WO2013114164A1 - Method for obtaining glycoprotein composition with increased afucosylation content - Google Patents

Method for obtaining glycoprotein composition with increased afucosylation content Download PDF

Info

Publication number
WO2013114164A1
WO2013114164A1 PCT/IB2012/057090 IB2012057090W WO2013114164A1 WO 2013114164 A1 WO2013114164 A1 WO 2013114164A1 IB 2012057090 W IB2012057090 W IB 2012057090W WO 2013114164 A1 WO2013114164 A1 WO 2013114164A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
temperature
process according
cultured
glycoprotein
Prior art date
Application number
PCT/IB2012/057090
Other languages
French (fr)
Inventor
Asma AHMED
Indraneel DASARI
Mohit NARESH
Original Assignee
Dr. Reddy's Laboratories Limited
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 Dr. Reddy's Laboratories Limited filed Critical Dr. Reddy's Laboratories Limited
Publication of WO2013114164A1 publication Critical patent/WO2013114164A1/en

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

Definitions

  • the invention describes a method for obtaining a glycoprotein composition with increased percentage of afucosylated glycoforms.
  • Protein glycosylation is one of the most important post-translation
  • N-linked glycosylation in which glycans are attached to the asparagine of the recognition sequence Asn-X-Thr/Ser, where "X" is any amino acid except proline
  • O-linked glycosylation in which glycans are attached to serine or threonine
  • N-linked glycans are of further two types - high mannose type consisting of two N-acetylglucosamines plus a large number of mannose residues (more than 4), and complex type that contain more than two N- acetylglucosamines plus any number of other types of sugars.
  • Endoplasmic Reticulum where a complex set of reactions result in the attachment of Glc 3 NAc 2 Man 9 (3 glucose, 2 N-acetylglucosamine and 9 mannose) to a carrier molecule called dolichol, that is then transferred to the appropriate point on the polypeptide chain (Schwarz F. and Aebi M., Current Opinion in Structural Biology, 2011, Vol.21, Issue 5, pages 576 to 582 and Burda P., and Aebi M., Biochemica et Biophysica acta (BBA) General Subjects, 1999, Volume 1426, Issue 2, pages 239 to 257).
  • Glc 3 NAc 2 Man 9 glucose, 2 N-acetylglucosamine and 9 mannose
  • the glycan complex so formed in the ER lumen is modified by action of enzymes in the Golgi apparatus. If the saccharide is relatively inaccessible, it is likely to stay in the original high-mannose form. If it is accessible, then many of the mannose residues may be cleaved off and the saccharide further modified, resulting in the complex type N-glycans structure.
  • mannosidase-1 may cleave/hydrolyze a high mannose glycan, while further on,fucosyltransferase FUT-8 fucosylates the glycan in the medial-Go ⁇ g ⁇ (Harue Imai-Nishiya et al, BMC
  • sugar composition as well as the structural configuration of a glycan structure depends on the protein being glycosylated, the cells/cell lines, the glycosylation machinery in the Endoplasmic Reticulum and the Golgi apparatus, the accessibility of the machinery enzymes to the glycan structure, the order of action of each enzyme and the stage at which the protein is released from the glycosylation machinery.
  • external factors may also affect the glycan structure and composition of a protein. These include the conditions in which the cell line expressing the protein is cultured, such as the medium composition, the composition and timing of the feed, osmolality, pH, temperature etc.
  • Pacisef a/ has shown that higher osmolality may result in increase in the number of Man5 residues on recombinant antibodies, with a simultaneous reduction in G 0 F and G-
  • reducing temperature can increase overall protein production by prolonging cell viability, which should, in principle, improve glycosylation (Moore A, Mercer J, Dutina G, Donahue CJ, Bauer KD,Mather JP, Etchverry T, Ryll T. Cytotechnology, 1997, Vol.23, pages 47 to 54).
  • Borys et al has shown that a deviation from optimum pH results in decrease in the expression rate as well as the extent of glycosylation of proteins ⁇ Borys M.C., Linzer, D.I.H., Papoutsakis 1993, ⁇ /technology, Vol.11, pages 720 to 724).
  • the culture pH of a hybridoma cell line has been shown to affect the resulting galactosylation and sialylation of the monoclonal antibodies (Muthing J, Kemminer SE, Conradt HS, Sagi D, Nimtz M, Karst U, Peter- Katalinic J., BiotechnolBioeng. 2003, Vol.83, Issue 3, pages 321 to 334).
  • rMAb therapeutic monoclonal antibodies
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Fucose-deficient IgGI s have shown a significant enhancement of ADCC up to 100-fold (Mori K., Cytotechnology, 2007, Vol. 55(2-3), pages 109 to 114 and Shields RL, The Journal of Biological Chemistry, 2002, Vol. 277(30), pages 26733 to 26740).
  • non-fucosylated antibodies may be the promising next-generation therapeutic antibodies with improved efficacy and reduced dose based toxicities.
  • the present invention describes a process of obtaining an antibody
  • composition comprising an increased percentage of afucosylated glycans.
  • the invention describes a cell culture process wherein the cells are subjected to simultaneous temperature downshift and pH upshift to obtain an antibody composition having increased percentage of afucosylated glycoform.
  • the invention describes a process for obtaining a glycoform composition comprising increased percentage of afucosylated glycoforms.
  • the method describes culturing cells at a temperature and pH for a period of time followed by subjecting cells to a second temperature and pH to obtain a glycoform composition having increased percentage of afucosylated glycans.
  • FIG. 1 is an illustration of effect of pH upshift and temperature downshift on the viable cell count during cell culture process, as described in examples 1 to 4.
  • FIG. 2 is an illustration of effect of pH upshift and temperature downshift on afucosylation content of antibodies obtained by cell culture process, as described in examples 1 to 4.
  • glycoprotein refers to protein or polypeptide having at least one glycan moiety wherein glycan refers to a monosaccharide or polysaccharide moeity. Thus, any polypeptide attached to a saccharide moiety is termed as glycoprotein.
  • glycoprotein refers to a monosaccharide or polysaccharide moiety.
  • glycoform or "glycovariant” have been used interchangeably herein, and refers to various oligosaccharide entities or moieties linked in their entirety to the Asparagine 297 (as per Kabat numbering) of the human Fc region of the glycoprotein in question, co translationally or post translationally within a host cell.
  • the glycan moieties that may be added during protein glycosylation include M3, M4, M5-8, M3NAG etc. Examples of such glycans and their structures are listed in Table 1 . However, Table 1 may in no way be considered to limit the scope of this invention to these glycans.
  • glycoform composition or distribution as used herein pertains to the quantity or percentage of different glycoforms present in a glycoprotein.
  • complex glycovariant as used herein consists of glycan moieties comprising any number of sugars.
  • Total afucosylated glycans described here, consists of glycan moieties wherein fucose is not linked to the non reducing end of N-acetlyglucosamine.
  • afucosylated glycans include GO, G1 A, G1 B, G2, M3- M9NAG, M3-M9.
  • osmolality as used herein is defined as a measure of the osmoles of solute per kilogram of solvent (osmol/kg) and may include ionized or non-ionized molecules. The osmolality may change during the cell culture process for e.g. by addition of feed, salts, additives or metabolites.
  • temperature shift as used herein is defined as the change in temperature during the cell culture process. For the purpose of this invention, the initial temperature of the cell culture process is higher than the final temperature i.e. cells are subjected to a temperature downshift wherein cells are first cultured at a higher temperature for certain time period after
  • Table I Representative table of various glycans
  • pH shift is defined as the change in pH during the cell culture process. As used herein, cells are first cultured at a pH for a certain period of time after which pH of the cell culture medium is increased, and cells are cultured at this increased pH for a certain period of time.
  • IVCC or “Integral viable cell concentration” refers to cell growth over time or integral of viable cells with respect to culture time that is used for calibration of specific protein production.
  • the integral of viable cell concentration can be increased either by increasing the viable cell concentration or by lengthening the process time.
  • the viable cell concentration or cell viability is defined as number of live cells in the total cell population.
  • the present invention discloses a cell culture process wherein cells are maintained at a particular pH and temperature to attain optimum growth, followed by decrease in temperature to a lower value, while pH is increased to a higher value to obtain a glycoform composition comprising increased percentage of afucosylated glycoforms.
  • the present invention provides a process for obtaining a glycoprotein composition comprising increased percentage of total afucosylated glycans comprising, culturing cells expressing said glycoprotein a) at a first temperature and a first pH, for a first period of time, followed by b) subjecting cells to a second temperature and second pH, for second period of time wherein the temperature and pH shift are carried out simultaneously.
  • the shift in temperature and pH may be accompanied by addition of nutrient feed, and further wherein the shift in temperature is towards lower value while shift in pH is towards higher values.
  • the glycoprotein comprises about 10.0% to about 22.0 % total afucosylated glycans.
  • the glycoprotein comprises about 10.0 %total afucosylated glycans.
  • the glycoprotein comprises about 13.0 % total afucosylated glycans.
  • the glycoprotein comprises about 15.0 % total afucosylated glycans. In yet another embodiment the glycoprotein comprises about 22.0 % total afucosylated glycans.
  • the application provides method for production of glycoproteins with a particular glycoform composition by first culturing cells at temperatures about 35°C -37°C and pH about 7.05, followed by lowering of temperature by about 2-7°C and increasing pH to about 7.2.
  • the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 35 °C and pH about 7.2. In yet another embodiment, the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 33 °C and pH about 7.2.
  • the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 31 °C and pH about 7.2.
  • the cell culture media that are useful in the application include but are not limited to, the commercially available products PF CHO (HyClone ® ), PowerCHO ® 2 (Lonza), Zap-CHO (Invitria), CD CHO, CDOptiCHOTM and CHO-S-SFMII
  • DMEM/F12 Invitrogen
  • Ham's F10 Ham's F10
  • Minimal Essential Media Sigma
  • RPMI -1 640 Sigma
  • the feeds in the present invention may be added in a continuous, profile or a bolus manner. Also it may be that one or more feeds are in one mode (e.g. profile mode) and others are in second mode (e.g. bolus or continuous mode). Further, the feed may be composed of nutrients or other medium components that have been depleted or metabolized by the cells. It may include hormones, growth factors, ions, vitamins, nucleoside, nucleotides, trace elements, amino acids, lipids or glucose. These supplementary components may be added at one time or in series of additions to replenish. Thus feed can be a solution of depleted nutrient(s), mixture of nutrient(s) or a mixture of cell culture medium/feed providing such nutrient(s). In one aspect of the invention, specific commercial feeds may be used while in other concentrated basal media may be added to the cell culture medium.
  • An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference.
  • rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone ® , Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05.
  • the cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone ® ) is added and temperature is lowered to 35 Q C.An additional feed is added at an IVCC of 10 million cells/mL.
  • the culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early.
  • the antibody titer at the time of harvest has been shown in Table 2.
  • the VCC values and afucosylation content are disclosed in Figures (FIG.) 1 and 2.
  • An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference.
  • rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone ® , Catalog no: SH30335 & SH30334) comprising galactose (6g/L) at 37C and pH 7.05.
  • the cells are cultured to attain VCC of -2.0 million cells/ml after which 75ml/L Cell Boost 2 (HyClone ® ) is added,temperature is lowered to 35°C and pH of the culture medium is raised to 7.2.
  • the culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early.
  • the resulting antibody (II) titre at the time of harvest is disclosed in Table 2.
  • the VCC values and afucosylation content are shown in FIG. 1 and
  • An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference.
  • rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone ® , Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05.
  • the cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone ® ) is added, temperature is lowered to 33°C and pH of the culture medium is raised to 7.2.
  • the culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early.
  • the resulting antibody (III) titre at the time of harvest has been shown in Table 2.
  • the VCC values and afucosylation content are disclosed in FIG.
  • Example IV An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference. rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone ® , Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05. The cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone ® ) is added, temperature is lowered to 31 °C and pH of the culture medium is raised to 7.2. The culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early. The resulting antibody (IV) titre at the time of harvest has been shown in Table 2. The VCC values and afucosylation content are disclosed in FIG. 1 and 2.

Abstract

The invention describes a method for obtaining a glycoprotein composition with increased percentage of afucosylated glycoforms.

Description

METHOD FOR OBTAINING GLYCOPROTEIN COMPOSITION WITH INCREASED AFUCOS YLATION CONTENT
RELATED APPLICATION
This application is related to and takes priority from the Indian Provisional Application 335/CHE/2012 filed 30 Jan 2012 and is incorporated herein in its entirety.
INTRODUCTION
The invention describes a method for obtaining a glycoprotein composition with increased percentage of afucosylated glycoforms.
Protein glycosylation is one of the most important post-translation
modifications associated with the eukaryotic proteins. The two major types of glycosylations in eukaryotic cells are N-linked glycosylation, in which glycans are attached to the asparagine of the recognition sequence Asn-X-Thr/Ser, where "X" is any amino acid except proline, and O-linked glycosylation in which glycans are attached to serine or threonine. N-linked glycans are of further two types - high mannose type consisting of two N-acetylglucosamines plus a large number of mannose residues (more than 4), and complex type that contain more than two N- acetylglucosamines plus any number of other types of sugars. In both N- and O- glycosylation, there is usually a range of glycan structures associated with each site (microheterogeneity). Macroheterogeneity results from the fact that not all N-glycan or O- glycan consensus sequences (Asn-X-Ser/Thr for N-glycan and serine or threonine for O-glycan present in the glycoproteins) may actually be glycosylated. This may be a consequence of the competitive action of diverse enzymes during biosynthesis and are key to understanding glycoprotein heterogeneity (Marino K. et ai, Nature Chemical Biology, 2010, Vol. 6, No. 10, pages 713 to723).
The process of N-linked glycosylation begins co-translationally in the
Endoplasmic Reticulum (ER) where a complex set of reactions result in the attachment of Glc3NAc2Man9 (3 glucose, 2 N-acetylglucosamine and 9 mannose) to a carrier molecule called dolichol, that is then transferred to the appropriate point on the polypeptide chain (Schwarz F. and Aebi M., Current Opinion in Structural Biology, 2011, Vol.21, Issue 5, pages 576 to 582 and Burda P., and Aebi M., Biochemica et Biophysica acta (BBA) General Subjects, 1999, Volume 1426, Issue 2, pages 239 to 257).
The glycan complex so formed in the ER lumen is modified by action of enzymes in the Golgi apparatus. If the saccharide is relatively inaccessible, it is likely to stay in the original high-mannose form. If it is accessible, then many of the mannose residues may be cleaved off and the saccharide further modified, resulting in the complex type N-glycans structure. In the c/'s-Golgi, mannosidase-1 may cleave/hydrolyze a high mannose glycan, while further on,fucosyltransferase FUT-8 fucosylates the glycan in the medial-Go\g\ (Harue Imai-Nishiya et al, BMC
Biotechnology, 2007, 7:84).
Thus the sugar composition as well as the structural configuration of a glycan structure depends on the protein being glycosylated, the cells/cell lines, the glycosylation machinery in the Endoplasmic Reticulum and the Golgi apparatus, the accessibility of the machinery enzymes to the glycan structure, the order of action of each enzyme and the stage at which the protein is released from the glycosylation machinery.
In addition to the "in vivo" factors listed above, "external factors" may also affect the glycan structure and composition of a protein. These include the conditions in which the cell line expressing the protein is cultured, such as the medium composition, the composition and timing of the feed, osmolality, pH, temperature etc.
There is a significant variability observed in terminal galactosylation which is found to be dependent on the medium. It has been shown that feeding cultures with galactose up to a concentration 36mM results in high levels of galactosylation (Davies, J.; Jiang, L; Pan, L.Z.; LaBarre, M.J. ; Anderson, D.; Reff, M.
Biotechnol.Bioeng.2001 , Vol.74, Issue 4 pages 288 to 294). However in a separate study it was shown that galactose feeding does not affect the sialic acid content.
Pacisef a/ has shown that higher osmolality may result in increase in the number of Man5 residues on recombinant antibodies, with a simultaneous reduction in G0F and G-| F glycoforms, resulting in its faster clearance from the body and thereby reducing its efficacy (Pads E., Yu, M., Autsen, J., Bayer, R., Li F., Bitechnol. Bioeng., 2011, Vol. 108, IssueW, pages 2348 to 2358). The studies by Kaufman et al and Yoon et a/ show a reduction in protein sialylation upon decrease in temperature {Kaufman, H., Mazur X., Fussenegger, M., Bailey, J.E., Biotechnology and Bioengineering., 1999, Vol. 63, Issue 5, pages 573 to 582;Trummer, E., Fauland, K., et.al, Biotechnol. Bioeng., 2006, Vol. 94, Issue 6, pages 1045 to 1052 and Yoon S.K., Song, J. Y., Lee, G.M., Biotechnol. Bioeng., 2003, Vol. 82, Issue 3, pages 289 to 298). Further, reducing temperature can increase overall protein production by prolonging cell viability, which should, in principle, improve glycosylation (Moore A, Mercer J, Dutina G, Donahue CJ, Bauer KD,Mather JP, Etchverry T, Ryll T. Cytotechnology, 1997, Vol.23, pages 47 to 54). Likewise, Borys et al has shown that a deviation from optimum pH results in decrease in the expression rate as well as the extent of glycosylation of proteins {Borys M.C., Linzer, D.I.H., Papoutsakis 1993, ΒΙΟ/technology, Vol.11, pages 720 to 724). The culture pH of a hybridoma cell line has been shown to affect the resulting galactosylation and sialylation of the monoclonal antibodies (Muthing J, Kemminer SE, Conradt HS, Sagi D, Nimtz M, Karst U, Peter- Katalinic J., BiotechnolBioeng. 2003, Vol.83, Issue 3, pages 321 to 334).
Most of the therapeutic monoclonal antibodies (rMAb) that have been licensed and developed as medical agents are of the human lgG1 isotype. The effectiveness of these antibodies is dependent on sensitization of target cells for subsequent killing by antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) mediated through the interaction of the Fc with FcyRs receptors or complement respectively (Jefferis, R. Expert Opinion on Biological Therapy, 2007, Vol.7, Number 9, pages 1401 to 1413; Shields, R. L. et al, The Journal of Biological Chemistry, 2002, Vol.277 (30), pages 26733 to 26740 ;Shinkawa, T. et al. The Journal of Biological Chemistry, 2003, Vol.278 (5), pages 3466 to 3473; Niwa, R. et al., Journal of Immunological Methods, 2005, Vol.306, Issues 1-2, pages 151 to 160; Ferrara, C. et al., Biotechnol. Bioeng., 2006, Vol. 93, Issue 5, pages 851 to 861 and Krapp, S. et al., J. Mol. Biol., 2003, Vol. 325, pages 979 to 989). Further, the binding to the Fcylll receptor is dependent on the fucose content of the Fc glycans wherein a reduction in fucose can increase effector function. Fucose-deficient IgGI s have shown a significant enhancement of ADCC up to 100-fold (Mori K., Cytotechnology, 2007, Vol. 55(2-3), pages 109 to 114 and Shields RL, The Journal of Biological Chemistry, 2002, Vol. 277(30), pages 26733 to 26740). Hence, non-fucosylated antibodies may be the promising next-generation therapeutic antibodies with improved efficacy and reduced dose based toxicities.
Robust stable production of completely non-fucosylated therapeutic antibodies consistent in quality has been achieved by the generation of a unique host cell line, in which the endogenous a-1 ,6-fucosyltransferase (FUT8) gene is knocked out. Further, antibodies with high oligomannose content having high affinity for FCYRI I IA, which is key to high ADCC activity have been generated using specific inhibitors for e.g. KifunesinefSfren, A., Ng, D., Joly, J., Snedecor, B., Lu, Y., Meng, G., Nakamura, G., and Krummen, L. Metabolic engineering to control glycosylation,in cell culture and upstream processing (edM.butler)2007, Taylor and Francis group) .Antibodies with the altered glycoform profiles have been produced using glycosylation enzyme mutants/knockouts or by using glycosylation inhibitors in cell culture medium such as deoxymannojirimycin J Bischoff J., Liscum L., and Kornfeld R., The Journal of Biological Chemistry, 1986, Vol. 261(10), pages 4766 to
4774 .However, the prior art does not describe a cell culture process or modifications in process parameters to modify afucosylated glycan content in a glycoprotein composition.
The present invention describes a process of obtaining an antibody
composition comprising an increased percentage of afucosylated glycans. In particular the invention describes a cell culture process wherein the cells are subjected to simultaneous temperature downshift and pH upshift to obtain an antibody composition having increased percentage of afucosylated glycoform.
SUMMARY
The invention describes a process for obtaining a glycoform composition comprising increased percentage of afucosylated glycoforms. In particular the method describes culturing cells at a temperature and pH for a period of time followed by subjecting cells to a second temperature and pH to obtain a glycoform composition having increased percentage of afucosylated glycans.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 is an illustration of effect of pH upshift and temperature downshift on the viable cell count during cell culture process, as described in examples 1 to 4. FIG. 2 is an illustration of effect of pH upshift and temperature downshift on afucosylation content of antibodies obtained by cell culture process, as described in examples 1 to 4. DETAILED DESCRIPTION OF THE INVENTION
Definitions
The term "glycoprotein" refers to protein or polypeptide having at least one glycan moiety wherein glycan refers to a monosaccharide or polysaccharide moeity. Thus, any polypeptide attached to a saccharide moiety is termed as glycoprotein. The term "glycan" refers to a monosaccharide or polysaccharide moiety.
The term "glycoform" or "glycovariant" have been used interchangeably herein, and refers to various oligosaccharide entities or moieties linked in their entirety to the Asparagine 297 (as per Kabat numbering) of the human Fc region of the glycoprotein in question, co translationally or post translationally within a host cell. The glycan moieties that may be added during protein glycosylation include M3, M4, M5-8, M3NAG etc. Examples of such glycans and their structures are listed in Table 1 . However, Table 1 may in no way be considered to limit the scope of this invention to these glycans.
The "glycoform composition" or distribution as used herein pertains to the quantity or percentage of different glycoforms present in a glycoprotein.
The "complex glycovariant" as used herein consists of glycan moieties comprising any number of sugars.
"Total afucosylated glycans" described here, consists of glycan moieties wherein fucose is not linked to the non reducing end of N-acetlyglucosamine.
Without limitation examples of afucosylated glycans include GO, G1 A, G1 B, G2, M3- M9NAG, M3-M9.
The term "osmolality" as used herein is defined as a measure of the osmoles of solute per kilogram of solvent (osmol/kg) and may include ionized or non-ionized molecules. The osmolality may change during the cell culture process for e.g. by addition of feed, salts, additives or metabolites. The term "temperature shift" as used herein is defined as the change in temperature during the cell culture process. For the purpose of this invention, the initial temperature of the cell culture process is higher than the final temperature i.e. cells are subjected to a temperature downshift wherein cells are first cultured at a higher temperature for certain time period after
Table I : Representative table of various glycans
Figure imgf000007_0001
Figure imgf000008_0001
&-A8 ia ijl -
which temperature is reduced, and cells are cultured at this lower temperature for a fixed period of time.
The term "pH shift" as used herein is defined as the change in pH during the cell culture process. As used herein, cells are first cultured at a pH for a certain period of time after which pH of the cell culture medium is increased, and cells are cultured at this increased pH for a certain period of time.
As used herein, "IVCC" or "Integral viable cell concentration" refers to cell growth over time or integral of viable cells with respect to culture time that is used for calibration of specific protein production. The integral of viable cell concentration can be increased either by increasing the viable cell concentration or by lengthening the process time.
The viable cell concentration or cell viability is defined as number of live cells in the total cell population. The present invention discloses a cell culture process wherein cells are maintained at a particular pH and temperature to attain optimum growth, followed by decrease in temperature to a lower value, while pH is increased to a higher value to obtain a glycoform composition comprising increased percentage of afucosylated glycoforms. In one embodiment, the present invention provides a process for obtaining a glycoprotein composition comprising increased percentage of total afucosylated glycans comprising, culturing cells expressing said glycoprotein a) at a first temperature and a first pH, for a first period of time, followed by b) subjecting cells to a second temperature and second pH, for second period of time wherein the temperature and pH shift are carried out simultaneously.
The shift in temperature and pH may be accompanied by addition of nutrient feed, and further wherein the shift in temperature is towards lower value while shift in pH is towards higher values.
In an embodiment the glycoprotein comprises about 10.0% to about 22.0 % total afucosylated glycans.
In another embodiment the glycoprotein comprises about 10.0 %total afucosylated glycans.
In yet another embodiment the glycoprotein comprises about 13.0 % total afucosylated glycans.
In yet another embodiment the glycoprotein comprises about 15.0 % total afucosylated glycans. In yet another embodiment the glycoprotein comprises about 22.0 % total afucosylated glycans.
Various methods described in the art such as Wuhreret. al., Ruhaak L.R., and Geoffrey et. al. can be used for assessing glycovariants present in a glycoprotein composition (Wuhrer M. et ai, Journal of Chromatography B, 2005, Vol.825, Issue 2, pages 124-13;, Ruhaak L.R., Anal Bioanal Chem., 2010, Vol.397. -3457-3481 andGeoffrey, R. G. et. al. Analytical Biochemistry 1996, Vol. 240, pages 210-226).
In another embodiment, the application provides method for production of glycoproteins with a particular glycoform composition by first culturing cells at temperatures about 35°C -37°C and pH about 7.05, followed by lowering of temperature by about 2-7°C and increasing pH to about 7.2.
In yet another embodiment, the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 35 °C and pH about 7.2. In yet another embodiment, the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 33 °C and pH about 7.2.
In yet another embodiment, the application provides methods for expression of protein with particular glycoform composition by growing cells at about 37°C and pH about 7.05, followed by subjecting cells to about 31 °C and pH about 7.2.
The cell culture media that are useful in the application include but are not limited to, the commercially available products PF CHO (HyClone®), PowerCHO® 2 (Lonza), Zap-CHO (Invitria), CD CHO, CDOptiCHO™ and CHO-S-SFMII
(Invitrogen), ProCHO™ (Lonza), CDM4CHO™ (Hyclone), DMEM (Invitrogen),
DMEM/F12 (Invitrogen), Ham's F10 (Sigma), Minimal Essential Media (Sigma), and RPMI -1 640 (Sigma).
The feeds in the present invention may be added in a continuous, profile or a bolus manner. Also it may be that one or more feeds are in one mode (e.g. profile mode) and others are in second mode (e.g. bolus or continuous mode). Further, the feed may be composed of nutrients or other medium components that have been depleted or metabolized by the cells. It may include hormones, growth factors, ions, vitamins, nucleoside, nucleotides, trace elements, amino acids, lipids or glucose. These supplementary components may be added at one time or in series of additions to replenish. Thus feed can be a solution of depleted nutrient(s), mixture of nutrient(s) or a mixture of cell culture medium/feed providing such nutrient(s). In one aspect of the invention, specific commercial feeds may be used while in other concentrated basal media may be added to the cell culture medium.
Certain aspects and embodiments of the invention are more fully defined by reference to the following examples. These examples should not, however, be construed as limiting the scope of the invention.
Example I
An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference. rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone®, Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05. The cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone®) is added and temperature is lowered to 35QC.An additional feed is added at an IVCC of 10 million cells/mL.The culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early. The antibody titer at the time of harvest has been shown in Table 2. The VCC values and afucosylation content are disclosed in Figures (FIG.) 1 and 2.
Example II
An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference. rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone®, Catalog no: SH30335 & SH30334) comprising galactose (6g/L) at 37C and pH 7.05. The cells are cultured to attain VCC of -2.0 million cells/ml after which 75ml/L Cell Boost 2 (HyClone®) is added,temperature is lowered to 35°C and pH of the culture medium is raised to 7.2. The culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early. The resulting antibody (II) titre at the time of harvest is disclosed in Table 2. The VCC values and afucosylation content are shown in FIG. 1 and 2.
Example III
An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference. rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone®, Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05. The cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone®) is added, temperature is lowered to 33°C and pH of the culture medium is raised to 7.2. The culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early. The resulting antibody (III) titre at the time of harvest has been shown in Table 2. The VCC values and afucosylation content are disclosed in FIG. 1 and 2.
Example IV An anti-CD20 antibody was cloned and expressed in a CHO cell line as described in U.S. Patent No. 7381560 which is incorporated herein by reference. rCHO cells expressing antibody at a seeding density of 0.8-1 .0 million cells/ml are seeded in PF CHO (HyClone®, Catalog no: SH30335 & SH30334) comprising galactose (6 g/L) at 37°C and pH 7.05. The cells are cultured to attain VCC of -2.0 million cells/ml, at which 75ml/L Cell Boost 2 (HyClone®) is added, temperature is lowered to 31 °C and pH of the culture medium is raised to 7.2. The culture is harvested after 180 - 288 hrs or at greater than 50% viability whichever is early. The resulting antibody (IV) titre at the time of harvest has been shown in Table 2. The VCC values and afucosylation content are disclosed in FIG. 1 and 2.
Table 2: Antibody titre at the time of harvest (180-288 hrs)
Figure imgf000012_0001

Claims

We claim
A process for obtaining a glycoprotein composition comprising increased percentage of total afucosylated glycans comprising, culturing cells expressing said glycoprotein at a first temperature and a first pH, for a first period of time, followed by subjecting cells to a second temperature and second pH, for a second period of time wherein the temperature shift and pH shift are carried out simultaneously.
A process according to claim 1 , wherein the said glycoprotein comprises about 10.0% to about 22.0% total afucosylated glycans.
A process according to claim 1 , wherein the said glycoprotein comprises about 10.0% total afucosylated glycans.
A process according to claim 1 , wherein the said glycoprotein comprises about 13.0% total afucosylated glycans.
A process according to claim 1 , wherein the said glycoprotein comprises about 15.0% total afucosylated glycans.
A process according to claim 1 , wherein the said glycoprotein comprises about 22.0% total afucosylated glycans.
A process according to claim 1 , wherein the cells are cultured in step a) at temperature of about 35 °C to about 37°C.
A process according to claim 1 , wherein the cells are cultured in step a) at temperature of about 37°C.
A process according to claim 1 , wherein cell are cultured in step a) at a pH of about 7.05.
A process according to claim 1 , wherein cells are cultured in step b) at a temperature of about 31 °C to about 35 °C. 1 1 ) A process according to claim 1 , wherein cells are cultured in step b) at a temperature of about 35 °C.
12) A process according to claim 1 , wherein cells are cultured in step b) at a temperature of about 33 °C.
13) A process according to claim 1 , wherein cells are cultured in step b) at a temperature of about 31 °C.
14) A process according to claim 1 , wherein cells are cultured in step b) at a pH of about 7.2.
15) A process according to claim 1 , wherein step b) further comprises addition of a feed.
PCT/IB2012/057090 2012-01-30 2012-12-08 Method for obtaining glycoprotein composition with increased afucosylation content WO2013114164A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN335CH2012 2012-01-30
IN335/CHE/2012 2012-01-30

Publications (1)

Publication Number Publication Date
WO2013114164A1 true WO2013114164A1 (en) 2013-08-08

Family

ID=48904473

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2012/057090 WO2013114164A1 (en) 2012-01-30 2012-12-08 Method for obtaining glycoprotein composition with increased afucosylation content

Country Status (1)

Country Link
WO (1) WO2013114164A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016089919A1 (en) * 2014-12-01 2016-06-09 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
WO2017120359A1 (en) * 2016-01-06 2017-07-13 Oncobiologics, Inc. Reduction of high molecular weight species, acidic charge species, and fragments in a monoclonal antibody composition
WO2018170099A1 (en) * 2017-03-14 2018-09-20 Amgen Inc. Control of total afucosylated glycoforms of antibodies produced in cell culture
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
WO2019236739A1 (en) 2018-06-05 2019-12-12 Amgen Inc. Modulating antibody dependent cellular phagocytosis
WO2020055900A1 (en) 2018-09-11 2020-03-19 Amgen Inc. Methods of modulating antibody-dependent cell-mediated cytotoxicity
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
CN111406112A (en) * 2017-11-30 2020-07-10 豪夫迈·罗氏有限公司 Process for culturing mammalian cells
WO2020227726A1 (en) 2019-05-06 2020-11-12 Amgen Inc. Modulating antibody effector functions
WO2021062372A1 (en) 2019-09-26 2021-04-01 Amgen Inc. Methods of producing antibody compositions
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
WO2022081824A1 (en) 2020-10-15 2022-04-21 Amgen Inc. Relative unpaired glycans in antibody production methods

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008015339A2 (en) * 2006-08-01 2008-02-07 Lfb Biotechnologies Recombinant factor vii composition
WO2008063776A2 (en) * 2006-10-12 2008-05-29 Genentech, Inc. Antibodies to lymphotoxin-alpha

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008015339A2 (en) * 2006-08-01 2008-02-07 Lfb Biotechnologies Recombinant factor vii composition
WO2008063776A2 (en) * 2006-10-12 2008-05-29 Genentech, Inc. Antibodies to lymphotoxin-alpha

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10376582B2 (en) 2013-10-16 2019-08-13 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
AU2015355087B2 (en) * 2014-12-01 2020-04-09 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
EA036178B1 (en) * 2014-12-01 2020-10-09 Эмджен Инк. Process for manipulating the level of glycan content of a glycoprotein
EP3680344A1 (en) * 2014-12-01 2020-07-15 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
JP7066775B2 (en) 2014-12-01 2022-05-13 アムジエン・インコーポレーテツド Process for manipulating levels of glycoprotein glycan content
JP2017538446A (en) * 2014-12-01 2017-12-28 アムジエン・インコーポレーテツド Process for manipulating the level of glycan content of glycoproteins
US10167492B2 (en) 2014-12-01 2019-01-01 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
CN107109455A (en) * 2014-12-01 2017-08-29 美国安进公司 For the method for the glycan content level for manipulating glycoprotein
KR20170083636A (en) * 2014-12-01 2017-07-18 암젠 인크 Process for manipulating the level of glycan content of a glycoprotein
US10822630B2 (en) 2014-12-01 2020-11-03 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
EP3227454B1 (en) 2014-12-01 2020-01-29 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
JP2020124216A (en) * 2014-12-01 2020-08-20 アムジエン・インコーポレーテツド Processes for manipulating levels of glycan content of glycoproteins
WO2016089919A1 (en) * 2014-12-01 2016-06-09 Amgen Inc. Process for manipulating the level of glycan content of a glycoprotein
KR102623965B1 (en) 2014-12-01 2024-01-11 암젠 인크 Process for manipulating the level of glycan content of a glycoprotein
US10696735B2 (en) 2015-01-21 2020-06-30 Outlook Therapeutics, Inc. Modulation of charge variants in a monoclonal antibody composition
WO2017120359A1 (en) * 2016-01-06 2017-07-13 Oncobiologics, Inc. Reduction of high molecular weight species, acidic charge species, and fragments in a monoclonal antibody composition
US11285210B2 (en) 2016-02-03 2022-03-29 Outlook Therapeutics, Inc. Buffer formulations for enhanced antibody stability
CN110418846A (en) * 2017-03-14 2019-11-05 美国安进公司 The control for always going fucosylation sugar-type of the antibody generated in cell culture
WO2018170099A1 (en) * 2017-03-14 2018-09-20 Amgen Inc. Control of total afucosylated glycoforms of antibodies produced in cell culture
AU2018235928B2 (en) * 2017-03-14 2023-09-21 Amgen Inc. Control of total afucosylated glycoforms of antibodies produced in cell culture
JP2020513813A (en) * 2017-03-14 2020-05-21 アムジエン・インコーポレーテツド Regulation of total nonfucosylated glycoforms of antibodies produced in cell culture
CN111406112A (en) * 2017-11-30 2020-07-10 豪夫迈·罗氏有限公司 Process for culturing mammalian cells
WO2019236739A1 (en) 2018-06-05 2019-12-12 Amgen Inc. Modulating antibody dependent cellular phagocytosis
WO2020055900A1 (en) 2018-09-11 2020-03-19 Amgen Inc. Methods of modulating antibody-dependent cell-mediated cytotoxicity
WO2020227726A1 (en) 2019-05-06 2020-11-12 Amgen Inc. Modulating antibody effector functions
WO2021062372A1 (en) 2019-09-26 2021-04-01 Amgen Inc. Methods of producing antibody compositions
WO2022081824A1 (en) 2020-10-15 2022-04-21 Amgen Inc. Relative unpaired glycans in antibody production methods

Similar Documents

Publication Publication Date Title
WO2013114164A1 (en) Method for obtaining glycoprotein composition with increased afucosylation content
US10059770B2 (en) Process of modulating man5 and/or afucosylation content of a glycoprotein composition
Costa et al. Glycosylation: impact, control and improvement during therapeutic protein production
Wang et al. Antibody glycoengineering strategies in mammalian cells
Liu et al. The availability of glucose to CHO cells affects the intracellular lipid-linked oligosaccharide distribution, site occupancy and the N-glycosylation profile of a monoclonal antibody
Durocher et al. Expression systems for therapeutic glycoprotein production
Batra et al. Glycosylation of monoclonal antibody products: Current status and future prospects
WO2013114167A1 (en) Process of obtaining glycoform composition
EP1792987A1 (en) Method of producing glycoprotein composition
Konno et al. Fucose content of monoclonal antibodies can be controlled by culture medium osmolality for high antibody-dependent cellular cytotoxicity
WO2013114165A1 (en) Process of obtaining glycoprotein composition with increased afucosylation content
Blondeel et al. Supplementing glycosylation: A review of applying nucleotide-sugar precursors to growth medium to affect therapeutic recombinant protein glycoform distributions
Mastrangeli et al. The formidable challenge of controlling high mannose-type N-glycans in therapeutic mAbs
WO2003085119A1 (en) METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
WO2015140700A1 (en) Cell culture process
Wang et al. Strategies for engineering protein N-glycosylation pathways in mammalian cells
EP3127917B1 (en) Method for preparing antibody through regulation of sugar content of antibody
WO2015128793A1 (en) A process for modifying high mannose and galactosylation content of a glycoprotein composition
JPWO2005035741A1 (en) Genome-modified cells
Donini et al. Glycoengineering Chinese hamster ovary cells: a short history
Seo et al. Effect of glucose feeding on the glycosylation quality of antibody produced by a human cell line, F2N78, in fed-batch culture
US10590455B2 (en) Process for modifying galactosylation and G0F content of a glycoprotein composition by glutamine supplementation
WO2005035740A1 (en) Genomically modified cell neutralized to serum-free system
Spearman et al. Glycosylation in cell culture
BR112021008458A2 (en) method for modifying the glycosylation profile of a recombinant glycoprotein produced in cell culture

Legal Events

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

Ref document number: 12867131

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12867131

Country of ref document: EP

Kind code of ref document: A1