US20180230228A1 - Method for increasing the galactose content of recombinant proteins - Google Patents

Method for increasing the galactose content of recombinant proteins Download PDF

Info

Publication number
US20180230228A1
US20180230228A1 US15/749,978 US201615749978A US2018230228A1 US 20180230228 A1 US20180230228 A1 US 20180230228A1 US 201615749978 A US201615749978 A US 201615749978A US 2018230228 A1 US2018230228 A1 US 2018230228A1
Authority
US
United States
Prior art keywords
cell culture
antibody
composition
cells
feeding
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
US15/749,978
Other languages
English (en)
Inventor
Ákos PUTICS
Dénes ZALAI
Gáspár NAGY
László PÁRTA
Áron SCHLEICHER
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.)
Richter Gedeon Nyrt
Original Assignee
Richter Gedeon Nyrt
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 Richter Gedeon Nyrt filed Critical Richter Gedeon Nyrt
Assigned to RICHTER GEDEON NYRT. reassignment RICHTER GEDEON NYRT. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZALAI, Dénes, PUTICS, Ákos, SCHLEICHER, Áron, PÁRTA, László, NAGY, Gáspár
Publication of US20180230228A1 publication Critical patent/US20180230228A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • 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
    • 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
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/40Nucleotides, nucleosides or bases
    • 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
    • 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
    • C12N2523/00Culture process characterised by temperature

Definitions

  • the present invention relates to a method for increasing the galactose content of a recombinant protein produced in mammalian cells, wherein during the cultivation of said cells the pH of the cell culture is changed and a composition comprising nucleosides, transition metal salts and/or sugars is fed.
  • Naturally occurring antibodies derived from mammalian serum are glycosylated in their constant region and this glycosylation is important for the effector functions of the antibodies such as antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • recombinant monoclonal antibodies produced in eukaryotic cells show a specific glycosylation pattern.
  • biosimilar therapeutic antibodies care must also be taken that the biosimilar antibody is comparable to the originator product in terms of glycosylation.
  • the prior art discloses various methods for modulating the galactosylation profile of a glycoprotein composition.
  • WO 2012/149197 A2 provides a method for controlling galactosylation using a manganese and/or galactose containing cell culture supplement.
  • EP 2 511 293 A1 describes a method for controlling galactosylation by pCO 2 regulation.
  • WO 2014/170866 A2 discloses a method for increasing the galactose content of recombinant proteins by reducing the temperature during the cell culture process and maintaining the pCO 2 level in a specific range.
  • the present inventors have found that a combination of a pH reduction and feeding the mammalian cells with uridine, manganese chloride and galactose increases the galactosylation of a recombinantly produced antibody to a greater extent than the feeding with uridine, manganese chloride and galactose without pH reduction.
  • the present invention relates to a method for increasing the galactose content of a recombinant protein produced in mammalian cells, said method comprising:
  • composition comprising at least two of the following components:
  • the present invention relates to a method for producing a recombinant protein in mammalian cells, said method comprising:
  • composition comprising at least two of the following components:
  • the recombinant protein is produced at large scale.
  • the mammalian cells are Chinese hamster ovary cells.
  • the recombinant protein is an Fc-containing protein.
  • the second pH is lower than the first pH and more preferably the second pH is 0.05 to 0.3 pH units lower than the first pH.
  • the nucleoside is uridine and more preferably the concentration of uridine within the composition is 1 to 20 mM.
  • the transition metal salt is manganese (II) chloride and more preferably the concentration of manganese (II) chloride within the composition is 0.002 mM to 0.1 mM.
  • the sugar is galactose and more preferably the concentration of galactose within the composition is 5 mM to 100 mM.
  • the present invention relates to a method for producing a rituximab biosimilar antibody in Chinese hamster ovary cells, said method comprising:
  • composition comprising the following components:
  • the present invention relates to a method for improving the biosimilarity of a therapeutic antibody produced by Chinese hamster ovary cells to its reference antibody, said method comprising the steps of:
  • composition comprising the following components:
  • the cells are cultured at the first pH until the viable cell density is 4.5 to 6.0 ⁇ 10 6 cells/ml.
  • the cells are cultured at the second pH for 6 to 7 days.
  • the temperature is kept constant during steps (a), (b) and (c).
  • composition further contains at least one amino acid selected from the group consisting of L-valine, L-cysteine, L-phenylalanine and L-serine.
  • step (c) is performed at least twice.
  • step (c) is preceded by a feeding step with a composition to which the components (i) and (iii) have not been added.
  • the culture medium in steps (a) and (b) does not contain uridine and galactose.
  • the composition of step (c) does not contain one or more of thymidine, fructose, mannose, sucrose and N-acetylmannosamine.
  • the osmolality of the culture in steps (a), (b) and (c) is lower than 400 mOsm/kg.
  • the term “consisting of” is considered to be a preferred embodiment of the term “comprising”. If hereinafter a group is defined to comprise at least a certain number of embodiments, this is meant to also encompass a group which preferably consists of these embodiments only. Furthermore, the terms “first”, “second”, “third” or “(a)”, “(b)”, “(c)”, “(d)” etc. and the like in the description and in the claims, are used for distinguishing between similar elements and not necessarily for describing a sequential or chronological order. It is to be understood that the terms so used are interchangeable under appropriate circumstances and that the embodiments of the invention described herein are capable of operation in other sequences than described or illustrated herein.
  • first”, “second”, “third” or “(a)”, “(b)”, “(c)”, “(d)”, “i”, “ii” etc. relate to steps of a method or use or assay there is no time or time interval coherence between the steps, i.e. the steps may be carried out simultaneously or there may be time intervals of seconds, minutes, hours, days, weeks, months or even years between such steps, unless otherwise indicated in the application as set forth herein above or below.
  • the present invention is based on the finding that a change, preferably a reduction, of the cell culture pH and the feeding of a composition comprising nucleosides, transition metal salts and sugars, preferably of uridine, manganese (II) chloride and galactose, to the cell culture increases the galactose content of a recombinant protein, preferably a recombinant antibody.
  • the term “increase of galactose content” is intended to mean that the percentage of one or all of the galactosylated isoforms G1F, G1′F and G2F in the recombinant protein is higher when the pH of the cell culture is changed, preferably lowered, and a composition comprising nucleosides, transition metal salts and sugars, preferably a composition comprising uridine, manganese chloride and galactose, is fed to the cell culture compared to the percentage of these isoforms in the same recombinant protein produced by a cell culture which is maintained at constant pH and to which the composition as defined above has not been fed.
  • This increase in the percentage of G1F, G1′F and G2F is accompanied by a decrease of non-galactosylated glyco forms such as G0 and G0F.
  • the G0F, G1F, G1′F and G2F glycoforms have the following structures:
  • Gn is N-acetylglucosamine
  • Fuc is fucose
  • M is mannose
  • Gal galactose
  • the galactose content is increased, if the sum of the percentage of the G1F, G1′F and G2F isoforms in the recombinant protein produced according to the methods of the invention is increased by at least 1%, 2% or 3%, preferably by at least 4%, 5%, 6% or 7%, more preferably by at least 8%, 9% or 10% and most preferably by at least 11% or 12% compared to the sum of the percentage of the G1F, G1′F and G2F iso forms in the same recombinant protein produced by a cell culture which is maintained at constant pH and to which the composition as defined above has not been fed.
  • the galactose content is also increased, if the percentage of the G0F isoform in the recombinant protein produced according to the method of the present invention is decreased by at least 1%, 2% or 3%, preferably by at least 4%, 5% or 6%, more preferably by at least 7%, 8% or 9% and most preferably by at least 10% compared to the percentage of the G0F isoform in the same recombinant protein produced by a cell culture which is maintained at constant pH and to which the composition as defined above has not been fed.
  • the galactose content is determined eight to fourteen days after inoculation of the cells into the cell culture medium. In a preferred embodiment, the galactose content is determined nine to ten days after inoculation of the cells into the cell culture medium.
  • the relative ratio of the different glycan iso forms of the recombinant protein, in particular of the galactosylated isoforms G1F, G1′F and G2F, and consequently of the galactose content can be determined by any method known in the art.
  • capillary electrophoresis using laser-induced fluorescence detection CE-LIF
  • the relative content of each of the glycan isoforms is determined by fluorescence detection and calculated using area % values of the corresponding peaks. An exemplary method is described in the Examples section herein below.
  • the term “inoculation of the cells into the cell culture medium” refers to the step of contacting the cells with the cell culture medium under conditions which are suitable for growth and proliferation of the cells.
  • recombinant protein refers to any protein which can be produced by mammalian cell culture as the result of the transcription and translation of a gene encoding said recombinant protein which gene is carried on a recombinant nucleic acid molecule that has been introduced into the mammalian host cell.
  • the recombinant protein may not be produced naturally in the mammalian cells used or the recombinant protein may be produced naturally in the mammalian cells used, but at a lower level.
  • the recombinant protein is not produced naturally by the mammalian host cell.
  • the term “recombinant protein” encompasses therapeutic proteins such as cytokines, growth factors, clotting factors and antibodies in which the galactose content influences the biological function of the protein.
  • the recombinant protein is an Fc containing protein such as an antibody or a fusion protein of the Fc portion of an IgG antibody with parts or all of another protein.
  • fusion protein of the Fc portion of an IgG antibody with parts or all of another protein examples include etanercept (fusion with TNF receptor), aflibercept (fusion with extracellular domains of VEGF receptors 1 and 2), abatacept (fusion with extracellular domain of CTLA-4) and belatacept (fusion with extracellular domain of CTLA-4).
  • the recombinant protein is a recombinant antibody.
  • the term “recombinant antibody” refers to any antibody which can be produced by mammalian cell culture as the result of the transcription and translation of a gene encoding said recombinant antibody which gene is carried on a recombinant nucleic acid molecule that has been introduced into the mammalian host cell.
  • the recombinant antibody may not be produced naturally in the mammalian cells used or the recombinant antibody may be produced naturally in the mammalian cells used, but at a lower level.
  • the recombinant antibody is not produced naturally by the mammalian host cell used for its production.
  • immunoglobulin and “antibody” are used interchangeably herein.
  • the immunoglobulin may be a monoclonal antibody, multispecific antibody (e.g. bispecific antibody) or fragments thereof exhibiting the desired antigen binding activity.
  • Naturally occurring antibodies are molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 Daltons, composed of two identical light chains and two identical heavy chains that are linked by disulfide bonds. From N- to C-terminus, each heavy chain has a variable domain (VH), also called a variable heavy domain or a heavy chain variable domain followed by three or four constant domains (CH1, CH2, CH3 and optionally CH4).
  • VH variable domain
  • each light chain has a variable domain (VL), also called a variable light domain or a light chain variable domain followed by a constant light chain (CL) domain.
  • VL variable domain
  • CL constant light chain
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • Antibody fragments comprise a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; single-chain antibody molecules; diabodies; linear antibodies; and multispecific antibodies formed from antibody fragments.
  • the immunoglobulin is a monoclonal antibody.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e. the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. In contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the immunoglobulin may be of the murine classes IgG1, IgG2a, IgG2b, IgM, IgA, IgD or IgE, the human classes IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD or IgE, or combinations or fragments thereof.
  • the immunoglobulin may recognize any one or a combination of proteins including, but not limited to the following antigens: CD2, CD3, CD4, CD8, CD11a, CD14, CD18, CD20, CD22, CD23, CD25, CD33, CD40, CD44, CD52, CD80 (B7.1), CD86 (B7.2), CD147, CD152, IL-1a, IL-1ß, IL-1, IL-2, IL-3, IL-7, IL-4, IL-5, IL-8, IL-10, IL-12, IL-23, IL-2 receptor, IL-4 receptor, IL-6 receptor, IL-12 receptor, IL-13 receptor, IL-18 receptor subunits, PDGF- ⁇ , and analogues thereof, PLGF, VEGF, TGF, TGF- ⁇ 2, TGF-p1, EGF receptor, PLGF receptor, VEGF receptor, platelet receptor gpIIb/IIIa, thrombopoeitin receptor, apoptosis receptor PD-1,
  • the immunoglobulin may for example be afelimomab, abciximab, adalimumab, alemtuzumab, arcitumomab, belimumab, canakinumab, cetuximab, denosumab, trastuzumab, imciromab, capromab, infliximab, ipilimumab, abciximab, rituximab, basiliximab, palivizumab, natalizumab, nivolumab, nofetumomab, omalizumab, daclizumab, ibritumomab, muromonab-CD3, edrecolomab, gemtuzumab, golimumab, certolizumab, eculizumab, ustekinumab, ocrelizumab, ofatumumab, obi
  • the immunoglobulin of the invention is preferably an IgG molecule, such as an IgG1, IgG2, IgG3, or IgG4 molecule. More preferably, the immunoglobulin is IgG1. Even more preferably, the immunoglobulin is an IgG1 wherein at least the Fc part is human.
  • the immunoglobulin may be a murine-human chimeric IgG1 wherein the Fc part of the IgG1 is human and the variable region is of mouse origin. Most preferably, the chimeric immunoglobulin is rituximab or infliximab.
  • Rituximab is a chimeric anti-CD20 antibody which is described in detail in, for example, WO 94/11026.
  • Infliximab is a chimeric anti-TNF ⁇ antibody which is described in detail in, for example, WO 92/16553.
  • the immunoglobulin may be a humanized IgG1 form of a murine progenitor in that the CDRs of the variable domain are derived from mouse and the framework regions of the variable domain are derived from human.
  • the humanized antibody is trastuzumab or bevacizumab.
  • Trastuzumab is a humanized anti-HER2 antibody which is described in detail in, for example, WO 92/22653.
  • Bevacizumab is a humanized anti-VEGF antibody which is described in detail in, for example, WO 98/45331.
  • the immunoglobulin may be a fully human IgG1 antibody, i.e. an antibody in which all parts are derived from human origin. Most preferably the human antibody is adalimumab or denosumab.
  • Adalimumab is a human anti-TNF ⁇ antibody which is described in detail in, for example, WO 97/29131.
  • Denosumab is a human anti-RANKL antibody which is described in detail in, for example, WO 03/002713.
  • the antibody may be rituximab or bevacizumab.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, as long as they exhibit the desired biological activity.
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, as long as they exhibit the desired biological activity.
  • the monoclonal antibodies herein also include “humanized” antibodies. Such antibodies are obtained by “humanization” of non-human (for example murine) antibodies and contain only minimal sequences derived from the animal immunoglobulin. Most of the molecule is comprised of human amino acid sequence. Residues from a hypervariable region of the human recipient antibody are replaced by residues from a hypervariable region of a non-human donor antibody having the desired binding properties.
  • the monoclonal antibodies herein also include fully human antibodies which may initially be obtained by screening of a human antibody library.
  • the recombinant protein is produced in mammalian cells.
  • suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO) cells (including dhfr negative CHO cells used with a DHFR selectable marker), NS0 myeloma cells, COS cells, SP2 cells, monkey kidney CV1, human embryonic kidney line 293; baby hamster kidney cells (BHK), mouse Sertoli cells (TM4), African green monkey kidney cells (VERO-76), human cervical carcinoma cells (HELA); canine kidney cells (MDC), buffalo rat liver cells (BRL 3 A), human lung cells (W138), human liver cells (Hep G2), mouse mammary tumor cells (MMT 060562), TRI cells, MRC 5 cells and FS4 cells.
  • CHO Chinese Hamster Ovary
  • the host cells are derived from a rodent. More preferably, the mammalian cells are Chinese hamster ovary (CHO) cells, even more preferably the cells are CHO-K1 cells and most preferably the cells are CHO-K1 cells adapted for growth in serum-free media (CHO-S) and/or are obtainable from Invitrogen (Catalogue number R-800-07).
  • CHO Chinese hamster ovary
  • the cells are CHO-K1 cells and most preferably the cells are CHO-K1 cells adapted for growth in serum-free media (CHO-S) and/or are obtainable from Invitrogen (Catalogue number R-800-07).
  • the mammalian cells have been transformed, i.e. genetically modified, with at least one recombinant nucleic acid molecule such as an expression vector which enables the stable production of the recombinant protein in the mammalian host cells.
  • the mammalian cells may either be transformed with one recombinant nucleic acid molecule which encodes both the heavy and the light chain of the antibody or with two recombinant nucleic acid molecules of which one encodes the light chain of the antibody and the other one encodes the heavy chain of the antibody.
  • the recombinant antibody is produced from one recombinant nucleic acid molecule which encodes both the heavy and the light chain of the antibody.
  • the recombinant antibody is produced from one recombinant nucleic acid molecule and the expression of the heavy and the light chain is controlled by separate promoters which may be the same or different.
  • the recombinant antibody is produced from one recombinant nucleic acid molecule and the expression of the heavy and the light chain is controlled by separate promoters which are the same.
  • a cell culture medium provides essential and non-essential amino acids, vitamins, energy sources, lipids, and trace elements required by the cell for minimal growth and/or survival.
  • the medium is chemically defined in that all its components and their concentration are known.
  • the medium is serum-free and hydrolysate-free and does not contain any components derived from animals.
  • the medium is serum-free and hydrolysate-free and does not contain any components derived from animals, but contains HEPES and Pluronic® F-68.
  • the medium used in steps (a) and (b) of the method of the present invention i.e. the steps before feeding
  • PowerCHO-2 CD available from Lonza under Catalogue number BE12-771Q
  • the PowerCHO-2 CD medium is supplemented with recombinant insulin, lipids, ferric citrate and PEG20000 and extra amounts of L-tyrosine, L-phenylalanine and L-glutamine.
  • the extra amounts of L-tyrosine, L-phenylalanine and L-glutamine comprise 8 mM L-glutamine, 1.2 mM L-tyrosine and 2 mM L-phenylalanine.
  • the cell culture medium may contain any or all of these compounds, if they are present in the chemically defined medium used. More preferably, the cell culture medium does not comprise any galactose.
  • the cell culture medium is preferably subjected to sterile filtration, more preferably to sterile filtration using a filter with 0.1 micron pore size.
  • the pH of the cell culture medium in step a) of the method of the present invention which is also called “the first pH” is maintained within a range of between pH 7.15 to 7.25, preferably by adding Na 2 CO 3 or H 3 PO 4 , for a first period of time.
  • the first period of time is 60 to 80 hours, preferably 63 to 79 hours, more preferably 66 to 78 hours and most preferably 70 hours after inoculation of the cell culture medium with the mammalian cells.
  • the pH of the cell culture medium is changed, preferably lowered, to a second pH. More preferably, the second pH is 0.05 to 0.3 pH units lower than the first pH and even more preferably, the second pH is 0.15 to 0.25 pH units lower than the first pH. Most preferably, the second pH is pH 7.00.
  • the pH may be lowered by adding a suitable acid or CO 2 gas, preferably by adding H 3 PO 4 .
  • the pH is changed when a viable cell density of 4.0 to 7.0 ⁇ 10 6 has been reached.
  • the second period of time in which the cells are cultured at the second pH is about 6 to 11 days, or about 6 to 8 days, preferably about seven days.
  • the overall cultivation period in the method of the present invention is eight to fourteen days after inoculation of the cell culture medium with the mammalian cells.
  • the overall cultivation period in the method of the present invention is nine to ten days after inoculation of the cell culture medium with the mammalian cells.
  • the cells are fed in step (c) with a composition comprising at least two of the following components: (i) one or more nucleosides, (ii) one or more transition metal salts and (iii) one or more sugars (hereinafter also called components (i) to (iii)), in particular with a composition comprising uridine, manganese chloride and galactose.
  • a composition comprising at least two of the following components: (i) one or more nucleosides, (ii) one or more transition metal salts and (iii) one or more sugars (hereinafter also called components (i) to (iii)), in particular with a composition comprising uridine, manganese chloride and galactose.
  • feeding means that the composition is added to the cell culture of step (a) or (b) and no medium or cells is withdrawn during the feeding.
  • the feeding typically does not occur continuously, but at defined time points.
  • the composition is fed at defined time points as further detailed below.
  • the composition which is fed may only comprise components (i) to (iii), e.g. in water or a suitable buffer, or it may be based on a cell culture medium which additionally contains components (i) to (iii).
  • the composition which is fed in step (c) of the method is based on a cell culture medium which additionally contains components (i) to (iii).
  • This cell culture medium may be the same or different as the cell culture medium used in the initial culturing of the cells, i.e. after inoculation and before feeding (steps (a) and (b)).
  • the cell culture medium used for feeding is different from the one used in the initial culturing of the cells (i.e. steps (a) and (b)). More preferably, the cell culture medium used for feeding is ExCell® of Sigma Aldrich. In another preferred embodiment, the cell culture medium used for feeding is customized salt-free (SF) ExCell® of Sigma Aldrich.
  • the cell culture medium used for feeding may also contain other components such as amino acids and other supplements in addition to the basic cell culture medium.
  • the cell culture medium used for feeding additionally contains one or more of L-valine, L-cysteine, L-phenylalanine, L-serine and a chemically defined supplement such as BD Recharge.
  • the cell culture medium used for feeding comprises L-valine, L-cysteine, L-phenylalanine, L-serine and a chemically defined supplement in addition to components (i) to (iii).
  • the cell culture medium used for feeding is ExCell® or customized salt-free SF ExCell® and comprises L-valine, L-cysteine, L-phenylalanine, L-serine and a chemically defined supplement in addition to components (i) to (iii).
  • the concentration of L-valine in the cell culture medium used for feeding is 34 mM
  • the concentration of L-cysteine in the cell culture medium used for feeding is 8.3 mM
  • the concentration of L-phenylalanine in the cell culture medium used for feeding is 4.5 mM
  • the concentration of L-serine in the cell culture medium used for feeding is 38 mM.
  • the cell culture medium used for feeding is preferably subjected to sterile filtration, more preferably to sterile filtration using a filter with 0.2 or 0.1 micron pore size.
  • composition used for feeding does not contain thymidine, fructose, mannose, sucrose and N-acetylmannosamine.
  • the composition used for feeding comprises one or more nucleoside(s).
  • Nucleosides are composed of a nitrogenous base and a sugar comprising five carbon atoms such as ribose and desoxyribose.
  • Examples of nucleosides include cytidine, uridine, adenosine, guanosine, thymidine and inosine.
  • the nucleoside is uridine.
  • the concentration of the one or more nucleoside(s) within the composition used for feeding is 1 to 20 mM, preferably 1.5 to 15 mM, more preferably 2 to 12 mM, even more preferably 2.5 to 10 mM and most preferably it is 3 mM.
  • the concentration of uridine within the composition used for feeding is 1 to 20 mM, preferably 1.5 to 15 mM, more preferably 2 to 12 mM, even more preferably 2.5 to 10 mM and most preferably it is 3 mM.
  • the composition used for feeding further comprises one or more transition metal salt(s).
  • Transition metal salts are salts of a transition metal with a counterion. Transition metals include Fe, Co, Cr, Mn, Mo, Ni, Cu, Zn and suitable counterions include chloride (Cl ⁇ ), sulphate (SO 4 2 ⁇ ) and phosphate (PO 4 3 ⁇ ).
  • the transition metal salt is a manganese salt and most preferably it is manganese (II) chloride.
  • the concentration of the one or more transition metal salt(s) within the composition used for feeding is 0.002 mM to 0.1 mM, preferably 0.005 mM to 0.09 mM, more preferably 0.008 mM to 0.08 mM, even more preferably 0.01 mM to 0.07 mM and most preferably it is 0.06 mM.
  • the concentration of manganese (II) chloride within the composition used for feeding is 0.002 mM to 0.1 mM, preferably 0.005 mM to 0.09 mM, more preferably 0.008 mM to 0.08 mM, even more preferably 0.01 mM to 0.07 mM and most preferably it is 0.06 mM.
  • the composition used for feeding further comprises one or more sugar(s).
  • Sugars are short-chain carbohydrates and include glucose, fructose, sucrose, galactose, maltose and lactose.
  • the sugar is galactose.
  • the concentration of the one or more sugar(s) within the composition used for feeding is 5 mM to 100 mM, preferably 7.5 mM to 75 mM, more preferably 10 mM to 60 mM, even more preferably 12.5 mM to 50 mM and most preferably it is 15 mM.
  • the concentration of galactose within the composition used for feeding is 5 mM to 100 mM, preferably 7.5 mM to 75 mM, more preferably 10 mM to 60 mM, even more preferably 12.5 mM to 50 mM and most preferably it is 15 mM.
  • the concentration of the one or more nucleoside(s) within the composition used for feeding is 1 to 20 mM, the concentration of the one or more transition metal salt(s) within the composition used for feeding is 0.002 mM to 0.1 mM and the concentration of the one or more sugar(s) within the composition used for feeding is 5 mM to 100 mM.
  • the concentration of one or more nucleoside(s) within the composition used for feeding is 3 mM, the concentration of the one or more transition metal salt(s) within the composition used for feeding is 0.06 mM and the concentration of the one or more sugar(s) within the composition used for feeding is 15 mM.
  • the concentration of uridine within the composition used for feeding is 1 to 20 mM
  • the concentration of manganese (II) chloride within the composition used for feeding is 0.002 mM to 0.1 mM
  • the concentration of galactose within the composition used for feeding is 5 mM to 100 mM.
  • the concentration of uridine within the composition used for feeding is 3 mM
  • the concentration of manganese (II) chloride within the composition used for feeding is 0.06 mM
  • the concentration of galactose within the composition used for feeding is 15 mM.
  • the cell culture is fed with the composition comprising components (i) to (iii) at least once, preferably at least twice, more preferably it is fed twice.
  • the feeding with the composition comprising components (i) to (iii) preferably occurs four to six days after the inoculation of the cell culture medium with the cells, more preferably it occurs five days after the inoculation of the cell culture medium with the cells.
  • the first feeding with the composition comprising components (i) to (iii) is performed twice, the first feeding with the composition comprising components (i) to (iii) is performed four to six days, preferably five days, after the inoculation of the cell culture medium and the second feeding with the composition comprising components (i) to (iii) is performed six to eight days, preferably seven days, after the inoculation of the cell culture medium. More preferably, the first feeding with the composition comprising components (i) to (iii) is performed five days after inoculation and the second feeding with the composition comprising components (i) to (iii) is performed seven days after inoculation.
  • the composition comprising components (i) to (iii) is diluted by a factor of 8.5 to 10.5, preferably by a factor of 9.0 to 10.0 and most preferably by a factor of 9.3.
  • the composition comprising components (i) to (iii) is diluted by a factor of 9.5 to 11.5, preferably by a factor of 10.0 to 11.0 and most preferably by a factor of 10.3.
  • the one or more steps of feeding with the composition comprising components (i) to (iii) are preceded by a feeding step with a composition to which the components (i) to (iii) have not been added, but which is otherwise identical to the composition comprising components (i) to (iii).
  • the feeding with a composition to which the components (i) to (iii) have not been added, but which is otherwise identical to the composition comprising components (i) to (iii) takes place two to four days, preferably three days, after the inoculation of the cell culture medium.
  • the method of the present invention preferably comprises the following feeding steps:
  • the temperature of the cell culture i.e. the cell culture medium comprising the mammalian cells
  • the temperature during the culturing process is set to 36° C. to 38° C. and more preferably the temperature is set to 37° C.
  • the osmolality is preferably lower than 400 mOsm/kg throughout the whole process, i.e. steps (a) to (c), as defined in the claims.
  • the osmolality is in the range of 250 to 400 mOsm/kg, more preferably in the range of 300 to 380 mOsm/kg and most preferably in the range of 330 to 370 mOsm/kg.
  • the term “osmolality” as used herein is defined as osmoles of solute per kilogram of solvent and may include ionized or non-ionized molecules.
  • a low osmolality such as an osmolality lower than 400 mOsm/kg can be maintained by using media with a low salt concentration.
  • the composition used for feeding contains a low salt concentration or contains no salt other than the transition metal salt used in feeding step c) at all.
  • the cells are cultured under aerobic conditions, i.e. a level of dissolved oxygen of 50 ⁇ 40%.
  • the level of carbon dioxide is maintained within a range of between 0 to 90 mmHg, optionally by adjusting the mixing rate or the intensity of aeration.
  • the process of the present invention is performed without glucose limitation. Accordingly, glucose is added to the cell culture to keep the glucose level in the range of 5 to 35 mM, preferably in the range of 10 to 25 mM.
  • antifoam agent may be added to the culture at any time during the process of the present invention.
  • the product is harvested. Since recombinant proteins, in particular antibodies, expressed from mammalian cells are typically secreted into the cell culture fluid during the cultivation process, the product harvest at the end of the cultivation process occurs by separating cell culture fluid comprising the recombinant protein from the cells.
  • the cell separation method should be gentle to minimize cell disruption to avoid the increase of cell debris and release of proteases and other molecules that could affect the quality of the immunoglobulin product.
  • the harvesting of the cell culture fluid comprising the recombinant protein involves centrifugation and/or filtration, whereby the recombinant protein is present in the supernatant and the filtrate, respectively. Expanded bed adsorption chromatography is an alternative method to avoid centrifugation/filtration methods.
  • the recombinant protein After harvesting the cell culture fluid comprising the recombinant protein the recombinant protein has to be purified from the cell culture fluid.
  • the purification of recombinant proteins and in particular recombinant antibodies is usually accomplished by a series of chromatographic steps such as anion exchange chromatography, cation exchange chromatography, affinity chromatography, hydrophobic interaction chromatography, hydroxyapatite chromatography and size exclusion chromatography. Further, the purification process may comprise one or more ultra-, nano- or diafiltration steps.
  • One particularly suitable method which is described in PCT/EP2015/054862 involves the steps of anion exchange chromatography in the flow-through mode, affinity chromatography on a protein A resin and cation exchange chromatography in the bind-and-elute mode.
  • the processes of the present invention are suitable for producing the recombinant protein at large scale, meaning in a culture volume of at least 500 or 1,000 liters, preferably at least 5,000 or 8,000 liters and most preferably of 10,000 or 20,000 liters.
  • the process of the present invention improves the biosimilarity of a biosimilar therapeutic antibody to its reference product, i.e. the marketed therapeutic antibody.
  • a biosimilar therapeutic antibody is a therapeutic antibody which is marketed after the patent protection for the original product has expired and which has the same amino acid sequence as the original product, but may slightly differ in posttranslational modifications. Nevertheless, they show a physiological effect which is identical to that of the original product.
  • an application for a marketing authorization for a biosimilar of a marketed antibody is filed, it has to be shown that the structure of the biosimilar antibody is comparable to the reference product.
  • One important parameter for assessing biosimilarity of glycosylated proteins is the glycosylation pattern which may influence the effector functions of the antibody.
  • the glycosylation pattern and in particular the galactose level of the biosimilar antibody is comparable to that of the reference product, thereby improving the biosimilarity compared to the glycosylation pattern and the biosimilarity of a therapeutic antibody which has not been subjected to a pH reduction, and which has not been fed with a composition comprising uridine, manganese (II) chloride and galactose.
  • the invented methods do neither depend on specific antibodies nor on specific host cells used for the expression of the immunoglobulins. The same is true for the mode of expression and the selected culture conditions, which were optimized in terms of protein galactosylation profile and maximum yields in the harvest.
  • CHO-S Chinese Hamster Ovary Cell line S
  • CHO-S Chinese Hamster Ovary Cell line S
  • the CHO-S cells were adapted to growth in serum-free, chemically-defined PowerCHO-2 medium (Lonza Inc US).
  • CHO cells genetically engineered to express the model antibody were grown initially in basal media PowerCHO-2 (Lonza). On every second day from the 3rd day (post inoculation) of cultivation on three times concentrated (3 ⁇ ) ExCell feed (37 g/L; SAFC) at a feed to initial working volume (volume of the basal medium plus inoculum) ratio of 15% was added to the culture in shot-wise mode.
  • the cultivation temperature was maintained at 37° C.
  • the pH was kept in the range of between pH 7.05 and pH 7.15 by addition of 0.5 M Na 2 CO 3 or H 3 PO 4 .
  • Dissolved oxygen (DO) set point was 40%.
  • the relevant metabolites were measured every day.
  • the glucose level was maintained at about 20 mM.
  • Cells were cultivated for between 9 to 10 days.
  • the experiments were mainly performed with harvested culture fluid from a laboratory scale of 1, 5, 10 or 100 L.
  • the production scale and maximum culture volume used in the examples was 1000 L. If not specified otherwise, the scale always refers to the culture volume.
  • the obtained model antibody was affinity purified from the fermentation broth using Protein A chromatography. This capture offers an exceptional selectivity for Fc-bearing molecules, thereby removing more than 99.5% of contaminants in a single step.
  • Viable cell density and cell viability were determined by CountessTM Automated Cell Counter (Invitrogen Carlsbad, Calif., 2008) using the Trypan blue staining method.
  • Glucose concentration was measured with Accu-Chek blood glucose meter (Roche, Mannheim, Germany).
  • Dissolved carbon-dioxide content (pCO2) was determined with the ABL80 blood gas analyzer (Radiometer, Bronshoj, Denmark).
  • At-line pH measurement for in situ pH meter re-calibration was performed with a S47 SevenMulti pH meter (Mettler Toledo, Zurich, Switzerland).
  • Osmolality of the samples was determined with Advanced Model 2020 multi-sample osmometer (Advanced Instruments, Norwood, Mass.).
  • Protein titer of the (in-process) samples was determined by Protein A affinity HPLC.
  • Determination of the relative ratio of a glycan population expressed in migration time corrected area % of glycan forms was performed by capillary electrophoresis using laser-induced fluorescence detection (CE-LIF). Protein samples (200 ⁇ g) were deglycosylated by incubation with PNGase-F for 3 hours at 37° C. Precipitation of proteins was performed using chilled ethanol, followed by drying. Reductive amination using fluorescent derivatizing agent 9-Aminopyrene-1,4,6-trisulfonic acid (APTS) and sodium cyanoborohydride was followed by heating for 90 minutes at 55° C.
  • APTS fluorescent derivatizing agent 9-Aminopyrene-1,4,6-trisulfonic acid
  • the bioactivity of the model antibody was determined using the complement-dependent cytotoxicity (CDC) assay.
  • the basis of the CDC method is that the model antibody binds in a specific manner to its antigen expressed on the surface of the target cells; the thus formed antigen-antibody complex activates the complement system, as a result of which the cells die in a dose-dependent manner.
  • Surviving cells are detected by the addition of AlamarBlue® reagent.
  • the evaluation of CDC assay is based on the comparison of the sigmoid dose-response curves obtained for the dilution series of both the sample and the reference.
  • CHO cells genetically engineered to express the model antibody were grown initially in basal media (PowerCHO-2, Lonza Inc US). On every second day from the 3rd day (post inoculation) on 15% 3 ⁇ concentrated ExCell feed (37 g/L; SAFC) was added to the culture in shot-wise mode.
  • Table 1 shows the medium supplementation during the respective experiments.
  • Basal Medium + Basal medium PowerCHO-2 CD + 2 amino acids; 8 mM Gln + Tyr + Phe 15% (3x) SF ExCell Basal feed: 15% (37 g/L) 3x SF ExCell CD on day feed + 6 amino acids 3, 5, 7, 9 . . . Supplementation: Pro, Cys, Val, Phe, Ser on day 3, 5, 7, 9 . . .; Tyr (3x cc.) only on day 5
  • the cultivation temperature was maintained at 37° C.
  • the pH was kept in the range of between pH 7.05 to pH 7.15 by addition of 0.5 M Na 2 CO 3 or H 3 PO 4 .
  • Dissolved oxygen (DO) set point was 40%.
  • the relevant metabolites were measured every day.
  • the glucose level was maintained at about 20 mM.
  • Cells were cultivated for between 9-10 days.
  • the glycosylation pattern was analyzed daily from samples of the fermentation broth from the 3rd day (post inoculum) of cultivation on.
  • the obtained antibody was affinity purified from the fermentation broth using protein A.
  • Cell viability, titer and osmolality were assessed daily from the 3rd day post inoculum of cultivation on.
  • the bioactivity of the obtained antibody was determined using the complement-dependent cytotoxicity (CDC) assay.
  • Table 2 shows on the basis of several results of samples from fed-batch experiments that increasing osmolality led to a significantly poorer glycosylation pattern implying that an osmolality of higher than 400 mOsm/kg may have a negative impact on protein glycosylation. Due to the accumulation of salts in the culture, the amount of non-galactosylated forms (G0F) was increasing, while the amount of the galactosylated forms was decreasing. Analogously, bioactivity of the respective antibody samples according to a CDC Assay was decreasing with increasing osmolality of the fermentation broth.
  • the original ExCell feed composition was modified to contain 66% less sodium phosphate compared to the original medium.
  • the antibody glycosylation pattern after 10 days of cultivation (post inoculum) from fed-batch fermentation runs A and B is shown in Table 3.
  • the galactosylation pattern of the obtained antibody was investigated after UMG feed and slight pH shift at different time points of the application of the pH shift using the AMBR (Advanced Microscale Bioreactor) system of TAP Biosystems, UK, which is a high-throughput down-scale fermentation platform that mimics the characteristics of bench-top bioreactors in microscale.
  • AMBR Advanced Microscale Bioreactor
  • CHO cells genetically engineered to express the model antibody were cultivated for 9 days in basal medium (PowerCHO-2, Lonza Inc US). On every second day from the 3rd day (post inoculation) on 15% (3 ⁇ ) SF ExCell feed (37 g/L; SAFC) supplemented according to Experiment B (Example 2.1, Table 1 with the exception of Proline) was added to the culture in shot-wise mode.
  • UMG supplement (3 mM uridine, 0.06 mM manganese (II) chloride and 15 mM galactose was added on days 5 and 7 (post inoculation) of cultivation together with the customized SF ExCell feed.
  • the cultivation pH was kept at pH 7.15 by addition of 0.5 M Na 2 CO 3 or H 3 PO 4 until day 3 (post inoculum) of cultivation. Shifts towards pH 7.00 were performed at different time points between 65th-78th hours after inoculation, at a viable cell density of between 4.0-7.0 ⁇ 10 6 cells/mL by addition of H 3 PO 4 .
  • the galactosylation pattern of the produced antibody was analysed with capillary electrophoresis from the crude purified protein.
  • Table 5 shows the percental distribution of the, G0F, G1F, G1′F and G2F galactosylation of the obtained antibody samples on days 8 and 9 of cultivation (post inoculum), respectively, and the time point when the pH shift was applied.
  • the analysis of the galactosylation pattern on the 8 th day of cultivation shows a decrease in the percental amount of non-galactosylated glycoform G0F compared to the control samples, where pH 7.15 was kept constant during fermentation.
  • the combination of pH shift and UMG feed resulted in an increase of the percental amount of galactosylated glycoforms (G1F, G1′F and G2F) of the obtained antibody.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Analytical Chemistry (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
US15/749,978 2015-08-04 2016-08-04 Method for increasing the galactose content of recombinant proteins Abandoned US20180230228A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
HUP1500363A HU231463B1 (hu) 2015-08-04 2015-08-04 Módszer rekombináns proteinek galaktóz tartalmának növelésére
HUP1500363 2015-08-04
PCT/EP2016/068651 WO2017021493A1 (fr) 2015-08-04 2016-08-04 Procédé pour augmenter la teneur en galactose de protéines recombinantes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/068651 A-371-Of-International WO2017021493A1 (fr) 2015-08-04 2016-08-04 Procédé pour augmenter la teneur en galactose de protéines recombinantes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/749,499 Continuation US20220372157A1 (en) 2015-08-04 2022-05-20 Method for increasing the galactose content of recombinant proteins

Publications (1)

Publication Number Publication Date
US20180230228A1 true US20180230228A1 (en) 2018-08-16

Family

ID=89658303

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/749,978 Abandoned US20180230228A1 (en) 2015-08-04 2016-08-04 Method for increasing the galactose content of recombinant proteins
US17/749,499 Pending US20220372157A1 (en) 2015-08-04 2022-05-20 Method for increasing the galactose content of recombinant proteins

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/749,499 Pending US20220372157A1 (en) 2015-08-04 2022-05-20 Method for increasing the galactose content of recombinant proteins

Country Status (8)

Country Link
US (2) US20180230228A1 (fr)
EP (1) EP3331911A1 (fr)
JP (1) JP6971221B2 (fr)
KR (1) KR102301702B1 (fr)
CN (1) CN108350075B (fr)
CA (1) CA2994611C (fr)
HU (1) HU231463B1 (fr)
WO (1) WO2017021493A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111321188A (zh) * 2018-12-17 2020-06-23 嘉和生物药业有限公司 一种抗体糖型改造的配方、细胞培养方法以及在工业化生产中的应用
WO2021066772A1 (fr) * 2019-10-01 2021-04-08 Arven Ilac Sanayi Ve Ticaret Anonim Sirketi Milieu de culture cellulaire pour réduire la fucosylation et des variants basiques dans la production d'anticorps

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020033827A1 (fr) * 2018-08-10 2020-02-13 Genentech, Inc. Stratégies de culture cellulaire pour moduler la glycosylation de protéines
HU231514B1 (hu) * 2018-11-07 2024-07-28 Richter Gedeon Nyrt. Sejttenyészetben előállított rekombináns glikoprotein glikozilációs mintázatának megváltoztatására szolgáló módszer
US20220177818A1 (en) * 2019-04-01 2022-06-09 The Automation Partnership (Cambridge ) Ltd. Operation process for a cell cultivation system
KR20220019725A (ko) * 2019-06-10 2022-02-17 다케다 야쿠힌 고교 가부시키가이샤 항체를 생성하기 위한 세포 배양 방법 및 조성물
CN113403281B (zh) * 2020-03-16 2024-02-27 夏尔巴生物技术(苏州)有限公司 一种提高抗体半乳糖基化水平的细胞培养方法
JP2023538881A (ja) * 2020-08-14 2023-09-12 ブリストル-マイヤーズ スクイブ カンパニー タンパク質の製造方法

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2156859T5 (es) 1991-03-18 2008-03-16 New York University Anticuerpos monoclonales y quimericos especificos para el factor de necrosis tumoral humano.
JP4124480B2 (ja) 1991-06-14 2008-07-23 ジェネンテック・インコーポレーテッド 免疫グロブリン変異体
PT752248E (pt) 1992-11-13 2001-01-31 Idec Pharma Corp Aplicacao terapeutica de anticorpos quimericos e marcados radioactivamente contra antigenios de diferenciacao restrita de linfocitos b humanos para o tratamento do linfoma de celulas b
HU230515B1 (hu) 1996-02-09 2016-10-28 Abbvie Biotechnology Ltd Humán TNFalfa-kötő antitestek alkalmazásai
CA2286330C (fr) 1997-04-07 2008-06-10 Genentech, Inc. Anticorps anti-vegf
MEP32608A (en) 2001-06-26 2011-02-10 Amgen Fremont Inc Antibodies to opgl
AR059065A1 (es) * 2006-01-23 2008-03-12 Amgen Inc Metodos para modular el contenido de manosa de las proteinas recombinantes
RU2011116319A (ru) * 2008-09-26 2012-11-10 Шеринг Корпорейшн (US) Производство высокого титра антител
KR101828624B1 (ko) * 2010-04-26 2018-02-12 노파르티스 아게 개선된 세포 배양 방법
EP2511293A1 (fr) 2011-04-13 2012-10-17 LEK Pharmaceuticals d.d. Procédé pour le contrôle des structures de N-glycane de complexes principaux et les variantes acides et variabilité dans les bioprocessus fabriquant des protéines recombinantes
EP2702077A2 (fr) * 2011-04-27 2014-03-05 AbbVie Inc. Procédé de contrôle du profil de galactosylation de protéines exprimées de manière recombinante
WO2014170866A2 (fr) 2013-04-18 2014-10-23 Dr. Reddy's Laboratories Limited Procédé d'obtention d'une composition glycoprotéique ayant une teneur accrue en galactosylation

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111321188A (zh) * 2018-12-17 2020-06-23 嘉和生物药业有限公司 一种抗体糖型改造的配方、细胞培养方法以及在工业化生产中的应用
WO2021066772A1 (fr) * 2019-10-01 2021-04-08 Arven Ilac Sanayi Ve Ticaret Anonim Sirketi Milieu de culture cellulaire pour réduire la fucosylation et des variants basiques dans la production d'anticorps

Also Published As

Publication number Publication date
WO2017021493A1 (fr) 2017-02-09
JP2018521676A (ja) 2018-08-09
JP6971221B2 (ja) 2021-11-24
HUP1500363A2 (en) 2017-02-28
CN108350075B (zh) 2022-03-25
EP3331911A1 (fr) 2018-06-13
KR20180070553A (ko) 2018-06-26
KR102301702B1 (ko) 2021-09-15
CA2994611C (fr) 2024-06-18
CN108350075A (zh) 2018-07-31
CA2994611A1 (fr) 2017-02-09
HU231463B1 (hu) 2024-01-28
US20220372157A1 (en) 2022-11-24

Similar Documents

Publication Publication Date Title
CA2994611C (fr) Procede pour augmenter la teneur en galactose de proteines recombinantes
US9206390B2 (en) Methods to control protein heterogeneity
US12043845B2 (en) Methods of cell culture
AU2021258023B2 (en) Methods for modulating protein galactosylation profiles of recombinant proteins using peracetyl galactose
CN111954719A (zh) 细胞培养物中产生的抗体的总去岩藻糖基化糖型
JP2024112833A (ja) 細胞培養で生産される組換え糖タンパク質のグリコシル化プロファイルを変更する方法
JP2021506323A (ja) ポリエーテルイオノフォアを使用するタンパク質マンノシル化プロファイルの調節方法
KR20230109674A (ko) Fab 고 만노스 당형
JP2024527450A (ja) 抗体産物のアフコシル化を調節するための方法
WO2016162514A1 (fr) Procédés de modulation des profils de glycosylation des protéines de protéines de recombinaison

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: RICHTER GEDEON NYRT., HUNGARY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PUTICS, AKOS;ZALAI, DENES;NAGY, GASPAR;AND OTHERS;SIGNING DATES FROM 20180404 TO 20180418;REEL/FRAME:046396/0510

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

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

Free format text: ADVISORY ACTION MAILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

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

Free format text: ADVISORY ACTION MAILED

STCB Information on status: application discontinuation

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