US20190010531A1 - Cell culture process for making a glycoprotein - Google Patents

Cell culture process for making a glycoprotein Download PDF

Info

Publication number
US20190010531A1
US20190010531A1 US16/026,539 US201816026539A US2019010531A1 US 20190010531 A1 US20190010531 A1 US 20190010531A1 US 201816026539 A US201816026539 A US 201816026539A US 2019010531 A1 US2019010531 A1 US 2019010531A1
Authority
US
United States
Prior art keywords
glycoprotein
glycan
ornithine
putrescine
soy hydrolysate
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
US16/026,539
Other languages
English (en)
Inventor
John Chen
Shawn Lawrence
Amy Johnson
Theodore Loney
Ravindra Pangule
Ta-Chun Hang
Scott Carver
Bernhard Schilling
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.)
Regeneron Pharmaceuticals Inc
Original Assignee
Regeneron Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals Inc filed Critical Regeneron Pharmaceuticals Inc
Priority to US16/026,539 priority Critical patent/US20190010531A1/en
Assigned to REGENERON PHARMACEUTICALS, INC. reassignment REGENERON PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PANGULE, Ravindra, LAWRENCE, SHAWN, CARVER, Scott, HANG, Ta-Chun, JOHNSON, AMY, SCHILLING, BERNHARD, CHEN, JOHN, LONEY, Theodore
Publication of US20190010531A1 publication Critical patent/US20190010531A1/en
Priority to US16/742,723 priority patent/US20200131554A1/en
Priority to US16/864,689 priority patent/US20200255880A1/en
Priority to US17/409,158 priority patent/US20210388407A1/en
Priority to US17/409,193 priority patent/US20210388408A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • 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
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4705Regulators; Modulating activity stimulating, promoting or activating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • 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
    • C12N5/0681Cells of the genital tract; Non-germinal cells from gonads
    • C12N5/0682Cells of the female genital tract, e.g. endometrium; Non-germinal cells from ovaries, e.g. ovarian follicle 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
    • C12P21/06Preparation of peptides or proteins produced by the hydrolysis of a peptide bond, e.g. hydrolysate products
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"
    • 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
    • C12N2500/46Amines, e.g. putrescine

Definitions

  • the invention relates to methods for the culturing of cells and for the production of recombinant proteins.
  • the invention specifically relates to methods for the culturing of cells in soy hydrolysate-containing media to achieve consistent production of high quality recombinant protein.
  • Protein hydrolysates are commonly used in the production of recombinant proteins from cultured cells.
  • protein hydrolysates may contain compounds that negatively impact cell growth or recombinant protein production.
  • protein hydrolysates have been widely used as supplements in cell culture.
  • the present disclosure identifies certain components in plant protein hydrolysate (e.g., soy hydrolysate) that can vary from batch-to-batch and alter the composition and yield of high quality glycoproteins produced in culture using soy hydrolysate.
  • plant protein hydrolysate e.g., soy hydrolysate
  • the present disclosure addresses the need for improved cell culture methods by, among other things, screening batches of plant protein hydrolysate and selecting those batches that include a desirable concentration of a component of plant protein hydrolysate for use in the production of biopharmaceuticals.
  • the present disclosure is predicated in part on the discovery that the concentration of ornithine or putrescine in a batch of soy hydrolysate affects the quality and composition of proteins produced in cell culture using soy hydrolysate.
  • the present disclosure also provides that cells cultured in media including soy hydrolysate comprising certain concentrations of ornithine or putrescine produce greater amounts of high quality proteins exhibiting more consistent glycosylation patterns, amounts of glycan and sialic acid profiles from lot-to-lot.
  • the invention relates to a method of culturing a population of cells expressing a recombinant heterologous glycoprotein in cell culture media comprising soy hydrolysate to produce the recombinant heterologous glycoprotein, and wherein the soy hydrolysate comprises ⁇ 0.067% (w/w) ornithine or putrescine.
  • the method includes the steps of culturing a population of cells expressing a recombinant heterologous glycoprotein in cell culture media comprising soy hydrolysate containing ⁇ less than 0.67 milligram (mg) of ornithine per gram (g) of soy (w/w), or about 0.003%-0.067% (w/w) ornithine.
  • the culture media contains ⁇ 5 mg/L ornithine, or about 0.6-3 mg/L ornithine.
  • the population of cells is obtained by clonal expansion of a cell expressing a recombinant heterologous glycoprotein.
  • the invention relates to a method for producing a glycoprotein.
  • the method includes the steps of culturing a population of cells expressing a recombinant heterologous glycoprotein in culture media containing soy hydrolysate containing ⁇ less than 0.67 milligram (mg) of putrescine per gram (g) of soy (w/w), or about 0.003%-0.067% (w/w) putrescine.
  • the culture media contains ⁇ 5 mg/L putrescine, or about 0.6-3 mg/L putrescine.
  • the population of cells is obtained by clonal expansion of a cell expressing a recombinant heterologous glycoprotein.
  • the glycoprotein is a trap molecule, such as rilonacept (IL1-trap, disclosed, e.g., in U.S. Pat. No. 6,927,004), aflibercept (VEGF-trap, disclosed, e.g., in U.S. Pat. No. 7,087,411), conbercept (VEGF-trap, disclosed, e.g., in U.S. Pat. Nos. 7,750,138 and 8,216,575), and etanercept (TNF-trap, disclosed, e.g., in U.S. Pat. No. 5,610,279.)
  • ⁇ 10% (w/w) of the total amount of all N-glycan species of the glycoprotein is an A1 N-glycan.
  • the invention relates to a method of producing a glycoprotein. In another aspect, the invention relates to a method of using a soy hydrolysate in the producing of a glycoprotein. In another aspect, the invention relates to a method of selecting a soy hydrolysate for use in producing a glycoprotein by evaluating the quality of the produced glycoprotein.
  • the method comprises culturing a cell expressing a glycosylated protein in a cell culture media to produce the glycoprotein, purifying the glycosylated protein, subjecting the purified glycosylated protein to oligosaccharide fingerprint analysis, determining the relative amount of an A1 N-glycan compared to total amount of N-glycan species of the glycoprotein; and selecting a soy hydrolysate that provides for at least 10% (w/w) A1 N-glycan compared to total amount of N-glycan species of the glycoprotein.
  • the method includes the steps of preparing a cell culture media containing a soy hydrolysate, culturing a cell that expresses the glycoprotein in the cell culture media, purifying the glycosylated protein, subjecting the purified glycosylated protein to oligosaccharide fingerprint analysis, determining the relative amount of an A1 N-glycan compared to total amount of N-glycan species of the glycoprotein, and then selecting the soy hydrolysate that provides for the production of a glycoprotein with at least 10% (w/w) A1 N-glycan compared to total amount of N-glycan species of the glycoprotein.
  • the selected soy hydrolysate contains ⁇ 0.67 mg ornithine per g soy (w/w), or about 0.003%-0.067% (w/w) ornithine.
  • the culture media contains ⁇ 5 mg/L ornithine, or about 0.6-3 mg/L ornithine.
  • the selected soy hydrolysate contains ⁇ 0.67 mg putrescine per g soy (w/w), or about 0.003%-0.067% (w/w) putrescine.
  • the culture media contains ⁇ 5 mg/L putrescine, or about 0.6-3 mg/L putrescine.
  • the invention relates to a method of selecting a soy hydrolysate for use in producing a glycoprotein by measuring the amount of ornithine or putrescine in the soy hydrolysate.
  • the method includes the steps of measuring the amount of ornithine in a soy hydrolysate, selecting a soy hydrolysate with ⁇ 0.67 mg ornithine per g soy, or about 0.003%-0.067% (w/w) ornithine, and combining the selected soy hydrolysate with an additional ingredient to form a cell culture media with ⁇ 5 mg/L ornithine, or about 0.6-3 mg/L ornithine.
  • the method includes the steps of measuring the amount of putrescine in a potentially useful soy hydrolysate, selecting a soy hydrolysate with ⁇ 0.67 mg putrescine per g soy, or about 0.003%-0.067% (w/w) putrescine, and combining the selected soy hydrolysate with an additional ingredient to form a cell culture media with ⁇ 5 mg/L putrescine, or about 0.6-3 mg/L putrescine.
  • the invention relates to a glycoprotein comprising an A1 N-glycan and at least one other N-glycan species in which the relative amount of the A1 N-glycan is at least 10% (w/w) of the total amount of N-glycans of the glycoprotein is provided. In one embodiment, the relative amount of the A1 N-glycan is about 10%-17% (w/w).
  • the glycoprotein also has an A2 N-glycan, an A2F N-glycan, an A1F N-glycan, an NGA2F N-glycan, an NA2G1F N-glycan, an NA2 N-glycan, and an NA2F N-glycan.
  • the glycoprotein contains 8-65 moles of sialic acid per mole of glycoprotein.
  • any one of asparagine residues N37, N98, N418, and N511 of SEQ ID NO: 1 contains an A1 N-glycan.
  • any one of asparagine residues N123 and N196 of SEQ ID NO: 2 contains an A1 N-glycan.
  • the relative amount of the A1 N-glycan of the glycoprotein is determined by comparing the area under the peak of the A1 N-glycan to the total areas under the peak for all N-glycans obtained from an oligosaccharide fingerprint of the glycoprotein obtained by capillary electrophoresis.
  • a method of manufacturing soy hydrolysate having a reduced amount of ornithine or putrescine comprises the steps of enzymatically digesting soy extract in a residue-free reaction vessel, measuring the amount of ornithine in the soy hydrolysate, and selecting those lots of soy hydrolysate containing ⁇ 0.067% (w/w) ornithine or putrescine for use in a cell culture media.
  • the method comprises the steps of enzymatically digesting soy extract in a residue-free reaction vessel, measuring the amount of putrescine in the soy hydrolysate, and selecting soy hydrolysate with ⁇ 0.067% (w/w) ornithine or putrescine for use in a cell culture media.
  • FIG. 1 depicts a chromatographic elution profile of ninhydrin-derived amino acids.
  • the X-axis depicts time of elution from a chromatography column (retention time), and the Y-axis depicts light absorbance at 570 nm.
  • Panel A depicts batch that does not meet the criteria for producing an acceptable N-glycan mixture by FDA standards.
  • Panel B depicts an acceptable amino acid analysis of soy protein hydrolysate. The peak representing ornithine is circled in both chromatograms.
  • FIG. 2 depicts a capillary electrophoretogram of oligosaccharides released from a glycoprotein by peptide:N-glycosidase F (PNGase F) digestion.
  • the X-axis depicts time of elution from a capillary, and the Y-axis depicts light absorbance or fluorescence intensity. The peaks are numbered 1-21.
  • FIG. 3 depicts a dot blot of the relative amount of A1 N-glycan as a function of ornithine and citrulline concentration in soy protein hydrolysate.
  • the X-axis depicts the concentration of citrulline or ornithine in mg/L.
  • the Y-axis depicts the relative area of peak 11, which represents A1 N-glycan.
  • FIG. 4 is a correlation plot depicting the negative correlation of ornithine concentration in soy hydrolysate (lower right quadrant) to the relative amount of peak 11 in aflibercept (A1 N-glycan, upper left quadrant).
  • FIG. 5 is a correlation plot depicting (i) the negative correlation of ornithine concentration in soy hydrolysate (lower left quadrant) to the final titer of rilonacept (upper right quadrant); and (ii) the positive correlation of ornithine concentration in soy hydrolysate (lower left quadrant) to the accumulation of lactate in media (lower left quadrant).
  • FIG. 6A is a pair of graphs depicting the amount of polyamine synthesized from a CHO cell culture, depicted as either IVCD ⁇ 10 6 cell-day/ml or as titer (as grams per ml) as a function of batch day under various conditions including, control, high and low ornithine concentrations, putrescine, MFC and IPC.
  • FIG. 6B is a table providing the experimental conditions of each study group depicted in FIG. 6A .
  • relative amount means the amount of a molecular species over the total amount of all molecular species of a general type.
  • the relative amount of an A1 glycan i.e., (GlcNAc)2(Man)3(GlcNAc)2(Gal)2(SA)1 is calculated as amount of A1/sum of the amount of all N glycans.
  • the relative amount can be expresses as absolute mass-to-mass amounts (i.e. gram per gram) or a percentage i.e., % (w/w).
  • Ornithine is a non-protein coding amino acid involved in the urea cycle, polyamine synthesis and arginine metabolism. Ornithine is also known to influence glycoform content of recombinant proteins. See PCT/US2014/069378. Ornithine is acted on by several enzymes. For example, ornithine decarboxylase catalyzes the conversion of ornithine to putrescine in the polyamine biosynthetic pathway. See Pegg A, J. of Biol. Chem. (2006) 281:21 pp. 14532. Additionally, ornithine conversion to citrulline is catalyzed by ornithine transcarbamylase as part of the urea cycle.
  • Ornithine metabolism occurs in both cytosol and mitochondria of cells in culture.
  • the presence of putrescine or presence of ornithine has been deemed critical for growth and productivity of cells cultured in chemically defined media, however the impact on critical quality attributes of a protein produced by such cells has not been described.
  • “Putrescine” is a non-protein coding amino acid, a polyamine, involved in the urea cycle. Putrescine (also known as 1,4-Diaminobutane, having a chemical formula of C 4 H 12 N 2 ) is produced by the decarboxylation of ornithine and serves as a precursor to gamma-aminobutyrate ( ⁇ -aminobutyrate).
  • peptide As used herein “peptide”, “polypeptide” and “protein” are used interchangeably throughout and refer to a molecule comprising two or more amino acid residues joined to each other by a peptide bond. Peptides, polypeptides and proteins may also include modifications such as glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, alkylation, hydroxylation and ADP-ribosylation. Peptides, polypeptides, and proteins can be of scientific or commercial interest, including protein-based drugs (biotherapeutics). Peptides, polypeptides, and proteins include, among other things, antibodies and chimeric or fusion proteins. Peptides, polypeptides, and proteins can be produced by recombinant animal cell lines such as mammalian cell lines using cell culture methods.
  • the polynucleotide sequence may be a naturally occurring sequence that is introduced into an ectopic site within the host cell genome.
  • the peptide sequence may be heterologous, such as a naturally occurring sequence from another organism, a recombinant sequence, a genetically modified sequence, or inter alia a sequence expressed under the control of a different-than-wild type promoter, for example a nucleotide sequence encoding a human ortholog, whereby the host (production) cell is a CHO cell.
  • antigen-binding protein includes a protein that has at least one CDR and is capable of selectively recognizing an antigen, i.e., is capable of binding an antigen with a KD that is at least in the micromolar range.
  • Therapeutic antigen-binding proteins e.g., therapeutic antibodies
  • an antigen-binding protein includes two or more CDRs, e.g., 2, 3, 4, 5, or 6 CDRs.
  • antigen binding proteins include antibodies, antigen-binding fragments of antibodies such as polypeptides containing the variable regions of heavy chains and light chains of an antibody (e.g., Fab fragment, F(ab′)2 fragment), and proteins containing the variable regions of heavy chains and light chains of an antibody and containing additional amino acids from the constant regions of heavy and/or light chains (such as one or more constant domains, i.e., one or more of CL, CH1, hinge, CH2, and CH3 domains).
  • polypeptides containing the variable regions of heavy chains and light chains of an antibody e.g., Fab fragment, F(ab′)2 fragment
  • proteins containing the variable regions of heavy chains and light chains of an antibody and containing additional amino acids from the constant regions of heavy and/or light chains such as one or more constant domains, i.e., one or more of CL, CH1, hinge, CH2, and CH3 domains.
  • Antibody refers to an immunoglobulin molecule consisting of four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain has a heavy chain variable region (HCVR or VH) and a heavy chain constant region.
  • the heavy chain constant region contains three domains, CH1, CH2 and CH3.
  • Each light chain has a light chain variable region (VL) and a light chain constant region.
  • the light chain constant region consists of one domain (CL).
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from the amino-terminus to the carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • the term “antibody” includes both glycosylated and non-glycosylated immunoglobulins of any isotype or subclass.
  • the term “antibody” includes antibody molecules prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from a host cell transfected with a nucleotide sequence in order to express the antibody.
  • the term “antibody” also includes a bispecific antibody, which includes a heterotetrameric immunoglobulin that can bind to more than one epitope. Bispecific antibodies are generally described in U.S. Patent Application Publication No. 2010/0331527, which is incorporated by reference herein.
  • antigen-binding portion of an antibody (or antibody fragment) or a protein of interest refers to one or more fragments of an antibody or a protein of interest that retain the ability to specifically bind to an antigen.
  • protein binding fragments encompassed within the term “antigen-binding portion” of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., Nature (1989) 241:544-546), which consists of a VH domain, (vi) an isolated CDR, and (vii)
  • antibody forms of single chain antibodies, such as diabodies are also encompassed under the term “antibody”. See, e.g., Holliger et al., PNAS USA (1993) 90:6444-6448; Poljak et al., Structure (1994) 2:1121-1123.
  • an antibody or antigen-binding portion thereof may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides.
  • immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov et al., Human Antibodies and Hybridomas (1995) 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov et al. Mol. Immunol.
  • Antibody portions such as Fab and F(ab′)2 fragments, can be prepared from whole antibodies using conventional techniques, such as via papain or pepsin digestion of whole antibodies. Moreover, antibodies, antibody portions and immunoadhesion molecules can be obtained using standard recombinant DNA techniques commonly known in the art (see Sambrook et al., 1989).
  • human antibody is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences.
  • Human antibodies of the present disclosure may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and in particular CDR3.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor et al. Nucl. Acids Res. (1992) 20:6287-6295) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences.
  • Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences.
  • such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis), and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • Fc fusion proteins comprise part or all of two or more proteins, one of which is an Fc portion of an immunoglobulin molecule, which are not otherwise found together in nature. Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al., PNAS USA (1991) 88:10535; Byrn et al., Nature (1990) 344:677; and Hollenbaugh et al., Current Protocols in Immunology (1992) Suppl. 4, pp. 10.19.1-10.19.11.
  • Receptor Fe fusion proteins comprise one or more extracellular domain(s) of a receptor coupled to an Fc moiety, which in some embodiments comprises a hinge region followed by a CH2 and CH3 domain of an immunoglobulin.
  • the Fc-fusion protein contains two or more distinct receptor chains that bind to a one or more ligand(s).
  • an “Fc-fusion protein” is a “trap” molecule, which is a decoy receptor molecule that includes two distinct receptor components that mimic the binding domains of a corresponding endogenous receptor and the Fc portion of an antibody.
  • trap molecules include an IL-1 trap (e.g., rilonacept, which contains the IL-1RAcP ligand binding region fused to the IL-1R1 extracellular region which in turn is fused to the Fc of hIgG1) (e.g., SEQ ID NO:1) (see U.S. Pat. No.
  • a VEGF trap e.g., aflibercept, which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 which in turn is fused to Fc of hIgG1.
  • aflibercept which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 which in turn is fused to Fc of hIgG1.
  • TNF trap e.g., aflibercept, which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 which in turn is fused to Fc of hIgG1.
  • glycoproteins include the formation of glycoproteins where oligosaccharides are attached either to the side chain of an asparagine (Asn) residue (i.e., N-linked), or a serine (Ser) or threonine (Thr) residue (i.e., O-linked) of a protein.
  • Asn asparagine
  • Ser serine
  • Thr threonine
  • Glycoproteins include any protein that contains an O-linked glycan or an N-linked glycan. Glycans can be homo- or heteropolymers of monosaccharide residues, which can be linear or branched.
  • N-linked glycosylation is known to initiate primarily in the endoplasmic reticulum, whereas O-linked glycosylation is shown to initiate in either the ER or Golgi apparatus.
  • N-glycan is used interchangeably with “N-linked oligosaccharide.”
  • O-glycan is used interchangeably with “O-linked oligosaccharide.”
  • N-glycan protein includes proteins that contain or can accept N-linked oligosaccharides.
  • N-glycans can be composed of N-acetyl galactosamine (GalNAc), mannose (Man), fucose (Fuc), galactose (Gal), neuraminic acid (NANA), and other monosaccharides, however N-glycans usually have a common core pentasaccharide structure including: three mannose and two N-acetylglucosamine (GlcNAc) sugars.
  • Proteins with the consecutive amino acid sequence, Asn-X-Ser or Asn-X-Thr, where X is any amino acid except proline, can provide an attachment site for N-glycans.
  • N-glycans include those N-linked oligosaccharides listed in Table 1.
  • the shorthand designations of the listed oligosaccharides are used herein as simplified names to describe the oligosaccharide.
  • an A1 N-glycan contains an arginine linked to a oligosaccharide consisting of (SA)(Gal)2(GlcNAc)2(Man)3(GlcNAc)3.
  • Hydrolysates are complex materials derived from the hydrolysis of plant material, animal material, whey, yeast, and the like.
  • hydrolysate is used interchangeably with “protein hydrolysate”.
  • Plant hydrolysates plant protein hydrolysates
  • Protein hydrolysates can be manufactured by three general methods: acid hydrolysis, alkaline hydrolysis, and enzymatic hydrolysis. For biological applications, including biotherapeutic manufacturing, protein hydrolysates are mostly made by enzymatic hydrolysis.
  • soy hydrolysate made by pepsin digestion may be called “soy peptone”, or a yeast hydrolysate made by trpsin digestion may be called “yeast tryptone.”
  • soy peptone a soy hydrolysate made by pepsin digestion
  • yeast hydrolysate made by trpsin digestion may be called “yeast tryptone.”
  • Franek et al., Biotechnol. Prog. 16(5): 688-92 (2000) is incorporated herein for plant protein hydrolysates and methods of manufacturing them.
  • the subject hydrolysate is a plant hydrolysate.
  • the subject protein hydrolysate is a soy hydrolysate.
  • Soy hydrolysate is an enzymatically digested soy product derived from soybean grit, that is largely, chemically undefined. Generally, soy hydrolysate is composed of a conglomerate of amino acids, proteins, carbohydrates, minerals and vitamins.
  • Soy hydrolysate is a plant-derived protein hydrolysate that is commercially available, for example, in high concentration solution (e.g., HyCloneTM HyQ Soy Hydrolysate solution) or powder (e.g., Sigma Aldrich® S1674 (AmisoyTM), soy protein hydrolysate) form.
  • a “batch” or “lot” of soy hydrolysate, as used herein refers to a manufactured amount of soy hydrolysate resulting from hydrolysis of soybean grit. For example, each hydrolysis process can result in a unique “batch” or “lot” of soy hydrolysate with varying concentrations of components, such as vitamins, amino acids, peptides and sugars.
  • Soy hydrolysate is commonly used with animal protein-free cell culture medium for the growth of mammalian cell lines during the production of commercial biotherapeutics, such as antibodies. More specifically, soy hydrolysate is added to a cell culture media prior to or during inoculation with cells. The cells are then cultured in the hydrolysate-containing medium, until they are harvested. Due to the undefined nature of soy hydrolysate, batches of soy hydrolysate will vary from batch-to-batch (or lot-to-lot), which can lead to inconsistencies in commercial manufacturing of biotherapeutics.
  • the present disclosure has identified that concentrations of certain components in a batch of soy hydrolysate affect the quality and composition of proteins produced in cell culture using soy hydrolysate.
  • the present disclosure provides methods for screening batches of soy hydrolysate in order to select certain batches of soy hydrolysate that contain a desirable amount of a component such as, for example, ornithine, putrescine, citrulline, arginine or a combination thereof.
  • the screening method includes measuring the amount of ornithine or putrescine in at least a portion (i.e., a sample) of a batch of soy hydrolysate.
  • a soy hydrolysate sample is weighed and a portion thereof is dissolved to a desired concentration.
  • the soy hydrolysate solution is then diluted in a solvent to a second desired concentration (e.g., 1 g/L to 25 g/L) and the composition of the resulting soy hydrolysate solutions can then be determined.
  • the measuring step employs a suitable method for determining the molecular composition of a soy hydrolysate sample including, for example, colorimetric detection performed following post-column ninhydrin reaction, or chromatography f eluted ninhydrin-positive compounds, such as HPLC or UPLC, and the units used to express the measured amount of each component (e.g., ornithine or putrescine) can be any suitable units (e.g., micromoles/L, mg/L or g/L).
  • measuring the amount of ornithine or putrescine includes measuring the concentration of ornithine in a sample or measuring the total amount of ornithine in a soy hydrolysate sample. However, an amount of ornithine or putrescine is measured and whatever units are used to express the measured amount, the concentration of ornithine or putrescine in the selected batch of soy hydrolysate is less than or equal to 0.67 mg ornithine or putrescine per g soy.
  • a sample of a batch of soy hydrolysate is obtained and the ornithine or putrescine contents of the sample are measured by chromatography of the amino acids on an ion exchange column with post column ninhydrin detection.
  • screening methods include acid hydrolysis of a soy hydrolysate sample and reconstitution in a sample buffer. The hydrolyzed sample is then subject to high performance cation exchange separation on, for example, a column of a sulphonated polystyrene resin (Dowex 50) followed by a post-column derivatization that allows sensitive detection of individual amino acids within a sample. See, e.g., Moore and Stein. J. Biol. Chem.
  • liquid chromatography-mass spectrometry is used to screen a soy hydrolysate sample.
  • a sample of a batch of soy hydrolysate may be obtained as set forth herein and subjected to a chromatography run, or series of chromatography runs on a high-performance liquid chromatography (HPLC) system, such as Agilent 1100 or Agilent 1200SL.
  • HPLC high-performance liquid chromatography
  • Mass spectrometry analysis can be carried out to provide high-resolution quantitative data describing the composition of the soy hydrolysate sample being measured.
  • the present disclosure provides a method that includes screening batches of soy hydrolysate for a desired amount of a component, such as ornithine, putrescine and/or citrulline, and selecting those batches of soy hydrolysate that have a desired amount of such component.
  • a sample including a portion of a batch of soy hydrolysate powder can be screened as described above, and compared to an amino acid standard profile generated under the same conditions as the sample run. As shown in FIGS.
  • the resulting chromatogram(s) will provide the concentration of each amino acid component present in the soy hydrolysate sample (e.g., micromoles/L per amino acid, mg/L per amino acid, or g/L per amino acid). Analyzing the chromatogram, facilitates identification of batches of soy hydrolysate (i.e., samples) that contain a desired concentration of a component, such as ornithine, putrescine and/or citrulline. Each batch of soy hydrolysate that includes a desired amount of a specific component or components is then selected for further use, e.g., in cell culture, as describe herein.
  • FIG. 1 panel A depicts a rejected batch following amino acid identification.
  • FIG. 1 panel A depicts a rejected batch following amino acid identification.
  • panel B depicts an example of an acceptable soy hydrolysate batch run under identical conditions.
  • the amino acid peak corresponding to ornithine is circled in both figures.
  • the concentration of ornithine or putrescine can be determined generating a standard curve and interpolating the sample ornithine or putrescine concentration.
  • the relative amount of ornithine or putrescine can be determined by determining the area under the curve for the ornithine or putrescine peak and dividing it by the total of the areas under the peak for all amino acids, or comparing the peak area to a standard.
  • the desired concentration of the component of soy hydrolysate (e.g., ornithine or putrescine) to be selected is 5 mg/L or less. In one embodiment, the desired concentration of ornithine or putrescine in a batch of soy hydrolysate to be selected ranges from 0.5 mg/L to 5.0 mg/L or from 0.5 mg/L to 2.0 mg/L.
  • the concentration of ornithine or putrescine in a selected batch of soy hydrolysate ranges from 0.5 mg/L to 4.5 mg/L, 0.5 mg/L to 4.0 mg/L, 0.5 mg/L to 3.5 mg/L, 0.5 mg/L to 3.0 mg/L, 0.5 mg/L to 2.5 mg/L, 0.5 mg/L to 2.0 mg/L, 0.5 mg/L to 1.5 mg/L or 0.5 mg/L to 1.0 mg/L.
  • the concentration of ornithine or putrescine in a selected batch of soy hydrolysate ranges from 1.0 mg/L to 5.0 mg/L, 1.5 mg/L to 5.0 mg/L, 2.0 mg/L to 5.0 mg/L, 2.5 mg/L to 5.0 mg/L, 3.0 mg/L to 5.0 mg/L, 3.5 mg/L to 5.0 mg/L, 4.0 mg/L to 5.0 mg/L or 4.5 mg/L to 5.0 mg/L.
  • the desired concentration of ornithine or putrescine in a batch of soy hydrolysate is at least 0.5 mg/L, 0.6 mg/L, 0.7 mg/L, 0.8 mg/L, 0.9 mg/L, 1.1 mg/L, 1.2 mg/L, 1.3 mg/L, 1.4 mg/L, 1.5 mg/L, 1.6 mg/L, 1.7 mg/L, 1.8 mg/L, 1.9 mg/L, 2.0 mg/L, 2.1 mg/L, 2.2 mg/L, 2.3 mg/L, 2.4 mg/L, 2.5 mg/L, 2.6 mg/L, 2.7 mg/L, 2.8 mg/L, 2.9 mg/L, 3.0 mg/L, 3.1 mg/L, 3.2 mg/L, 3.3 mg/L, 3.4 mg/L, 3.5 mg/L, 3.6 mg/L, 3.7 mg/L, 3.8 mg/L, 3.9 mg/L, 4.0 mg/L, 4.1 mg/L, 4.2 mg/L, 4.3 mg
  • the desired concentration of ornithine or putrescine in a batch of soy hydrolysate is not more than 0.67 mg ornithine per g soy. In still other embodiments, the desired concentration of ornithine or putrescine in a batch of soy hydrolysate is not more than 0.27 mg ornithine per g soy. In another embodiment, the desired concentration of ornithine or putrescine in a batch of soy hydrolysate is not more than 0.24 mg ornithine or putrescine per g soy.
  • the desired concentration of ornithine or putrescine in a batch of soy is from 0.067 mg to 0.67 mg ornithine or putrescine per g soy.
  • the desired concentration of ornithine or putrescine in a batch of soy hydrolysate falls with the range of 0.067 mg to 0.27 mg ornithine per g soy.
  • the desired concentration of ornithine or putrescine in a batch of soy hydrolysate falls with the range of 0.067 mg to 0.24 mg ornithine or putrescine per g soy.
  • the relative amount by mass of ornithine or putrescine (% w/w) in the selected soy hydrolysate is ⁇ 0.067%, such as 0.0001%, 0.0002%, 0.0003%, 0.0004%, 0.0005%, 0.0006%, 0.0007%, 0.0008%, 0.0009%, 0.001%, 0.0015%, 0.002%, 0.0025%, 0.003%, 0.0035%, 0.004%, 0.0045%, 0.005%, 0.0055%, 0.006%, 0.0061%, 0.0062%, 0.0063%, 0.0064%, 0.0065%, 0.0066%, all by w/w.
  • the plant protein hydrolysate is selected on the basis of producing a glycoprotein with a specific quality attribute.
  • the quality of glycoprotein may be determined by assessing the level of one or more specific N-glycans on the glycoprotein, or by assessing the level of one or more specific sugars on the glycoprotein, or a combination of multiple attributes.
  • a glycoprotein having a specific fucose level can be a quality attribute criterion; or a specific sialic acid level, e.g., 5-15 moles of sialic acid per mole of glycoprotein; or a specific ratio of A1 N-glycan per total of all N-glycans, e.g., 10-17% (w/w) can be considered to have the requisite quality attribute.
  • a plant protein hydrolysate enabling the production of said glycoprotein would be considered selectable.
  • the plant protein hydrolysate is selected by producing a glycoprotein in a cell cultured in a medium containing the potential selected (potentially selectable) plant protein hydrolysate (e.g., soy hydrolysate), purifying the glycoprotein, subjecting the glycoprotein to oligosaccharide fingerprinting, and determining the relative amount of A1 N-glycan by calculating the area under the peak associated with the A1 N-glycan and dividing that value by the total area under the peaks of all N-glycans, and selecting a plant protein hydrolysate that enabled the production of a glycoprotein with a relative amount of A1 N-glycan of ⁇ 10%, ⁇ 10.5%, 10-17%, 10%, 10.5%, 11%, 11.5%, 12%, 12.5%, 13%, 13.5%, 14%, 14.5%, 15%, 15.5%, 16%, 16.5%, 17%, 17.5%, or 18%.
  • the potential selected (potentially selectable) plant protein hydrolysate e.g., so
  • the present disclosure provides a method for culturing cells expressing a protein of interest in a cell culture medium using a selected batch of soy hydrolysate as described above.
  • the instant disclosure has found, for the first time, that the use of selected batches of soy hydrolysate comprising 5.0 mg/L ornithine or less in cell culture media reduces lot-to-lot variability and improves protein product quality.
  • the instant disclosure has found, for the first time, that the use of selected batches of soy hydrolysate comprising 5.0 mg/L putrescine or less in cell culture media reduces lot-to-lot variability and improves protein product quality.
  • Cell culture or “culture” means the growth and propagation of cells outside of a multicellular organism or tissue. Suitable culture conditions for mammalian cells are known in the art. See, e.g., Animal cell culture: A Practical Approach, D. Rickwood, ed., Oxford University Press, New York (1992). Mammalian cells may be cultured in suspension or while attached to a solid substrate. Fluidized bed bioreactors, hollow fiber bioreactors, roller bottles, shake flasks, or stirred tank bioreactors, with or without microcarriers, and operated in a batch, fed batch, continuous, semi-continuous, or perfusion mode are available for mammalian cell culture. Cell culture media or concentrated feed media may be added to the culture continuously or at intervals during the culture. For example, a culture may be fed once per day, every other day, every three days, or may be fed when the concentration of a specific medium component, which is being monitored, falls outside a desired range.
  • cell culture media refers to any nutrient solution used for growing cells, e.g., animal or mammalian cells, and which generally provides at least one or more components from the following: an energy source (usually in the form of a carbohydrate such as glucose); one or more of all essential amino acids, and generally the twenty basic amino acids, plus cysteine; vitamins and/or other organic compounds typically required at low concentrations; lipids or free fatty acids; and trace elements, e.g., inorganic compounds or naturally occurring elements that are typically required at very low concentrations, usually in the micromolar range.
  • a cell culture media is formed by combining a soy or other plant protein hydrolysate with an additional ingredient.
  • additional ingredient includes any one or more of cell culture media components including but not limited to water, an energy source, one or more of all essential amino acids, and generally the twenty basic amino acids, plus cysteine; vitamins and/or other organic compounds typically required at low concentrations, lipids or free fatty acids, and trace elements.
  • the cell culture media is supplemented with an amount of a selected batch of soy hydrolysate. In certain embodiments, the cell culture medium is supplemented with about 0.5 g/L to about 25 g/L of a selected soy hydrolysate.
  • the cell culture medium is supplemented with about 0.5 g/L, 1 g/L, 1.5 g/L, 2 g/L, 2.5 g/L, 2 g/L, 2.5 g/L, 3 g/L, 3.5 g/L, 4 g/L, 4.5 g/L, 5 g/L, 5.5 g/L, 6 g/L, 6.5 g/L, 7 g/L, 7.5 g/L, 8 g/L, 8.5 g/L, 9 g/L, 9.5 g/L, 10 g/L, 10.5 g/L, 11 g/L, 11.5 g/L, 12 g/L, 12.5 g/L, 13 g/L, 13.5 g/L, 14 g/L, 14.5 g/L, 15 g/L, 15.5 g/L, 16 g/L, 16.5 g/L, 17 g/L, 17.5 g/L, 18
  • the concentration of ornithine or putrescine in the cell culture media after addition of the plant protein hydrolysate is ⁇ 5 mg/L, 0.6-3 mg/L, 0.01 mg/L, 0.02 mg/L, 0.03 mg/L, 0.04 mg/L, 0.05 mg/L, 0.06 mg/L, 0.07 mg/L, 0.08 mg/L, 0.09 mg/L, 0.010 mg/L, 0.015 mg/L, 0.02 mg/L, 0.025 mg/L, 0.03 mg/L, 0.035 mg/L, 0.04 mg/L, 0.045 mg/L, 0.05 mg/L, 0.055 mg/L, 0.06 mg/L, 0.065 mg/L, 0.07 mg/L, 0.075 mg/L, 0.08 mg/L, 0.085 mg/L, 0.09 mg/L, 0.095 mg/L, 0.1 mg/L, 0.15 mg/L, 0.2 mg/L, 0.25 mg/L, 0.3 mg/L, 0.35 mg/L, 0.10 mg/
  • the cells being cultured are cells of a cell line capable of producing a biotherapeutic protein.
  • cell lines that are used to produce protein biotherapeutics include inter alia primary cells, BSC cells, HeLa cells, HepG2 cells, LLC-MK cells, CV-1 cells, COS cells, VERO cells, MDBK cells, MDCK cells, CRFK cells, RAF cells, RK cells, TCMK-1 cells, LLCPK cells, PK15 cells, LLC-RK cells, MDOK cells, BHK cells, BHK-21 cells, CHO cells, CHO-K1 cells, NS-1 cells, MRC-5 cells, WI-38 cells, BHK cells, 3T3 cells, 293 cells, RK cells, Per.C6 cells and chicken embryo cells.
  • the cell line is a CHO cell line or one or more of several specific CHO cell variants optimized for large-scale protein production, e.g., CHO-K1, or the CHO-K1-derived EESYR® (enhanced expression and stability regions) cells (U.S. Pat. No. 7,771,997).
  • the cells that are cultured and express the heterologous glycoprotein are a population of cells obtained by clonal expansion of a cell (i.e., the progenitor cell) that harbors and expresses a polynucleotide encoding the glycoprotein or a subunit of the glycoprotein, where the glycoprotein is a complex multi-subunit protein like an antibody.
  • a cell i.e., the progenitor cell
  • the glycoprotein is a complex multi-subunit protein like an antibody.
  • at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, or about 100% of the constituent cells of the population of cells obtained or descended by clonal expansion from the progenitor cell contain the glycoprotein-encoding polynucleotide and express the glycoprotein.
  • Mammalian cells such as CHO cells, may be cultured in small scale cell culture containers, such as in 125 ml containers having about 25 ml of media, 250 ml containers having about 50 to 100 ml of media, 500 ml containers having about 100 to 200 ml of media.
  • the cultures can be large scale such as for example 1000 ml containers having about 300 to 1000 ml of media, 3000 ml containers having about 500 ml to 3000 ml of media, 8000 ml containers having about 2000 ml to 8000 ml of media, and 15000 ml containers having about 4000 ml to 15000 ml of media.
  • Cultures for manufacturing i.e., production cell cultures
  • production cell cultures such as for clinical manufacturing of protein therapeutics, are typically maintained for days, or even weeks, while the cells produce the desired protein(s). During this time the culture can be supplemented with a concentrated feed medium containing components, such as nutrients and amino acids, which are consumed during the course of the culture.
  • a concentrated feed medium is used.
  • Concentrated feed medium may be based on any cell culture media formulation.
  • Such a concentrated feed medium can contain most of the components of a cell culture medium described herein at, for example, about 5 ⁇ , 6 ⁇ , 7 ⁇ , 8 ⁇ , 9 ⁇ , 10 ⁇ , 12 ⁇ , 14 ⁇ , 16 ⁇ , 20 ⁇ , 30 ⁇ , 50 ⁇ , 100 ⁇ , 200 ⁇ , 400 ⁇ , 600 ⁇ , 800 ⁇ , or even about 1000 ⁇ of their normal useful amount.
  • Concentrated feed media are often used in fed batch culture processes.
  • the cell culture media is supplemented with “point-of-use additions”, also known as additions, point-of-use ingredients, or point-of-use chemicals, during the course of cell growth or protein production.
  • Point-of-use additions include any one or more of a growth factor or other proteins, a buffer, an energy source, a salt, an amino acid, a metal, and a chelator. Other proteins include transferrin and albumin.
  • Growth factors which include cytokines and chemokines, are generally known in the art and are known to stimulate cell growth, or in some cases, cellular differentiation.
  • a growth factor is usually a protein (e.g., insulin), a small peptide, or a steroid hormone, such as estrogen, DHEA, testosterone, and the like.
  • a growth factor may be a non-natural chemical that promotes cell proliferation or protein production, such as e.g., tetrahydrofolate (THF), methotrexate, and the like.
  • Non-limiting examples of protein and peptide growth factors include angiopoietins, bone morphogenetic proteins (BMPs), brain-derived neurotrophic factor (BDNF), epidermal growth factor (EGF), erythropoietin (EPO), fibroblast growth factor (FGF), glial cell line-derived neurotrophic factor (GDNF), granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), growth differentiation factor-9 (GDF9), hepatocyte growth factor (HGF), hepatoma-derived growth factor (HDGF), insulin, insulin-like growth factor (IGF), migration-stimulating factor, myostatin (GDF-8), nerve growth factor (NGF) and other neurotrophins, platelet-derived growth factor (PDGF), thrombopoietin (TPO), transforming growth factor alpha(TGF- ⁇ ), transforming growth factor beta(TGF- ⁇ ), tumor necrosis factor-al
  • the cell culture media is supplemented with the point-of-use addition growth factor insulin.
  • the concentration of insulin in the media i.e., the amount of insulin in the cell culture media after addition is from about 0.1 ⁇ M to 10 ⁇ m.
  • One or more the point-of-use additions can also be included in the media formulation of some embodiments.
  • Buffers are generally known in the art. The invention is not restricted to any particular buffer or buffers, and any one of ordinary skill in the art can select an appropriate buffer or buffer system for use with a particular cell line producing a particular protein.
  • a point-of-use addition buffer is NaHCO3/CO2 system. In one embodiment, the point-of-use addition buffer comprises NaHCO 3 . In another embodiment, the buffer is HEPES.
  • the point-of-use addition energy source is glucose.
  • the glucose can be added to a concentration of about 1 to 20 mM in the media.
  • Chelators are likewise well known in the art of cell culture and protein production. Tetrasodium EDTA dehydrate and citrate are two common chelators used in the art, although other chelators may be employed in the practice of this invention.
  • a point-of-use addition chelator is tetrasodium EDTA dihydrate.
  • a point-of-use addition chelator is citrate, such as Na3C6H5O7.
  • the cell culture may be supplemented with one or more point-of-use addition amino acids such as, glutamine.
  • Other point-of-use additions include one or more of various metal salts, such as salts of iron, nickel, zinc and copper.
  • the cell culture media is supplemented with any one or more of copper sulfate, zinc sulfate, ferric chloride; and nickel sulfate.
  • the media is supplemented at intervals during cell culture according to a fed-batch process.
  • Fed-batch culturing is generally known in the art and employed to optimized protein production. See, e.g., Y. M. Huang et al., Biotechnol Prog. (2010) 26(5) pp. 1400-1410.
  • cells cultured in medium comprising soy hydrolysate containing ornithine or putrescine at a desired concentration i.e., less than or equal to 5.0 mg/L, e.g., from 0.5 mg/L to 5.0 mg/L or from 0.5 mg/L to 2.0 mg/L
  • a desired concentration i.e., less than or equal to 5.0 mg/L, e.g., from 0.5 mg/L to 5.0 mg/L or from 0.5 mg/L to 2.0 mg/L
  • improved protein quality is measured by: the presence or absence of glycosylation at one or more amino acids on the protein of interest, the amount of glycan on the protein of interest, the presence of sialic acid at one or more glycosylation sites on the protein of interest, or a combination thereof.
  • improved quality can also refer to the more consistent quality, for example, post-translational modifications observed in a biotherapeutic protein production lot.
  • Consistent quality includes having, for example, a repeatable desired glycosylation profile after replicate production lines. Consistency, with respect to quality, refers to a degree of uniformity and standardization, whereas replicate production batches are essentially free from variation.
  • the protein product is an antibody, a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a multispecific antibody, a bispecific antibody, an antigen binding antibody fragment, a single chain antibody, a diabody, triabody or tetrabody, a Fab fragment or a F(ab′)2 fragment, an IgD antibody, an IgE antibody, an IgM antibody, an IgG antibody, an IgG1 antibody, an IgG2 antibody, an IgG3 antibody, or an IgG4 antibody.
  • the antibody is an IgG1 antibody.
  • the antibody is an IgG2 antibody.
  • the antibody is an IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1/IgG4 antibody.
  • the antibody is selected from the group consisting of an anti-Programmed Cell Death 1 antibody (e.g., an anti-PD1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/0203579A1), an anti-Programmed Cell Death Ligand-1 (e.g., an anti-PD-L1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/0203580A1), an anti-D114 antibody, an anti-Angiopoetin-2 antibody (e.g., an anti-ANG2 antibody as described in U.S. Pat. No.
  • an anti-Programmed Cell Death 1 antibody e.g., an anti-PD1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/0203579A1
  • an anti-Programmed Cell Death Ligand-1 e.g., an anti-PD-L1 antibody as described in U.S. Pat. Appln. Pub. No. US2015/02035
  • an anti-Angiopoetin-Like 3 antibody e.g., an anti-AngPt13 antibody as described in U.S. Pat. No. 9,018,356
  • an anti-platelet derived growth factor receptor antibody e.g., an anti-PDGFR antibody as described in U.S. Pat. No. 9,265,827
  • an anti-Erb3 antibody e.g., an anti-Prolactin Receptor antibody as described in U.S. Pat. No. 9,302,015
  • an anti-Complement 5 antibody e.g., an anti-05 antibody as described in U.S. Pat. Appln. Pub.
  • an anti-TNF antibody an anti-epidermal growth factor receptor antibody (e.g., an anti-EGFR antibody as described in U.S. Pat. No. 9,132,192 or an anti-EGFRvIII antibody as described in U.S. Pat. Appln. Pub. No. US2015/0259423A1)
  • an anti-Proprotein Convertase Subtilisin Kexin-9 antibody e.g. an anti-PCSK9 antibody as described in U.S. Pat. No. 8,062,640 or U.S. Pat. Appln. Pub. No.
  • an anti-Growth And Differentiation Factor-8 antibody e.g., an anti-GDF8 antibody, also known as anti-myostatin antibody, as described in U.S. Pat No. 8,871,209 or 9,260,51
  • an anti-Glucagon Receptor e.g., anti-GCGR antibody as described in U.S. Pat. Appln. Pub. Nos. US2015/0337045A1 or US2016/0075778A1
  • an anti-VEGF antibody an anti-IL1R antibody
  • an interleukin 4 receptor antibody e.g., an anti-IL4R antibody as described in U.S. Pat. Appln. Pub. No.
  • an anti-interleukin 6 receptor antibody e.g., an anti-IL6R antibody, as described in U.S. Pat. Nos. 7,582,298, 8,043,617 or 9,173,880
  • an anti-IL1 antibody e.g., an anti-IL2 antibody, an anti-IL3 antibody, an anti-IL4 antibody, an anti-IL5 antibody, an anti-IL6 antibody, an anti-IL7 antibody
  • an anti-interleukin 33 e.g., anti-IL33 antibody as described in U.S. Pat. Appln. Pub. Nos.
  • an anti-Respiratory syncytial virus antibody e.g., anti-RSV antibody as described in U.S. Pat. Appln. Pub. No. US2014/0271653A1
  • an anti-Cluster of differentiation 3 e.g., an anti-CD3 antibody, as described in U.S. Pat. Appln. Pub. Nos. US2014/0088295A1 and US20150266966A1, and in U.S. Application No. 62/222,605
  • an anti-Cluster of differentiation 20 e.g., an anti-CD20 antibody as described in U.S. Pat. Appln. Pub. Nos.
  • the bispecific antibody is selected from the group consisting of an anti-CD3 ⁇ anti-CD20 bispecific antibody (as described in U.S. Pat. Appln. Pub. Nos.
  • the protein of interest is selected from the group consisting of alirocumab, sarilumab, fasinumab, nesvacumab, dupilumab, trevogrumab, evinacumab, and rinucumab. All publications mentioned throughout this disclosure are incorporated herein by reference in their entirety.
  • the protein of interest is a recombinant protein that contains an Fc moiety and another domain, (e.g., an Fc-fusion protein).
  • an Fc-fusion protein is a receptor Fc-fusion protein, which contains one or more extracellular domain(s) of a receptor coupled to an Fc moiety.
  • the Fc moiety comprises a hinge region followed by a CH2 and CH3 domain of an IgG.
  • the receptor Fc-fusion protein contains two or more distinct receptor chains that bind to either a single ligand or multiple ligands.
  • an Fc-fusion protein is a trap protein, such as for example an IL-1 trap (e.g., rilonacept, which contains the IL-1RAcP ligand binding region fused to the Il-1R1 extracellular region fused to Fc of hIgG1; see U.S. Pat. No. 6,927,004, which is herein incorporated by reference in its entirety), a VEGF trap (e.g., aflibercept or ziv-aflibercept, which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 fused to Fc of hIgG1; see U.S. Pat. Nos.
  • an IL-1 trap e.g., rilonacept, which contains the IL-1RAcP ligand binding region fused to the Il-1R1 extracellular region fused to Fc of hIgG1; see U.S. Pat. No. 6,927,004,
  • conbercept which contains the Ig domain 2 of the VEGF receptor Flt1 fused to the Ig domain 3 of the VEGF receptor Flk1 fused to the Ig domain 4 of the VEGF receptor Flk1 fused to Fc of hIgG1; see U.S. Pat. No. 8,216,575), or a TNF trap (e.g., etanercept, which contains the TNF receptor fused to Fc of hIgG1; see U.S. Pat. No. 5,610,279).
  • TNF trap e.g., etanercept, which contains the TNF receptor fused to Fc of hIgG1; see U.S. Pat. No. 5,610,279
  • an Fc-fusion protein is a ScFv-Fc-fusion protein, which contains one or more of one or more antigen-binding domain(s), such as a variable heavy chain fragment and a variable light chain fragment, of an antibody coupled to an Fc moiety.
  • a protein of interest can be expressed by a host cell using methods known by those of ordinary skill in the art.
  • any protein of interest suitable for expression in mammalian cells can be produced by the instant methods, however glycoproteins will especially benefit from the present methods.
  • the protein of interest is an antibody or antigen-binding fragment thereof, a bispecific antibody or fragment thereof, a chimeric antibody or fragment thereof, an ScFv or fragment thereof, an Fc-tagged protein (e.g., Trap protein) or fragment thereof, a growth factor or a fragment thereof, a cytokine or a fragment thereof, or an extracellular domain of a cell surface receptor or fragment thereof.
  • Glycoproteins with asparagine-linked (N-linked) glycans are ubiquitous in eukaryotic cells. Biosynthesis of these glycans and their transfer to polypeptides takes place in the endoplasmic reticulum (ER). N-glycan structures are further modified by a number of glycosidases and glycosyl-transferases in the ER and the Golgi complex. Protein production using the present methods is directed at improving consistency of desired N-glycan structure in order to eliminate immunogenic epitopes (“glycotopes”). Detailed structural analysis of glycan-linked proteins may be correlated to functional features of the protein.
  • Such analysis characterizing protein glycosylation typically involves several steps: i) an enzymatic or chemical release of the attached glycans; ii) derivatization of the released glycans via reductive amination with aromatic or aliphatic amines or permethylation; iii) analysis of the glycans.
  • Many variations of analyzing glycosylation patterns in known to the skilled person Glycoproteins may carry several types of glycoforms occupying various sites in specific quantities, and therefore their complexity may make it difficult to reproduce in certain production methods. Consistency of type and quantity of glycoform is measurable and represents a desirable outcome for therapeutic protein production.
  • the present disclosure shows that producing numerous batches of a protein of interest in batch or fed-batch culture, by culturing cells expressing the protein of interest in media comprising soy hydrolysate with specific concentrations of ornithine or putrescine result in increased quality of the proteins being produced and improved consistency from batch-to-batch. Therefore, another aspect of the present disclosure provides a plurality of protein preparations that have each been produced by culturing cells in media comprising separate batches of soy hydrolysate containing a predetermined amount of ornithine or putrescine.
  • each batch of soy hydrolysate selected for use in cell culture has a concentration of 0.67 mg ornithine or putrescine per g soy or less, particularly from 0.0067 mg to 0.67 mg ornithine or putrescine per g soy, or from 0.0067 to 0.27 mg ornithine or putrescine per g soy.
  • the concentration of ornithine or putrescine in a soy hydrolysate-containing cell culture media ranges from 0.5 mg/L to 4.5 mg/L, 0.5 mg/L to 4.0 mg/L, 0.5 mg/L to 3.5 mg/L, 0.5 mg/L to 3.0 mg/L, 0.5 mg/L to 2.5 mg/L, 0.5 mg/L to 2.0 mg/L, 0.5 mg/L to 1.5 mg/L or 0.5 mg/L to 1.0 mg/L.
  • the concentration of ornithine or putrescine in soy hydrolysate-containing cell culture media ranges from 1.0 mg/L to 5.0 mg/L, 1.5 mg/L to 5.0 mg/L, 2.0 mg/L to 5.0 mg/L, 2.5 mg/L to 5.0 mg/L, 3.0 mg/L to 5.0 mg/L, 3.5 mg/L to 5.0 mg/L, 4.0 mg/L to 5.0 mg/L or 4.5 mg/L to 5.0 mg/L.
  • a cell culture media containing soy hydrolysate includes ornithine or putrescine at an amount of 0.5 mg/L, 0.6 mg/L, 0.7 mg/L, 0.8 mg/L, 0.9 mg/L, 1.1 mg/L, 1.2 mg/L, 1.3 mg/L, 1.4 mg/L, 1.5 mg/L, 1.6 mg/L, 1.7 mg/L, 1.8 mg/L, 1.9 mg/L, 2.0 mg/L, 2.1 mg/L, 2.2 mg/L, 2.3 mg/L, 2.4 mg/L, 2.5 mg/L, 2.6 mg/L, 2.7 mg/L, 2.8 mg/L, 2.9 mg/L, 3.0 mg/L, 3.1 mg/L, 3.2 mg/L, 3.3 mg/L, 3.4 mg/L, 3.5 mg/L, 3.6 mg/L, 3.7 mg/L, 3.8 mg/L, 3.9 mg/L, 4.0 mg/L, 4.1 mg/L, 4.2 mg/L, 4.3 mg
  • the desired concentration of ornithine or putrescine in a batch of media containing soy hydrolysate is not more than 5.0 mg/L. In still other embodiments, the desired concentration of ornithine or putrescine in a batch of media containing soy hydrolysate is not more than 2.0 mg/L. In another embodiment, the desired concentration of ornithine or putrescine in a batch of media containing soy hydrolysate is not more than 1.8 mg/L. In some embodiments, the desired concentration of ornithine or putrescine in a batch of media containing soy is from 0.5 mg/L to 5.0 mg/L.
  • the desired concentration of ornithine or putrescine in a batch of media containing soy hydrolysate falls with the range of 0.5 mg/L to 2.0 mg/L. In yet another embodiment, the desired concentration of ornithine or putrescine in a batch of media containing soy hydrolysate falls with the range of 0.5 mg/L to 1.8 mg/L.
  • quality of the protein of interest or amount of certain glycans produced in each protein preparation of a plurality of protein preparations is improved when compared to a protein preparation produced by a method including culturing cells in media supplemented with soy hydrolysate containing ornithine or putrescine at a concentration of greater than 5 mg/L.
  • improved protein quality exhibited by each protein preparation is measured by: the presence or absence of glycosylation at one or more amino acids of the protein of interest, the amount of glycan on the protein of interest, the presence of sialic acid at one or more glycosylation sites on the protein of interest, or a combination thereof.
  • the protein quality corresponds to the glycosylation state of individual members of a population of proteins produced in culture.
  • quality is improved by modulating the glycosylation substitutions present on individual glycoproteins of a population of proteins produced in culture by culturing the cells in media supplemented with soy hydrolysate having a concentration of 5.0 mg/L or less of ornithine or putrescine, from 0.5 mg/L to 5.0 mg/L of ornithine or putrescine, or from 0.5 mg/L to 2.0 mg/L of ornithine or putrescine.
  • protein quality is determined by comparing the abundance of at least one glycan molecule in each batch of proteins from a plurality of protein preparations to the abundance of the same glycan molecule(s) in another batch of proteins.
  • the term “abundance” as used herein refers to the percentage of proteins having a particular glycan molecule in a particular production lot, or the amount of proteins having a particular glycan molecule relative to the amount of all types of glycan molecules in a production lot.
  • the glycan molecule is selected from the group consisting of A1, A1F, A2, A2F, Man5, NA2, NA2F, NA2G1, NA2G1F, NGA2, and NGA2FI.
  • the glycan molecule is A1 (e.g., Peak 11 of FIG. 2 ).
  • Proteins of interest produced by the cell culture methods of the instant disclosure display favorable quality characteristics.
  • Protein quality can be measure, for example, by using methods well known to those skilled in the art, such as weak cation exchange chromatography, capillary isoelectric focusing, size-exclusion chromatography, High Performance Liquid Chromatography (HPLC), ELISA, and/or western blot analysis.
  • protein quality is measured by mass spectrometry, such as capillary electrophoresis mass spectrometry (CE-MS).
  • CE-MS capillary electrophoresis mass spectrometry
  • protein quality is determined by comparing mass spectrometry read outs of each batch of proteins from a plurality of protein preparations.
  • HPLC High Performance Liquid Chromatography
  • proteins of interest glycoproteins
  • soy hydrolysate with an ornithine or putrescine concentration from 0.5 mg/L to 5.0 mg/L have more consistent glycan expression and glycosylation patterns, as exemplified in Tables 2-4, herein.
  • the extent and distribution of specific N-linked sugar chains on glycoproteins can be ascertained by oligosaccharide profiling.
  • the glycoprotein is deglycosylated with peptide:N-glycosidase F (PNGase F) to cleave and remove the N-linked oligosaccharides from asparagine side chains.
  • PNGase F peptide:N-glycosidase F
  • the oligosaccharides are then derivatized with a fluorescent reagent, such as anthranilic acid.
  • the sugar chains are then separated by normal phase anion-exchange HPLC and detected with a fluorescence detector, generating an HPLC chromatogram.
  • individual glycopeptides are isolated following trypsin digestion of reduced and alkylated glycoprotein.
  • Individual tryptic glycopeptides are separated by reverse phase HPLC, coupled with a subsequent C18 column for increased resolution as needed.
  • the oligosaccharides are released from each of the separated glycopeptides by PNGase F digestion, derivatized with anthranilic acid, and analyzed by fluorescence HPLC to obtain a site-specific oligosaccharide profile of the glycoprotein.
  • glycoprotein is rilonacept (SEQ ID NO: 1)
  • asparagine residues at N37, N87, N91, N98, optionally N176, N189, N279, N418, N511, N551, N567, N581, N615, and N730 are glycosylated.
  • any one or more of residues N37, N98, N418, and N511 of rilonacept (residue positions correlating to SEQ ID NO: 1) contain an A1 oligosaccharide.
  • glycoprotein is aflibercept (SEQ ID NO: 2)
  • asparagine residues at N36, N68, N123, N196, and N282 are glycosylated.
  • any one or both of residues N123 and N196 of aflibercept contain an A1 oligosaccharide.
  • oligosaccharide pools from the glycoprotein are generated by deglycosylation of the proteins with PNGase F, followed by anthranilic acid derivatization and subsequent solid phase extraction (SPE). The masses of oligosaccharides are then measured using MALDI-TOF in a negative linear mode with 2, 4, 6-trihydroxyacetophenone (THAP) as matrix.
  • SPE solid phase extraction
  • Each observed mass is assigned to a unique oligosaccharide structure based on the masses of commonly observed N-linked glycans in recombinant proteins.
  • the expected mass assignments of all the peaks are summarized in Table 1.
  • the expected masses are the average mass calculated based on the proposed N-linked sugar chain structures with addition of anthranilic acid residue mass.
  • the monosaccharide compositions are listed based on the proposed N-linked sugar chain structures as well.
  • a quantitative oligosaccharide fingerprint assay using capillary electrophoresis is used to characterize the N-glycan (oligosaccharide) structure of the subject glycoprotein.
  • the glycoprotein is denatured and then deglycosylated by treatment with PNGase F.
  • Released oligosaccharides are then isolated by precipitation following removal of the protein.
  • Isolated oligosaccharide pools are labeled with the fluorophore 8-aminopyrene 1,3,6-trisulfonate (APTS). Labeled oligosaccharides are then separated by capillary electrophoresis and monitored with a laser induced fluorescence detector using an excitation wavelength of 488 nm and emission wavelength of 520 nm.
  • An electropherogram is generated, as depicted in FIG. 2 for the aflibercept glycoprotein, with all quantifiable peaks numbered (total of 21 peaks in this example).
  • the complete integrated peak area (total peak area) for the oligosaccharide fingerprint is determined.
  • the relative amount of each oligosaccharide can be determined by dividing the peak area for that particular oligosaccharide (e.g., A1 peak area) by the total peak area.
  • the quality of the subject glycoprotein is assessed by determining the level of sialylation (the amount of sialic residues per glycoprotein) or fucosylation (the amount of fucose residues per glycoprotein).
  • the total number of sialic acids on a glycoprotein are determined using a quantitative HPLC assay. In this assay, the sialic acids are released from the glycoprotein using mild acid hydrolysis, then derivatized with o-Phenylenediamine, separated by HPLC, and detected with either UV or fluorescence detectors. Quantitation of sialic acid can be assessed relative to a standard curve using e.g., sialyllactose. The sialic acid content is calculated from the moles of sialic acid released and the moles of the glycoprotein used in the reaction.
  • the sialic acid content of the rilonacept glycoprotein is about 30-70 moles sialic acid per 1 mole of glycoprotein (mol/mol), about 35-65 mol/mol, 30 mol/mol, 31 mol/mol, 32 mol/mol, 33 mol/mol, 34 mol/mol, 35 mol/mol, 36 mol/mol, 37 mol/mol, 38 mol/mol, 39 mol/mol, 40 mol/mol, 41 mol/mol, 42 mol/mol, 43 mol/mol, 44 mol/mol, 45 mol/mol, 46 mol/mol, 47 mol/mol, 48 mol/mol, 49 mol/mol, 50 mol/mol, 51 mol/mol, 52 mol/mol, 53 mol/mol, 54 mol/mol, 55 mol/mol, 56 mol/mol, 57 mol/mol, 58 mol/mol, 59 mol/mol, 60 mol/mol, 61 mol/mol, 62 mol/mol,
  • the sialic acid content of the aflibercept glycoprotein is about 5-15 moles sialic acid per 1 mole of glycoprotein (mol/mol), about 8-12 mol/mol, 4 mol/mol, 5 mol/mol, 6 mol/mol, 7 mol/mol, 8 mol/mol, 9 mol/mol, 10 mol/mol, 11 mol/mol, 12 mol/mol, 13 mol/mol, 14 mol/mol, 15 mol/mol, 16 mol/mol, 17 mol/mol, 18 mol/mol, 19 mol/mol, or 20 mol/mol.
  • oligosaccharide profiling is employed to determine the extent and distribution of sialylation of N-linked sugar chains on the glycoprotein.
  • the glycoprotein is deglycosylated with PNGase F, and then derivatized with the fluorescent reagent, anthranilic acid.
  • the oligosaccharides are then separated by normal phase anion-exchange HPLC and detected with a fluorescence detector to generate an HPLC chromatogram of the oligosaccharide profile.
  • the Z number (which measures the average degree of sialylation) for the glycoprotein is calculated from the following formula:
  • the Z number is the area of each peak from the oligosaccharide profile.
  • the total sialic acid is calculated as the sum of the areas of the 0 sialic acid/chain peaks multiplied by 0, the 1 sialic acid/chain peaks multiplied by 1, the 2 sialic acid/chain peaks multiplied by 2, and the 3 sialic acid/chain peaks multiplied by 3, etc.
  • the total number of sugar chains is generated as the sum of the areas of all of the peaks.
  • the Z number is the total sialic acid area divided by the total sugar chain area.
  • the sialic acid Z number of the rilonacept glycoprotein is about 1.3-1.6, 1.4-1.5, 1.41-1.48, 1.3, 1.31, 1.32, 1.33, 1.34, 1.35, 1.36, 1.37, 1.38, 1.39, 1.4, 1.41, 1.42, 1.43, 1.44, 1.45, 1.46, 1.47, 1.48, 1.49, 1.5, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, or 1.60.
  • the sialic acid Z number of the aflibercept glycoprotein is about 0.5-2, 1-1.5, 1-1.2, 0.5, 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.6, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, 0.7, 0.71, 0.72, 0.73, 0.74, 0.75, 0.76, 0.77, 0.78, 0.79, 0.8, 0.81, 0.82, 0.83, 0.84, 0.86, 0.87, 0.88, 0.89, 0.9, 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, 0.99, 1, 1.01, 1.02, 1.03, 1.04, 1.05, 1.06, 1.07, 1.08, 1.09, 1.1, 1.11, 1.12, 1.13, 1.14, 1.15, 1.16, 1.17, 1.18, 1.19, 1.2, 1.21, 1.22, 1.23, 1.24, 1.25, 1.
  • a soy hydrolysate sample was weighed and a 20 gram portion thereof was dissolved in 1 L water to a starting concentration of 20 g/L.
  • the resulting soy hydrolysate solution was then further diluted in water to a desired concentration for use in cell culture and the molecular composition of the resulting soy hydrolysate solution was determined using by chromatography.
  • the concentration of amino acids in the soy hydrolysate sample was measured by chromatography on an ion exchange column with post column ninhydrin detection. See, e.g., Moore and Stein. J. Biol. Chem. (1954) Vol. 211 pp. 907-913. Soy hydrolysate samples were diluted to permit sensitive separation and resolution of individual peaks (amino acids) as eluted from the HPLC column and compared to a standard. Each peak area of the chromatogram, as shown in FIGS. 1A and 1B , was compared to a standard to determine concentration of each eluate.
  • FIG. 1A shows a batch of soy hydrolysate with an eluate containing ornithine at retention time 89.02, which reveals a peak area equivalent to 1.57 mg ornithine per g soy of ornithine, as compared to the standard.
  • FIG. 1B illustrates a batch of soy hydrolysate having an ornithine concentration of less than 0.67 mg ornithine per g soy.
  • soy hydrolysate batches with between 0.067 and 0.67 mg ornithine per g soy were selected for use in cell culture methods to produce biotherapeutic proteins with more consistent protein glycosylation from lot-to lot.
  • soy hydrolysate batches containing ornithine at a concentration greater than less than 0.67 mg ornithine per g soy were employed in further experiments, as described below, to determine the effects of soy hydrolysate ornithine concentration on protein production.
  • CHO cells expressing a trap protein were cultured in proprietary medium including soy hydrolysate containing varying amounts of ornithine, putrescine and citrulline, or a combination thereof, in order to determine which amino acid components affect the quality of proteins produced.
  • Table 2 shows that the levels of ornithine in the hydrolysate correlate negatively with the quality of protein production lots, as indicated by the increased area under the curve for a key N-glycan for protein lots produced as a result of culturing CHO cells in media supplemented with soy hydrolysate containing ornithine at a concentration less than 5.0 mg/L independent of citrulline concentration.
  • VEGF-trap protein product lots produced by cells cultured in media including soy hydrolysate with an ornithine concentration of 2.0 mg/L or less produce higher quality protein product, when compared to cells cultured in media comprising more than 5.0 mg/L of ornithine, citrulline or putrescine.
  • VEGF-trap protein product lots produced by cells cultured in medium including soy hydrolysate with an ornithine concentration of 0.67 mg ornithine per g soy or less exhibit increased product quality and more consistent quality from lot-to-lot.
  • Each production lot was also compared (with respect to glycan profile) to a reference standard which represents a therapeutically acceptable batch of protein for the exemplary VEGF-trap protein.
  • Representative glycan analysis is shown in Table 4 for protein lots produced from cells cultured in medium supplemented with soy hydrolysate resulting in final concentration of ornithine between 0.5 mg/L and 2.0 mg/L.
  • each produced trap protein comprises a consistent glycan profile having peaks within an acceptable range (75% of lots analyzed).
  • each lot produced by cells cultured in medium supplemented with soy hydrolysate comprising over 5.0 mg/L ornithine failed to meet FDA acceptance criteria.
  • Protein product lots produced by cells cultured in media including soy hydrolysate with an ornithine concentration from 0.5 mg/L to 2.0 mg/L or less produce higher quality lots, as demonstrated by A1 N-glycan levels falling below product acceptance criteria, than cells cultured in media comprising more soy hydrolysate having an ornithine concentration greater than 5.0 mg/L.
  • FIG. 4 shows the strong negative correlation between the level of ornithine in soy hydrolysate and glycoprotein (aflibercept) quality as demonstrated by A1 N-glycan levels.
  • FIG. 5 depicts the loading plots of correlations between the soy hydrolysate analytes and maximum lactate and final glycoprotein titer. The determinations of lactate and glycoprotein titer demonstrate a negative correlation of ornithine in the soy hydrolysate.
  • FIGS. 6A and 6B depict CHO cell cultures under control media and feed conditions that received a spike of either ornithine or putrescine to demonstrate the effect of ornithine and putrescine, respectively, on cell growth and glycosylation.
  • Table 6B highlights the effect at peak 11, which is particularly pronounced.
US16/026,539 2017-07-06 2018-07-03 Cell culture process for making a glycoprotein Abandoned US20190010531A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US16/026,539 US20190010531A1 (en) 2017-07-06 2018-07-03 Cell culture process for making a glycoprotein
US16/742,723 US20200131554A1 (en) 2017-07-06 2020-01-14 Cell culture process for making a glycoprotein
US16/864,689 US20200255880A1 (en) 2017-07-06 2020-05-01 Cell culture process for making a glycoprotein
US17/409,158 US20210388407A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein
US17/409,193 US20210388408A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762529471P 2017-07-06 2017-07-06
US201862625744P 2018-02-02 2018-02-02
US16/026,539 US20190010531A1 (en) 2017-07-06 2018-07-03 Cell culture process for making a glycoprotein

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US16/742,723 Continuation US20200131554A1 (en) 2017-07-06 2020-01-14 Cell culture process for making a glycoprotein
US17/409,193 Division US20210388408A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein
US17/409,158 Division US20210388407A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein

Publications (1)

Publication Number Publication Date
US20190010531A1 true US20190010531A1 (en) 2019-01-10

Family

ID=64904487

Family Applications (5)

Application Number Title Priority Date Filing Date
US16/026,539 Abandoned US20190010531A1 (en) 2017-07-06 2018-07-03 Cell culture process for making a glycoprotein
US16/742,723 Abandoned US20200131554A1 (en) 2017-07-06 2020-01-14 Cell culture process for making a glycoprotein
US16/864,689 Abandoned US20200255880A1 (en) 2017-07-06 2020-05-01 Cell culture process for making a glycoprotein
US17/409,193 Pending US20210388408A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein
US17/409,158 Pending US20210388407A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein

Family Applications After (4)

Application Number Title Priority Date Filing Date
US16/742,723 Abandoned US20200131554A1 (en) 2017-07-06 2020-01-14 Cell culture process for making a glycoprotein
US16/864,689 Abandoned US20200255880A1 (en) 2017-07-06 2020-05-01 Cell culture process for making a glycoprotein
US17/409,193 Pending US20210388408A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein
US17/409,158 Pending US20210388407A1 (en) 2017-07-06 2021-08-23 Cell culture process for making a glycoprotein

Country Status (13)

Country Link
US (5) US20190010531A1 (fr)
EP (2) EP3649144A4 (fr)
JP (3) JP7265494B2 (fr)
KR (3) KR20210084695A (fr)
CN (2) CN114075269A (fr)
AU (3) AU2018298039B2 (fr)
BR (1) BR112020000127A2 (fr)
CA (1) CA3067847A1 (fr)
IL (1) IL271524A (fr)
MX (1) MX2020000228A (fr)
SG (1) SG11201912548XA (fr)
TW (1) TW201934570A (fr)
WO (1) WO2019010191A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10927342B2 (en) 2015-08-04 2021-02-23 Regeneran Pharmaceuticals, Inc. Taurine supplemented cell culture medium and methods of use
CN114058673A (zh) * 2021-09-15 2022-02-18 江苏先思达生物科技有限公司 一种脂肪肝检测试剂及其在脂肪肝检测中的应用
US11332771B2 (en) 2013-03-15 2022-05-17 Regeneron Pharmaceuticals, Inc. Serum-free cell culture medium

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3638697A4 (fr) 2017-06-12 2021-07-07 Bluefin Biomedicine, Inc. Anticorps anti-il1rap et conjugués anticorps-médicament
CN113679744B (zh) * 2020-05-18 2023-12-29 中国人民解放军军事科学院军事医学研究院 寨卡病毒或其与用于免疫检查点治疗的药物在治疗胶质母细胞瘤中的用途

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101603026A (zh) * 2009-06-19 2009-12-16 华东理工大学 适于动物细胞产品生产的无动物来源低蛋白培养基
US20160076068A1 (en) * 2013-05-29 2016-03-17 Roche Diagnostics Operations, Inc. Quantitative control of sialylation
US9447431B2 (en) * 2012-02-15 2016-09-20 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US20160333385A1 (en) * 2014-01-13 2016-11-17 Amgen Inc. Regulating orinthine metabolism to manipulate the high mannose glycoform content of recombinant proteins
US20170305999A1 (en) * 2016-04-26 2017-10-26 La Jolla Biologics, Inc. Cell culture medium

Family Cites Families (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK0417563T3 (da) 1989-09-12 2000-11-06 Hoffmann La Roche TNF-bindende proteiner
AT409379B (de) * 1999-06-02 2002-07-25 Baxter Ag Medium zur protein- und serumfreien kultivierung von zellen
US7087411B2 (en) 1999-06-08 2006-08-08 Regeneron Pharmaceuticals, Inc. Fusion protein capable of binding VEGF
DE60309238T2 (de) 2002-03-08 2007-06-06 Asml Netherlands B.V. Lithographische Maske, lithographischer Apparat und Verfahren zur Herstellung einer Vorrichtung
ES2381014T3 (es) 2004-06-08 2012-05-22 Chengdu Kanghong Biotechnologies Co., Ltd. Proteína quimérica inhibidora de la angiogénesis y el uso
WO2006047380A2 (fr) * 2004-10-22 2006-05-04 Amgen, Inc. Technique et milieu pour culture de cellules cho sans serum unique
US20060094104A1 (en) * 2004-10-29 2006-05-04 Leopold Grillberger Animal protein-free media for cultivation of cells
US7642078B2 (en) * 2005-12-28 2010-01-05 Crucell Holland B.V. Methods to obtain recombinant proteins with increased sialylation from cells that express adenovirus E1A protein, and proteins obtained thereby
JP5259418B2 (ja) * 2006-01-04 2013-08-07 バクスター・インターナショナル・インコーポレイテッド 無オリゴペプチド細胞培地
US8216575B2 (en) 2006-03-31 2012-07-10 Chengdu Kanghong Biotechnologies Co., Ltd. Inhibition of neovascularization with a soluble chimeric protein comprising VEGF FLT-1 and KDR domains
CN102585002A (zh) 2006-06-02 2012-07-18 瑞泽恩制药公司 人il-6受体的高亲和力抗体
US7608693B2 (en) 2006-10-02 2009-10-27 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to human IL-4 receptor
ES2522615T3 (es) 2007-06-04 2014-11-17 Regeneron Pharmaceuticals, Inc. Regiones de expresión y estabilidad potenciadas
WO2008154014A2 (fr) * 2007-06-11 2008-12-18 Amgen Inc. Methode de culture de cellules mammiferes servant a ameliorer la production de proteines recombinees
CN101809037B (zh) 2007-07-31 2014-01-15 瑞泽恩制药公司 人cd20的人抗体及其使用方法
US8309088B2 (en) 2007-08-10 2012-11-13 Regeneron Pharmaceuticals, Inc. Method of treating osteoarthritis with an antibody to NGF
JO3672B1 (ar) 2008-12-15 2020-08-27 Regeneron Pharma أجسام مضادة بشرية عالية التفاعل الكيماوي بالنسبة لإنزيم سبتيليسين كنفرتيز بروبروتين / كيكسين نوع 9 (pcsk9).
MX2011014008A (es) 2009-06-26 2012-06-01 Regeneron Pharma Anticuerpos biespecificos facilmente aislados con formato de inmunoglobulina original.
CN102115728B (zh) * 2009-12-31 2012-09-26 北京清大天一科技有限公司 无血清动物细胞培养基干粉、液体培养基及其制备方法
JO3417B1 (ar) 2010-01-08 2019-10-20 Regeneron Pharma الصيغ المستقرة التي تحتوي على الأجسام المضادة لمضاد مستقبل( interleukin-6 (il-6r
JO3340B1 (ar) 2010-05-26 2019-03-13 Regeneron Pharma مضادات حيوية لـعامل تمايز النمو 8 البشري
JOP20190250A1 (ar) 2010-07-14 2017-06-16 Regeneron Pharma صيغ مستقرة تحتوي على الأجسام المضادة لمضاد عامل نمو الأعصاب
CA2807895A1 (fr) * 2010-08-31 2012-03-08 Friesland Brands B.V. Milieu de culture pour cellules eucaryotes
AR083044A1 (es) 2010-09-27 2013-01-30 Regeneron Pharma Anticuerpos anti-cd48 y usos de los mismos
NZ609557A (en) 2010-10-06 2014-12-24 Regeneron Pharma Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies
JO3756B1 (ar) 2010-11-23 2021-01-31 Regeneron Pharma اجسام مضادة بشرية لمستقبلات الجلوكاجون
JO3412B1 (ar) 2011-06-17 2019-10-20 Regeneron Pharma أجسام مضادة ل angptl3 واستخداماتها
US20140286946A1 (en) * 2011-10-31 2014-09-25 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
CA2855822C (fr) 2011-11-14 2022-11-15 Regeneron Pharmaceuticals, Inc. Compositions et methodes permettant d'augmenter la masse et la force musculaires par un effet antagoniste specifiquement dirige contre la myostatine et/ou l'activine a
EP2807190B1 (fr) 2012-01-23 2018-12-26 Regeneron Pharmaceuticals, Inc. Formulations stabilisées contenant des anticorps anti-ang2
JO3820B1 (ar) 2012-05-03 2021-01-31 Regeneron Pharma أجسام مضادة بشرية لـ fel d1وطرق لاستخدامها
SG11201408490PA (en) * 2012-06-21 2015-01-29 Baxter Int Virus filtration of cell culture media
TW201843172A (zh) 2012-06-25 2018-12-16 美商再生元醫藥公司 抗-egfr抗體及其用途
WO2014020160A1 (fr) * 2012-08-02 2014-02-06 Sanofi Article de manufacture comprenant de l'aflibercept ou du ziv-aflibercept
AU2013302925B2 (en) 2012-08-13 2018-07-05 Regeneron Pharmaceuticals, Inc. Anti-PCSK9 antibodies with pH-dependent binding characteristics
JOP20200236A1 (ar) 2012-09-21 2017-06-16 Regeneron Pharma الأجسام المضادة لمضاد cd3 وجزيئات ربط الأنتيجين ثنائية التحديد التي تربط cd3 وcd20 واستخداماتها
JO3405B1 (ar) 2013-01-09 2019-10-20 Regeneron Pharma الأجسام المضادة لمضاد مستقبل عامل النمو المشتق من الصفائح الدموية - بيتا واستخداماتها
JO3532B1 (ar) 2013-03-13 2020-07-05 Regeneron Pharma الأجسام المضادة لمضاد انترلوكين-33 واستعمالاتها
TWI659968B (zh) 2013-03-14 2019-05-21 再生元醫藥公司 針對呼吸道融合病毒f蛋白質的人類抗體及其使用方法
US9637535B2 (en) 2013-03-15 2017-05-02 Regeneron Pharmaceuticals, Inc. IL-33 antagonists and uses thereof
US20160237400A1 (en) * 2013-03-15 2016-08-18 The Jackson Laboratory Isolation of non-embryonic stem cells and uses thereof
AR095196A1 (es) * 2013-03-15 2015-09-30 Regeneron Pharma Medio de cultivo celular libre de suero
TWI641620B (zh) 2013-08-21 2018-11-21 再生元醫藥公司 抗-prlr抗體及其用途
TWI680138B (zh) 2014-01-23 2019-12-21 美商再生元醫藥公司 抗pd-l1之人類抗體
TWI681969B (zh) 2014-01-23 2020-01-11 美商再生元醫藥公司 針對pd-1的人類抗體
CA2940685C (fr) 2014-03-11 2023-10-24 Regeneron Pharmaceuticals, Inc. Anticorps anti-egfrviii et utilisations associees
TWI754319B (zh) 2014-03-19 2022-02-01 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
ES2779070T3 (es) 2014-05-05 2020-08-13 Regeneron Pharma Animales con C5 y C3 humanizadas
JO3701B1 (ar) 2014-05-23 2021-01-31 Regeneron Pharma مضادات حيوية بشرية لمتلازمة الشرق الأوسط التنفسية - بروتين كورونا فيروس الشوكي
CA2950423A1 (fr) * 2014-05-27 2015-12-03 Academia Sinica Compositions et procedes concernant des glycoformes universelles pour une efficacite d'anticorps amelioree
CA2951025A1 (fr) * 2014-06-03 2015-12-10 Lupin Limited Processus de culture cellulaire destine a produire une proteine
US9657099B2 (en) 2014-09-16 2017-05-23 Regeneron Pharmaceuticals, Inc. Anti-glucagon antibodies
TWI710573B (zh) 2015-01-26 2020-11-21 美商再生元醫藥公司 抗伊波拉病毒醣蛋白之人類抗體

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101603026A (zh) * 2009-06-19 2009-12-16 华东理工大学 适于动物细胞产品生产的无动物来源低蛋白培养基
US9447431B2 (en) * 2012-02-15 2016-09-20 Curevac Ag Nucleic acid comprising or coding for a histone stem-loop and a poly(A) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
US20160076068A1 (en) * 2013-05-29 2016-03-17 Roche Diagnostics Operations, Inc. Quantitative control of sialylation
US20160333385A1 (en) * 2014-01-13 2016-11-17 Amgen Inc. Regulating orinthine metabolism to manipulate the high mannose glycoform content of recombinant proteins
US20170305999A1 (en) * 2016-04-26 2017-10-26 La Jolla Biologics, Inc. Cell culture medium

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11332771B2 (en) 2013-03-15 2022-05-17 Regeneron Pharmaceuticals, Inc. Serum-free cell culture medium
US11970724B2 (en) 2013-03-15 2024-04-30 Regeneron Pharmaceuticals, Inc. Serum-free cell culture medium
US10927342B2 (en) 2015-08-04 2021-02-23 Regeneran Pharmaceuticals, Inc. Taurine supplemented cell culture medium and methods of use
US11312936B2 (en) 2015-08-04 2022-04-26 Regeneron Pharmaceuticals, Inc. Taurine supplemented cell culture medium and methods of use
CN114058673A (zh) * 2021-09-15 2022-02-18 江苏先思达生物科技有限公司 一种脂肪肝检测试剂及其在脂肪肝检测中的应用

Also Published As

Publication number Publication date
JP7265494B2 (ja) 2023-04-26
CN110914293A (zh) 2020-03-24
US20200255880A1 (en) 2020-08-13
CN114075269A (zh) 2022-02-22
EP3649144A1 (fr) 2020-05-13
EP3967765A1 (fr) 2022-03-16
CA3067847A1 (fr) 2019-01-10
BR112020000127A2 (pt) 2020-07-07
KR20200026248A (ko) 2020-03-10
EP3649144A4 (fr) 2021-07-21
KR102659791B1 (ko) 2024-04-23
US20210388408A1 (en) 2021-12-16
AU2021229121A2 (en) 2021-10-14
WO2019010191A1 (fr) 2019-01-10
AU2018298039B2 (en) 2023-02-02
KR20240055885A (ko) 2024-04-29
TW201934570A (zh) 2019-09-01
JP2020526196A (ja) 2020-08-31
IL271524A (en) 2020-02-27
KR20210084695A (ko) 2021-07-07
AU2018298039A1 (en) 2020-01-16
AU2023202659A1 (en) 2023-05-18
JP2023052471A (ja) 2023-04-11
MX2020000228A (es) 2020-08-10
US20200131554A1 (en) 2020-04-30
AU2021229121A1 (en) 2021-09-30
JP2021166537A (ja) 2021-10-21
US20210388407A1 (en) 2021-12-16
SG11201912548XA (en) 2020-01-30

Similar Documents

Publication Publication Date Title
US20200255880A1 (en) Cell culture process for making a glycoprotein
ES2695169T3 (es) Métodos de cultivo de células
KR20210005689A (ko) 조절된 글리칸 프로파일을 갖는 항체
JP2023166478A (ja) グリコシル化タンパク質の電気泳動のための脱グリコシル化方法
EA042958B1 (ru) Процесс культивирования клеток для получения гликопротеина
Roucka et al. Glycosylation Pattern of Biotechnologically Produced Proteins-Lectin Array Technology as a Versatile Tool for Screening?
US20240002483A1 (en) Fab high mannose glycoforms
Gupta Implementation of CQA (critical quality attribute) based approach for development of biosimilars

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: REGENERON PHARMACEUTICALS, INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEN, JOHN;LAWRENCE, SHAWN;JOHNSON, AMY;AND OTHERS;SIGNING DATES FROM 20180829 TO 20180930;REEL/FRAME:047045/0828

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: NON FINAL ACTION MAILED

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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