10-24 2010-07-08 Amgen, Inc. PROCESS FOR THE PURIFICATION OF PROTEINS IN A FLOW-DIRECT FRACTION OF HYDROPHOBIC INTERACTION CHROMATOGRAPHY.
US20050100965A1 (en) 2003-11-12 2005-05-12 Tariq Ghayur IL-18 binding proteins
DE10355251A1 (en) 2003-11-26 2005-06-23 Merck Patent Gmbh Water-based pharmaceutical preparation for treatment of tumors has active ingredient effective against receptor of endothelial growth factor receptor
WO2005056600A2 (en) 2003-12-08 2005-06-23 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind or neutralize dengue virus
AU2004309114A1 (en) 2003-12-23 2005-07-14 Laboratoires Serono Sa Process for the production of tumor necrosis factor-binding proteins
JP4943161B2 (en) 2003-12-23 2012-05-30 ジェネンテック, インコーポレイテッド Treatment of cancer with a novel anti-IL13 monoclonal antibody
US20080058507A1 (en) 2004-02-11 2008-03-06 Hui Liu Method For The Removal Of Aggregate Proteins From Recombinant Samples Using Ion Exchange Chromatography
RU2390353C2 (en) 2004-02-12 2010-05-27 Мерк Патент Гмбх High-concentration liquid anti-egfr antibody compositions
CA2552639C (en) 2004-02-27 2012-05-01 Ge Healthcare Bio-Sciences Ab A process for the purification of antibodies
CN1234725C (en) 2004-04-07 2006-01-04 陈志南 High performance quick purifying method for preparing piecewise antibody
TWI439284B (en) 2004-04-09 2014-06-01 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating tnfα-related disorders
US20060127950A1 (en) 2004-04-15 2006-06-15 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
WO2005111627A2 (en) 2004-04-15 2005-11-24 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
AU2005233387B2 (en) 2004-04-15 2011-05-26 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
ATE455861T1 (en) 2004-05-04 2010-02-15 Novo Nordisk Healthcare Ag O-LINKED GLYCOFORMS OF FACTOR VII AND METHOD FOR THE PRODUCTION THEREOF
EP1771477A2 (en) 2004-07-21 2007-04-11 Glycofi, Inc. Immunoglobulins comprising predominantly a gal2glcnac2man3glcnac2 glycoform
US20060223147A1 (en) 2004-08-05 2006-10-05 Kyowa Hakko Kogyo Co., Ltd., Process for producing glycoprotein composition
TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
DK1807101T3 (en) 2004-09-30 2016-05-23 Bayer Healthcare Llc Devices and methods for integrated continuous production of biological molecules
WO2006041970A2 (en) 2004-10-08 2006-04-20 Abbott Biotechnology Ltd. Treatment of respiratory syncytial virus (rsv) infection
WO2007011390A2 (en) 2004-10-09 2007-01-25 Government Of The United States As Represented By The Secretary Of The Army Large-scale production of human serum butyrylcholinesterase as a bioscavenger
JP4831436B2 (en) 2004-10-21 2011-12-07 ジーイー・ヘルスケア・バイオサイエンス・アクチボラグ Chromatographic ligand
US20060094104A1 (en) 2004-10-29 2006-05-04 Leopold Grillberger Animal protein-free media for cultivation of cells
JP2008529495A (en) 2005-02-04 2008-08-07 グラクソ グループ リミテッド Optimization of heterologous polypeptide expression
WO2006084826A1 (en) 2005-02-11 2006-08-17 Novo Nordisk Health Care Ag Production of a polypeptide in a serum-free cell culture liquid containing plant protein hydrolysate
MX2007011129A (en) 2005-03-11 2007-11-06 Wyeth Corp A method of weak partitioning chromatography.
WO2006107990A2 (en) 2005-04-05 2006-10-12 The Johns Hopkins University Improving protein n-glycosylation of eukaryotic cells using dolichol-linked oligosaccharide synthesis pathway, other n-gylosylation-increasing methods, and engineered hosts expressing products with increased n-glycosylation
MX2007012499A (en) 2005-04-11 2007-12-06 Medarex Inc Protein purification using hcic amd ion exchange chromatography.
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US20090203055A1 (en) 2005-04-18 2009-08-13 Massachusetts Institute Of Technology Compositions and methods for RNA interference with sialidase expression and uses thereof
US8349991B2 (en) 2005-04-19 2013-01-08 Massachusetts Institute Of Technology Amphiphilic polymers and methods of use thereof
NZ591701A (en) 2005-05-16 2012-11-30 Abbott Biotech Ltd Use of tnf inhibitor for treatment of erosive polyarthritis
AU2006254217A1 (en) 2005-06-03 2006-12-07 Biovitrum Ab (Publ) Process for cultivating animal cells comprising the feeding of plant-derived peptones
WO2006133148A2 (en) 2005-06-03 2006-12-14 Genentech, Inc. Method of producing antibodies with modified fucosylation level
CN101506238B (en) 2005-06-30 2013-11-06 森托科尔公司 Methods and compositions with enhanced therapeutic activity
KR101419729B1 (en) 2005-07-26 2014-07-17 상가모 바이오사이언스 인코포레이티드 Targeted integration and expression of exogenous nucleic acid sequences
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
WO2007024743A2 (en) 2005-08-19 2007-03-01 Centocor, Inc. Proteolysis resistant antibody preparations
LT1917276T (en) 2005-08-26 2018-05-25 Ares Trading S.A. Process for the preparation of glycosylated interferon beta
JP2009507040A (en) 2005-09-02 2009-02-19 グライコフィ, インコーポレイテッド Immunoglobulin containing mainly GLCNACMAN3GLCNAC2 glycoform
PE20070796A1 (en) 2005-10-24 2007-08-15 Wyeth Corp PROTEIN PRODUCTION METHOD USING ANTI-SENESCENCE COMPOUNDS
EP1948235B1 (en) 2005-11-01 2013-08-28 AbbVie Biotechnology Ltd Methods for determining efficacy of adalimumab in subjects having ankylosing spondylitis using ctx-ii and mmp3 as biomarkers
AU2006312148B2 (en) 2005-11-07 2012-04-12 The Rockefeller University Reagents, methods and systems for selecting a cytotoxic antibody or variant thereof
US8470318B2 (en) 2005-11-07 2013-06-25 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
US20080206246A1 (en) 2006-04-05 2008-08-28 Ravetch Jeffrey V Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
ES2564790T5 (en) 2005-12-08 2023-08-25 Amgen Inc Enhanced glycoprotein production using manganese
RU2486236C2 (en) 2006-01-04 2013-06-27 Бакстер Интернэшнл Инк. Method of protein expression
US20070190057A1 (en) 2006-01-23 2007-08-16 Jian Wu Methods for modulating mannose content of recombinant proteins
US20070202051A1 (en) 2006-02-10 2007-08-30 Pari Gmbh Aerosols for sinunasal drug delivery
NZ611859A (en) 2006-04-05 2014-12-24 Abbvie Biotechnology Ltd Antibody purification
EP2815768A3 (en) 2006-04-05 2015-01-14 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
US20080118496A1 (en) 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
WO2007120656A2 (en) 2006-04-10 2007-10-25 Abbott Biotechnology Ltd. Uses and compositions for treatment of rheumatoid arthritis
US20090317399A1 (en) 2006-04-10 2009-12-24 Pollack Paul F Uses and compositions for treatment of CROHN'S disease
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
EP2012586A4 (en) 2006-04-10 2010-08-18 Abbott Biotech Ltd Uses and compositions for treatment of ankylosing spondylitis
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
US20080131374A1 (en) 2006-04-19 2008-06-05 Medich John R Uses and compositions for treatment of rheumatoid arthritis
EP2032600A2 (en) 2006-05-19 2009-03-11 Glycofi, Inc. Erythropoietin compositions
US20100021451A1 (en) 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
US20080311043A1 (en) 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
WO2009017491A1 (en) 2006-06-14 2009-02-05 Smithkline Beecham Corporation Methods for purifying antibodies using ceramic hydroxyapatite
TWI498137B (en) 2006-06-30 2015-09-01 Abbvie Biotechnology Ltd Automatic injection device
DK2041270T3 (en) 2006-07-13 2014-01-27 Wyeth Llc Preparation of glycoproteins
US9766217B2 (en) 2006-09-08 2017-09-19 Novo Nordisk A/S Methods of optimizing chromatographic separation of polypeptides
ES2620261T3 (en) 2006-09-10 2017-06-28 Glycotope Gmbh Use of human myeloid leukemic cells for antibody expression
AU2007294731B2 (en) 2006-09-13 2014-04-17 Abbvie Inc. Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
EP2500416A1 (en) * 2006-09-13 2012-09-19 Abbott Laboratories Cell culture improvements
US10982250B2 (en) 2006-09-18 2021-04-20 Genentech, Inc. Methods of protein production
EP2081553B1 (en) 2006-10-06 2020-08-12 Amgen Inc. Stable antibody formulations
BRPI0717335A2 (en) 2006-10-27 2013-12-10 Abbott Biotech Ltd CRYSTALINE ANTI-HTNFALFA ANTIBODIES
EP2091969A4 (en) 2006-10-27 2010-05-12 Univ Rockefeller Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
MX2009004519A (en) 2006-11-03 2009-05-12 Wyeth Corp Glycolysis-inhibiting substances in cell culture.
US20110105734A1 (en) 2006-12-06 2011-05-05 Jcr Pharmaceuticals Co., Ltd. Method for production of human erythropoietin
WO2008077545A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Selection method
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
CN105056231A (en) 2006-12-28 2015-11-18 詹森生物科技公司 Methods and vectors for generating asialylated immunoglobulins
US7691980B2 (en) 2007-01-09 2010-04-06 Bio-Rad Laboratories, Inc. Enhanced capacity and purification of antibodies by mixed mode chromatography in the presence of aqueous-soluble nonionic organic polymers
JP2010516651A (en) 2007-01-17 2010-05-20 メルク セローノ ソシエテ アノニム Methods for purification of Fc-containing proteins
KR20090127326A (en) 2007-03-02 2009-12-10 와이어쓰 Use of copper and glutamate in cell culture for production of polypeptides
NZ598881A (en) 2007-03-29 2013-11-29 Abbvie Inc Crystalline anti-human il-12 antibodies
JP5456658B2 (en) 2007-03-30 2014-04-02 メディミューン,エルエルシー Antibody preparation
WO2008120107A2 (en) 2007-04-03 2008-10-09 Oxyrane (Uk) Limited Glycosylation of molecules
EP2135090A1 (en) 2007-04-16 2009-12-23 Momenta Pharmaceuticals, Inc. Proteolytic release of glycans
EP2135088A1 (en) 2007-04-16 2009-12-23 Momenta Pharmaceuticals, Inc. Characterization of n-glycan mixtures by nuclear magnetic resonance
US20110213137A1 (en) 2007-04-16 2011-09-01 Momenta Pharmaceuticals, Inc. Isotopically-labeled glycans
CA2682738A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Defined glycoprotein products and related methods
AU2008240078B2 (en) 2007-04-16 2012-10-25 Momenta Pharmaceuticals, Inc. Multi-dimensional chromatographic methods for separating N-glycans
RU2475759C2 (en) 2007-04-16 2013-02-20 Момента Фармасьютикалз, Инк. Study of n-glycans with application of exoglycosidases
CA2682750A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Ms methods to evaluate glycans
US9182467B2 (en) 2007-04-16 2015-11-10 Momenta Pharmaceuticals, Inc. Comparative analysis of protein conformations by using 2D NOESY NMR spectra
US20120264927A1 (en) 2007-04-16 2012-10-18 Ian Christopher Parsons Methods for labeling glycans
WO2008128230A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Reference glycoprotein products and related methods
US20100279306A1 (en) 2007-04-16 2010-11-04 Momenta Pharmaceuticals, Inc. Analysis of phosphorylated glycans, glycopeptides or glycoproteins by imac
RU2475751C2 (en) 2007-04-16 2013-02-20 Момента Фармасьютикалз, Инк. Methods related to cell surface glycosylation
TW200902708A (en) 2007-04-23 2009-01-16 Wyeth Corp Methods of protein production using anti-senescence compounds
EP1988101A1 (en) 2007-05-04 2008-11-05 Novo Nordisk A/S Improvement of factor VIII polypeptide titers in cell cultures
WO2008135498A2 (en) 2007-05-04 2008-11-13 Novo Nordisk A/S Prevention of protein degradation in mammalian cell cultures
AU2008251405B2 (en) 2007-05-11 2012-05-17 Amgen Inc. Improved feed media
EP2679996A1 (en) 2007-05-31 2014-01-01 AbbVie Inc. Biomarkers predictive of the responsiveness to TNF-alfa inhibitors in autoimmune disorders
WO2008150490A2 (en) 2007-06-01 2008-12-11 Abbott Biotechnology Ltd. Uses and compositions for treatment of psoriasis and crohn's disease
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
US20100221823A1 (en) 2007-06-11 2010-09-02 Amgen Inc. Method for culturing mammalian cells to improve recombinant protein production
SI3597659T1 (en) 2007-07-09 2023-04-28 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
WO2009011782A2 (en) 2007-07-13 2009-01-22 Abbott Biotechnology Ltd. METHODS AND COMPOSITIONS FOR PULMONARY ADMINISTRATION OF A TNFa INHIBITOR
KR100897159B1 (en) 2007-07-26 2009-05-14 보령제약 주식회사 A plant recombinant human CTLA4Ig and a method for producing the same
US8753839B2 (en) 2007-08-08 2014-06-17 Abbvie Inc. Compositions and methods for crystallizing antibodies
WO2009023562A2 (en) 2007-08-09 2009-02-19 Wyeth Use of perfusion to enhance production of fed-batch cell culture in bioreactors
MX2010002269A (en) 2007-08-28 2010-03-25 Abbott Biotech Ltd Compositions and methods comprising binding proteins for adalimumab.
EP2031064A1 (en) 2007-08-29 2009-03-04 Boehringer Ingelheim Pharma GmbH & Co. KG Method for increasing protein titres
CN101784670A (en) 2007-08-31 2010-07-21 弗·哈夫曼-拉罗切有限公司 Glycosylation profile analysis
AU2008310583A1 (en) 2007-10-12 2009-04-16 Sigma-Aldrich Co. Llc Compositions and methods for improved glycoprotein sialylation
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2009058307A1 (en) 2007-10-29 2009-05-07 University Of Georgia Research Foundation, Inc. In vivo isotopic labeling method for quantitative glycomics
WO2009058769A1 (en) 2007-10-30 2009-05-07 Schering Corporation Purification of antibodies containing hydrophobic variants
CA2707483A1 (en) 2007-11-30 2009-06-11 Wolfgang Fraunhofer Protein formulations and methods of making same
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
US20130195888A1 (en) 2007-11-30 2013-08-01 Abbvie Ultrafiltration and diafiltration formulation methods for protein processing
ES2644089T3 (en) 2007-12-27 2017-11-27 Baxalta GmbH Cell culture procedures
WO2009083246A1 (en) 2007-12-31 2009-07-09 Bayer Schering Pharma Aktiengesellschaft Antibodies to tnf alpha
WO2009086550A1 (en) 2008-01-03 2009-07-09 Abbott Laboratories Predicting long-term efficacy of a compound in the treatment of psoriasis
CA2711307C (en) 2008-01-03 2020-10-27 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
EP2784089A1 (en) 2008-01-15 2014-10-01 AbbVie Inc. Improved mammalian expression vectors and uses thereof
MX2010007728A (en) 2008-01-15 2010-12-21 Abbott Gmbh & Co Kg Powdered protein compositions and methods of making same.
CA2713342A1 (en) 2008-01-30 2009-08-13 Abbott Laboratories Compositions and methods for crystallizing antibody fragments
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
EP2260054B1 (en) 2008-02-29 2015-06-03 Biogen Idec MA Inc. Purified immunoglobulin fusion proteins and methods of their purification
CA2717614A1 (en) 2008-03-11 2009-09-17 Genentech, Inc. Antibodies with enhanced adcc function
MX2010010503A (en) 2008-03-24 2010-11-09 Abbott Biotech Ltd Methods and compositions for treating bone loss.
JP2011517410A (en) 2008-04-08 2011-06-09 アミリス バイオテクノロジーズ, インコーポレイテッド Heterologous sequence expression
ES2458541T3 (en) 2008-05-02 2014-05-06 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
WO2009135656A1 (en) 2008-05-06 2009-11-12 Lonza Biologics Plc. A method for the purification of antibodies using displacement chromatography
CA2726087A1 (en) 2008-06-03 2009-12-10 Tariq Ghayur Dual variable domain immunoglobulins and uses thereof
US8318416B2 (en) 2008-08-08 2012-11-27 Biogen Idec Ma Inc. Nutrient monitoring and feedback control for increased bioproduct production
HUE047316T2 (en) 2008-08-14 2020-04-28 Genentech Inc Methods for removing a contaminant using indigenous protein displacement ion exchange membrane chromatography
US20100069617A1 (en) 2008-09-12 2010-03-18 Ge Healthcare Bio-Sciences Ab Enhanced protein aggregate removal by mixed mode chromatography on hydrophobic interaction media in the presence of protein-excluded zwitterions
KR20110084196A (en) 2008-09-26 2011-07-21 유레카 쎄라퓨틱스, 인코포레이티드 Cell lines and proteins with variant glycosylation pattern
WO2010043703A1 (en) 2008-10-17 2010-04-22 Dsm Ip Assets B.V. Removal of host cell proteins
RU2514657C2 (en) 2008-10-20 2014-04-27 Эббви Инк, Method of obtaining preparation of antibody against il-18 or its antigen-binding part (versions)
JP5856845B2 (en) 2008-10-20 2016-02-10 アッヴィ・インコーポレイテッド Virus inactivation during antibody purification
CA2932207A1 (en) 2008-10-20 2010-12-09 Abbvie Inc. Isolation and purification of antibodies using protein a affinity chromatography
GB0821100D0 (en) 2008-11-18 2008-12-24 Hansa Medical Ab Antibodies
WO2010071817A2 (en) 2008-12-19 2010-06-24 Momenta Pharmaceuticals, Inc. Characterization of o-linked glycans
CN102239173A (en) 2008-12-19 2011-11-09 动量制药公司 Methods related to modified glycans
US8614297B2 (en) 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
WO2010080062A1 (en) 2009-01-08 2010-07-15 Ge Healthcare Bio-Sciences Ab Separation method using single polymer phase systems
KR20110119702A (en) 2009-01-22 2011-11-02 모멘타 파머슈티컬스 인코포레이티드 Galactose-alpha-1,3-galactose-containing n-glycans in glycoprotein products derived from cho cells
WO2010098863A1 (en) 2009-02-26 2010-09-02 Lpath, Inc. Humanized platelet activating factor antibody design using anti-lipid antibody templates
US9127043B2 (en) 2009-03-05 2015-09-08 Biogen Ma Inc. Purification of immunoglobulins
EP2233499A1 (en) 2009-03-26 2010-09-29 CSL Behring AG Antibody composition with altered Fab sialylation
BRPI1013648B1 (en) 2009-04-01 2023-05-09 Nobelpharma Co., Ltd ANTIBODY OR ANTIGEN-BINDING FRAGMENT, PHARMACEUTICAL COMPOSITION AND METHOD FOR PRODUCTION OF ANTIBODY OR ANTIGEN-BINDING FRAGMENT
BRPI1015019A2 (en) 2009-04-20 2018-02-14 Pfizer glycosylation control of protein and related compositions and methods.
WO2010127069A1 (en) 2009-04-29 2010-11-04 Schering Corporation Antibody purification
JP5677411B2 (en) 2009-04-29 2015-02-25 アッヴィ バイオテクノロジー リミテッド Automatic injection equipment
RU2560701C2 (en) 2009-05-04 2015-08-20 Эббви Байотекнолоджи Лтд. Stable compositions with high concentrations of proteins of human antibodies against tnf-alpha
WO2010138502A2 (en) 2009-05-26 2010-12-02 Momenta Pharmaceuticals, Inc. Production of glycoproteins
KR20120042744A (en) 2009-05-28 2012-05-03 베링거 인겔하임 인터내셔날 게엠베하 Method for a rational cell culturing process
WO2010136209A1 (en) 2009-05-28 2010-12-02 ETH Zürich N-glycan core beta-galactosyltransferase and uses thereof
EP2438185A4 (en) 2009-06-05 2016-10-05 Momenta Pharmaceuticals Inc Methods of modulating fucosylation of glycoproteins
CN102482633A (en) 2009-07-06 2012-05-30 弗·哈夫曼-拉罗切有限公司 Method of culturing eukaryotic cells
CA2768325C (en) 2009-07-24 2019-10-29 F. Hoffmann-La Roche Ag Optimizing the production of antibodies
US8945895B2 (en) 2009-07-31 2015-02-03 Baxter International Inc. Methods of purifying recombinant ADAMTS13 and other proteins and compositions thereof
WO2011015926A1 (en) 2009-08-03 2011-02-10 Avesthagen Limited A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein
US20110039300A1 (en) 2009-08-10 2011-02-17 Robert Bayer Antibodies with enhanced adcc functions
HUE048980T2 (en) 2009-08-11 2020-08-28 Hoffmann La Roche Production of proteins in glutamine-free cell culture media
US20110053223A1 (en) 2009-08-14 2011-03-03 Robert Bayer Cell culture methods to make antibodies with enhanced adcc function
WO2011024025A1 (en) 2009-08-28 2011-03-03 Avesthagen Limited An erythropoietin analogue and a method thereof
US9540426B2 (en) 2009-10-06 2017-01-10 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
US8470552B2 (en) 2009-10-12 2013-06-25 Keck Graduate Institute Strategy to reduce lactic acid production and control PH in animal cell culture
EP2325296A1 (en) 2009-11-20 2011-05-25 LEK Pharmaceuticals d.d. Production of glycoproteins with low N-glycolylneuraminic acid (Neu5Gc) content
US9096879B2 (en) 2009-11-24 2015-08-04 Biogen Ma Inc. Method of supplementing culture media to prevent undesirable amino acid substitutions
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
EP2507627A2 (en) 2009-12-04 2012-10-10 Momenta Pharmaceuticals, Inc. Antennary fucosylation in glycoproteins from cho cells
PL3715356T3 (en) 2009-12-18 2024-03-11 Csl Limited Method of purifying polypeptides
US20120014956A1 (en) 2010-02-02 2012-01-19 Hartmut Kupper Methods and compositions for predicting responsiveness to treatment with tnf-alpha inhibitor
US20120309056A1 (en) 2010-02-04 2012-12-06 Leon Arnaud Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
KR20120118065A (en) 2010-02-12 2012-10-25 디에스엠 아이피 어셋츠 비.브이. Single unit antibody purification
KR101555740B1 (en) 2010-03-10 2015-09-25 에프. 호프만-라 로슈 아게 Method for purifying immunoglobulin solutions
US8981081B2 (en) 2010-03-12 2015-03-17 Purecircle Usa Inc. High-purity steviol glycosides
BR112012025645A2 (en) 2010-04-07 2017-12-12 Momenta Pharmaceuticals Inc high mannose glycans.
US20110262965A1 (en) * 2010-04-23 2011-10-27 Life Technologies Corporation Cell culture medium comprising small peptides
ES2617777T5 (en) 2010-04-23 2022-10-13 Hoffmann La Roche Production of heteromultimeric proteins
LT2563906T (en) 2010-04-26 2018-02-26 Novartis Ag Process for cultivation of cho cells
WO2011134920A1 (en) 2010-04-26 2011-11-03 Novartis Ag Improved cell culture medium
JP6050226B2 (en) 2010-05-28 2016-12-21 ジェネンテック, インコーポレイテッド Lowering lactic acid levels and increasing polypeptide production by downregulating LDH and PDHK expression
US8747854B2 (en) 2010-06-03 2014-06-10 Abbvie Biotechnology Ltd. Methods of treating moderate to severe hidradenitis suppurativa with anti-TNF-alpha antibodies
US9428546B2 (en) 2010-07-30 2016-08-30 Pfizer Inc. Tandem purification of proteins
CN103080300B (en) 2010-08-05 2015-11-25 安姆根有限公司 Increase the productive rate of cell culture and the dipeptides of vigor
DE102010038990A1 (en) 2010-08-06 2012-02-09 Robert Bosch Gmbh Shaft bearing device for a hand tool
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
WO2012037430A1 (en) 2010-09-17 2012-03-22 Abbott Laboratories Raman spectroscopy for bioprocess operations
US20120122076A1 (en) 2010-09-20 2012-05-17 Abbott Laboratories Purification of antibodies using simulated moving bed chromatography
US20120258496A1 (en) 2010-09-27 2012-10-11 Boehringer Ingelheim International Gmbh Production of low fucose antibodies in h4-ii-e rat cells
US20130244280A1 (en) 2010-10-08 2013-09-19 Cadila Healthcare Limited Expression vector for high level expression of recombinant proteins
KR20130142128A (en) 2010-10-11 2013-12-27 애브비 인코포레이티드 Processes for purification of proteins
CN103154243A (en) 2010-10-15 2013-06-12 日本化学研究株式会社 Method for producing glycoprotein having mannose residue as non-reducing end of sugar chain
EP2450375A1 (en) 2010-11-09 2012-05-09 Sandoz Gmbh Cell culture medium and process for protein expression, said medium and process comprising a PAM inhibitor
CA2815689C (en) 2010-11-11 2016-11-22 Abbvie Biotechnology Ltd. Improved high concentration anti-tnf.alpha. antibody liquid formulations
EP2640483A1 (en) 2010-11-15 2013-09-25 Biogen Idec Inc. Enrichment and concentration of select product isoforms by overloaded bind and elute chromatography
WO2012078376A1 (en) 2010-12-08 2012-06-14 Amgen Inc. Ion exchange chromatography in the presence of an amino acid
AU2012204237A1 (en) 2011-01-07 2013-07-04 Abbvie Inc. Anti-IL-12/IL-23 antibodies and uses thereof
TW201309330A (en) 2011-01-28 2013-03-01 Abbott Lab Compositions containing glycosylated antibodies and uses thereof
AR085302A1 (en) 2011-02-24 2013-09-18 Sanofi Sa METHOD OF PRODUCTION OF STIRATED ANTIBODIES
AU2012226398B9 (en) 2011-03-06 2017-04-13 Merck Serono S.A. Low fucose cell lines and uses thereof
JP2014514275A (en) 2011-03-10 2014-06-19 ボード・オブ・リージエンツ,ザ・ユニバーシテイ・オブ・テキサス・システム Protein nanoparticle dispersion system
WO2012125735A1 (en) 2011-03-15 2012-09-20 Abott Laboratories An integrated approach to the isolation and purification of antibodies
TWI622597B (en) 2011-03-28 2018-05-01 賽諾菲公司 Dual variable region antibody-like binding proteins having cross-over binding region orientation
EP2511293A1 (en) 2011-04-13 2012-10-17 LEK Pharmaceuticals d.d. A method for controlling the main complex N-glycan structures and the acidic variants and variability in bioprocesses producing recombinant proteins
WO2012145682A1 (en) 2011-04-21 2012-10-26 Amgen Inc. A method for culturing mammalian cells to improve recombinant protein production
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9670519B2 (en) 2011-04-29 2017-06-06 Biocon Limited Methods for reducing accumulation of lactate during culturing and method for producing polypeptide
US20140108084A1 (en) 2012-10-12 2014-04-17 Crestron Electronics, Inc. Initiating Schedule Management Via Radio Frequency Beacons
WO2012149393A2 (en) 2011-04-29 2012-11-01 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of t effector cells
KR101591671B1 (en) 2011-04-29 2016-02-04 바이오콘 리서치 리미티드 method for reducing heterogeneity of antibodies and a process of producing the antibodies thereof
US9562252B2 (en) 2011-05-13 2017-02-07 Biogen Ma Inc. Methods of preventing and removing trisulfide bonds
KR102108663B1 (en) 2011-07-01 2020-05-07 암젠 인크 Mammalian cell culture
WO2013006461A1 (en) 2011-07-01 2013-01-10 Biogen Idec Ma Inc. Cholesterol-based media supplementals for cell culture
US10323081B2 (en) 2011-07-06 2019-06-18 Genmag A/S Modulation of complement-dependent cytotoxicity through modifications of the C-terminus of antibody heavy chains
US9475858B2 (en) 2011-07-08 2016-10-25 Momenta Pharmaceuticals, Inc. Cell culture process
GB201112429D0 (en) 2011-07-19 2011-08-31 Glaxo Group Ltd Antigen-binding proteins with increased FcRn binding
WO2013013013A2 (en) 2011-07-21 2013-01-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for producing modified glycoproteins
US20140296490A1 (en) 2011-08-10 2014-10-02 Laboratoire Français Du Fractionnement Et Des Biotechnologies Highly galactosylated antibodies
US20130149300A1 (en) 2011-09-27 2013-06-13 Icon Genetics Gmbh MONOCLONAL ANTIBODIES WITH ALTERED AFFINITIES FOR HUMAN FCyRI, FCyRIIIa, AND C1q PROTEINS
US20140288278A1 (en) 2011-10-31 2014-09-25 Joseph Nti-Gyabaah Chromatography process for resolving heterogeneous antibody aggregates
JP6356607B2 (en) 2011-11-02 2018-07-11 ジェネンテック, インコーポレイテッド Overload / elution chromatography
AU2012355710B2 (en) 2011-12-19 2017-11-02 The Rockefeller University Non-sialylated anti-inflammatory polypeptides
EP3412310B1 (en) 2012-03-07 2022-09-07 Cadila Healthcare Limited Pharmaceutical formulations of tnf-alpha antibodies
SG10201701224UA (en) 2012-03-12 2017-04-27 Merck Patent Gmbh Removal of protein aggregates from biopharmaceutical preparations in a flowthrough mode
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158275A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Cell culture methods to reduce acidic species
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US20130281355A1 (en) * 2012-04-24 2013-10-24 Genentech, Inc. Cell culture compositions and methods for polypeptide production
US20140154270A1 (en) 2012-05-21 2014-06-05 Chen Wang Purification of non-human antibodies using kosmotropic salt enhanced protein a affinity chromatography
WO2013177115A2 (en) 2012-05-21 2013-11-28 Abbvie Inc. Novel purification of human, humanized, or chimeric antibodies using protein a affinity chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2013181585A2 (en) 2012-06-01 2013-12-05 Momenta Pharmaceuticals, Inc. Methods related to adalimumab
JP2015519382A (en) 2012-06-12 2015-07-09 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング Pharmaceutical formulations for therapeutic antibodies
WO2014018747A2 (en) 2012-07-26 2014-01-30 Momenta Pharmaceuticals, Inc. Glycoproteins with anti-inflammatory properties
EP3540438A1 (en) 2012-08-17 2019-09-18 MorphoSys AG Complex-specific antibodies and antibody fragments and its use
KR20150043523A (en) 2012-09-02 2015-04-22 애브비 인코포레이티드 Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
KR102238677B1 (en) 2012-09-07 2021-04-12 코히러스 바이오사이언시즈, 인코포레이티드 Stable aqueous formulations of adalimumab
US20150252108A1 (en) 2012-09-26 2015-09-10 Momenta Pharmaceuticals, Inc. Glycoprotein preparations
EP2931749B1 (en) 2012-12-17 2019-04-24 Laboratoire Francais du Fractionnement et des Biotechnologies Societe Anonyme Use of monoclonal antibodies for the treatment of inflammation and bacterial infections
US9844594B2 (en) 2012-12-18 2017-12-19 Merck Sharp & Dohme Corp. Liquid formulations for an anti-TNF α antibody
EP2956480B1 (en) 2013-02-13 2019-09-04 Laboratoire Français du Fractionnement et des Biotechnologies Highly galactosylated anti-tnf-alpha antibodies and uses thereof
CA2904146C (en) 2013-03-08 2023-03-21 Neuclone Biologics Pty Ltd A cell expression system
US8956830B2 (en) 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture
US9677105B2 (en) 2013-03-14 2017-06-13 Momenta Pharmaceuticals, Inc. Methods of cell culture
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
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
US20140271633A1 (en) 2013-03-14 2014-09-18 Abbvie Inc. Mammalian cell culture performance through surfactant supplementation of feed media
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
SG11201504260UA (en) 2013-03-14 2015-10-29 Abbvie Inc Low acidic species compositions and methods for producing and using the same
US20140271622A1 (en) 2013-03-14 2014-09-18 Momenta Pharmaceuticals, Inc. Methods of cell culture
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
US9518082B2 (en) 2013-03-15 2016-12-13 Alderbio Holdings Llc Antibody purification and purity monitoring
LT2970980T (en) 2013-03-15 2018-10-25 Janssen Biotech, Inc. Manufacturing methods to control c-terminal lysine, galactose and sialic acid content in recombinant proteins
EP2991666B1 (en) 2013-05-02 2020-03-25 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
KR101569783B1 (en) 2013-06-05 2015-11-19 한화케미칼 주식회사 A Method of Antibody Purification
TWI596107B (en) 2013-06-25 2017-08-21 卡地拉保健有限公司 Novel purification process for monoclonal antibodies
IN2013MU02285A (en) 2013-07-06 2015-06-19 Cadila Healthcare Ltd
CN105377237B (en) 2013-07-19 2019-04-19 德国赫素制药集团 Make the adjusting method and formulation relevant to the administration of biologics of immune response
US9751881B2 (en) 2013-07-31 2017-09-05 Merck Sharp & Dohme Corp. Inhibitors of the renal outer medullary potassium channel
EP3036320B2 (en) 2013-08-19 2024-04-03 Biogen MA Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
BR112016002622B1 (en) 2013-08-20 2023-04-11 Lek Pharmaceuticals D.D CELL CULTURE MEDIA AND USE OF SELENIUM
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
MX2016009848A (en) 2014-01-29 2017-02-17 Lg Life Sciences Ltd Method for controlling galactosylation of recombinant protein through optimization of culture medium.
US20160185848A1 (en) 2014-07-09 2016-06-30 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using sugars
US20160039924A1 (en) 2014-08-05 2016-02-11 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using dissolved oxygen
JP6807327B2 (en) 2014-12-22 2021-01-06 ユーシービー バイオファルマ エスアールエル Protein production
CN105777896B (en) 2015-03-19 2019-08-16 广东东阳光药业有限公司 A kind of purification process at antibody acidity peak
CN105777895A (en) 2015-03-19 2016-07-20 广东东阳光药业有限公司 Application of sodium phenylbutyrate in purification of antibody acidic peak
CN105777904B (en) 2015-03-23 2019-12-10 广东东阳光药业有限公司 cation exchange chromatography purification method of anti-TNF alpha monoclonal antibody

Also Published As

Publication number Publication date
US20150133639A1 (en) 2015-05-14
US9598667B2 (en) 2017-03-21
WO2015051293A3 (en) 2015-06-25
WO2015051293A2 (en) 2015-04-09
EP3052640A2 (en) 2016-08-10

Similar Documents

Publication Publication Date Title
US9598667B2 (en) Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9206390B2 (en) Methods to control protein heterogeneity
DK1896071T3 (en) Methods and compositions with increased therapeutic activity
KR102182885B1 (en) Compositions and methods for producing glycoproteins
JP2011512875A (en) Antibody with enhanced ADCC function
JP6971221B2 (en) How to increase the galactose content of recombinant proteins
SG192183A1 (en) Compositions containing glycosylated antibodies and uses thereof
AU2021258023B2 (en) Methods for modulating protein galactosylation profiles of recombinant proteins using peracetyl galactose
CA2746350C (en) Anti-rhd monoclonal antibodies
JP2021521854A (en) Antibodies with regulated glycan profiles
JP2022523475A (en) How to produce multimeric proteins in eukaryotic host cells
US20240002483A1 (en) Fab high mannose glycoforms
JP2021526014A (en) Regulation of antibody-dependent cellular cytotoxicity
AU2014200308A1 (en) Anti-RhD monoclonal antibodies
Dionne The study of culture redox potential’s effect on glycosylation and production of monoclonal antibodies in mammalian cell cultures

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WENTZ, ALANE E.;HEMMAVANH, DON;MATUCK, JOSEPH G.;REEL/FRAME:041900/0087

Effective date: 20150116

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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