US20220185843A1 - Protein purification method - Google Patents

Protein purification method Download PDF

Info

Publication number
US20220185843A1
US20220185843A1 US17/562,536 US202117562536A US2022185843A1 US 20220185843 A1 US20220185843 A1 US 20220185843A1 US 202117562536 A US202117562536 A US 202117562536A US 2022185843 A1 US2022185843 A1 US 2022185843A1
Authority
US
United States
Prior art keywords
antibody
physiologically active
active protein
dna
aqueous solution
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.)
Pending
Application number
US17/562,536
Inventor
Kozo Takeda
Norimichi Ochi
Kimie Ishii
Manabu Matsuhashi
Akinori Imamura
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.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=31986593&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20220185843(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Priority to US17/562,536 priority Critical patent/US20220185843A1/en
Assigned to CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment CHUGAI SEIYAKU KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IMAMURA, AKINORI, ISHII, KIMIE, MATSUHASHI, MANABU, OCHI, NORIMICHI, TAKEDA, KOZO
Publication of US20220185843A1 publication Critical patent/US20220185843A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/30Extraction; Separation; Purification by precipitation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/22Affinity chromatography or related techniques based upon selective absorption processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • the present invention relates to a method for purifying proteins, more specifically to a method for removing impurities such as DNA contaminants from a sample containing a physiologically active protein such as antibody molecules.
  • the aqueous medium is treated by affinity column chromatography on Protein A or G before being purified by various types of chromatography, based on the binding property of Protein A or Protein G to IgG Fc chain.
  • JP KOHYO 5-504579 an antibody-containing aqueous medium obtained from mammalian cell culture is subjected to Protein A/G column chromatography to adsorb antibody molecules onto the column, followed by elution with an acidic solution (about 0.1 M citric acid, pH 3.0-3.5) to release the antibody molecules.
  • the resulting acidic eluate is subjected sequentially to ion-exchange column chromatography and size exclusion column chromatography to give the purified antibody molecules.
  • the object of the present invention is to provide a simpler and less expensive method for purifying physiologically active proteins, especially antibodies, which can ensure removal of impurities such as DNA contaminants and viruses, and which can minimize a loss of physiologically active proteins.
  • the inventors of the present invention have made the surprising finding that impurities such as DNA contaminants and viruses can be efficiently removed from a physiologically active protein-containing sample without using complicated chromatographic processes when the sample is formed into an aqueous solution of low conductivity at a pH below the isoelectric point of the physiologically active protein and then filtrated through a filter to remove the resulting particles. This finding led to the completion of the present invention.
  • the present invention provides the following.
  • a method for removing impurities in a physiologically active protein-containing sample which comprises the steps of
  • step 1) is accomplished by forming the physiologically active protein-containing sample into an acidic or alkaline aqueous solution of low conductivity, and adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
  • physiologically active protein is an antibody
  • step 1) is accomplished by subjecting the antibody-containing sample to affinity chromatography on Protein A or G, eluting the sample with an acidic aqueous solution of low conductivity, and adjusting the resulting eluate with a buffer to a pH equal to or lower than the isoelectric point of the antibody.
  • a physiologically active protein contained in a sample to be purified by the method of the present invention examples include, but are not limited to, hematopoietic factors such as granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), erythropoietin (EPO) and thrombopoietin, cytokines such as interferons, IL-1 and IL-6, monoclonal antibodies, tissue plasminogen activator (TPA), urokinase, serum albumin, blood coagulation factor VIII, leptin, insulin, and stem cell growth factor (SCF).
  • G-CSF granulocyte colony-stimulating factor
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • EPO erythropoietin
  • cytokines such as interferons
  • IL-1 and IL-6 monoclon
  • G-CSF preferred are G-CSF and antibodies including monoclonal antibodies , and more preferred are monoclonal antibodies.
  • monoclonal antibodies are preferred for purification.
  • Antibodies are categorized into IgG, IgA, IgE, IgD and IgM classes, with IgG antibodies being preferred.
  • physiologically active protein is intended to mean a protein having substantially the same biological activities as a corresponding physiologically active protein of mammalian (especially human) origin.
  • a protein may either be native or genetically recombinant, preferably genetically recombinant.
  • Genetically recombinant physiologically active proteins may be prepared by production in bacterial cells such as E. coli; yeast cells; or animal-derived cultured cells such as Chinese hamster ovary (CHO) cells, C127 cells or COS cells. The proteins thus prepared are isolated and purified in various manners before use.
  • Such genetically recombinant proteins encompass those having the same amino acid sequence as the corresponding native protein, as well as those comprising deletion, substitution or addition of one or more amino acids in the amino acid sequence, but retaining the biological activities mentioned above. Further, such proteins include those chemically modified with PEG, etc.
  • a physiologically active protein When a physiologically active protein is a glycoprotein, it may have sugar chains of any origin, preferably of mammalian origin. Mammalian origins include, for example, Chinese hamster ovary (CHO) cells, BHK cells, COS cells and human-derived cells, with CHO cells being most preferred.
  • CHO Chinese hamster ovary
  • a physiologically active protein When a physiologically active protein is EPO, it may be prepared in any manner, for example, by obtaining from human urine in various manners or by producing with genetic engineering techniques in bacterial cells such as E. coli, yeast cells, Chinese hamster ovary (CHO) cells, BHK cells, COS cells, human-derived cells or the like (e.g., as described in JP KOKAI 61-12288). EPO thus prepared is extracted, isolated, and purified in various manners before use. In addition, EPO may be chemically modified with PEG, etc. (see International Publication No. WO90/12874). EPO as used herein further includes those originally unglycosylated but chemically modified with PEG, etc.
  • EPO analogs are also included, which are modified to have at least one additional site for N-linked or O-linked glycosylation in the amino acid sequence of EPO (see, e.g., JP KOKAI 08-151398, JP KOHYO 08-506023). Instead of increasing the number of glycosylation sites, EPO analogs may also be modified to have an increased content of sugar chains such as sialic acid for an increased amount of sugar chains.
  • G-CSF When a physiologically active protein is G-CSF, any G-CSF can be used as long as it is highly purified.
  • G-CSF as used herein may be prepared in any manner, for example, by obtaining from cultured human tumor cell lines or by producing with genetic engineering techniques in bacterial cells such as E. coli; yeast cells; or animal-derived cultured cells such as Chinese hamster ovary (CHO) cells, C127 cells or COS cells. G-CSF thus prepared is extracted, isolated, and purified in various manners before use. Preferred are those produced recombinantly in E. coli cells, yeast cells or CHO cells. The most preferred are those produced recombinantly in CHO cells.
  • G-CSF may be chemically modified with PEG, etc. (see International Publication No. WO90/12874).
  • a physiologically active protein when a physiologically active protein is a monoclonal antibody, it may be prepared in any manner.
  • a monoclonal antibody can be produced using known techniques by immunizing a sensitizing antigen in accordance with conventional procedures for immunization, fusing the resulting immunocytes with known parent cells through conventional procedures for cell fusion, and then screening monoclonal antibody-producing cells through conventional procedures for screening.
  • antibody genes are cloned from hybridomas, integrated into appropriate vectors, and then transformed into hosts to produce antibody molecules using gene recombination technology.
  • the genetically recombinant antibodies thus produced may also be used in the present invention (see, e.g., Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). More specifically, cDNA of antibody variable domains (V domains) is synthesized from hybridoma mRNA using reverse transcriptase.
  • the DNA is ligated to DNA encoding desired antibody constant domains (C domains) and integrated into an expression vector.
  • the DNA encoding the antibody V domains may be integrated into an expression vector carrying the DNA of the antibody C domains.
  • the DNA construct is integrated into an expression vector such that it is expressed under control of an expression regulatory region, e.g., an enhancer or a promoter. Host cells are then transformed with this expression vector for antibody expression.
  • chimeric antibodies e.g., chimeric antibodies, humanized antibodies
  • modified antibodies may be prepared in a known manner.
  • a chimeric antibody is composed of variable domains of heavy and light chains from a non-human mammalian (e.g., mouse) antibody and constant domains of heavy and light chains from a human antibody.
  • DNAs encoding such mouse antibody variable domains may be ligated to DNAs encoding the human antibody constant domains, and then integrated into an expression vector, followed by transformation into a host for antibody production.
  • Humanized antibodies are also called reshaped human antibodies and are obtained by grafting complementarity determining regions (CDRs) of non-human mammalian (e.g., mouse) antibodies to replace those of human antibodies. Standard gene recombination procedures for this purpose are also known. More specifically, a DNA sequence designed to allow ligation between CDRs of mouse antibody and framework regions (FRs) of human antibody is synthesized by PCR from several oligonucleotides which are prepared to have sections overlapping with one another at the ends. The DNA thus obtained is ligated to DNA encoding human antibody constant domains, and integrated into an expression vector, followed by transformation into a host for antibody production (see European Patent Publication No, EP 239400 and International Publication No.
  • CDRs complementarity determining regions
  • WO 96/02576 The FRs of human antibody, which is ligated via CDRs, are selected such that the complementarity determining regions form a favorable antigen-binding site. If necessary, amino acid substitutions may be made in the framework regions of antibody variable domains such that the complementarity determining regions of reshaped humanized antibody may form an appropriate antigen-binding site (Sato, K. et al., Cancer Res. (1993) 53, 851-856).
  • a humanized anti-IL-6 receptor antibody (hPM-1) can be presented as a preferred example for such reshaped humanized antibodies (see International Publication No. WO92-19759).
  • a humanized anti-HM1.24 antigen monoclonal antibody see International Publication No. WO98-14580
  • a humanized anti-parathyroid hormone-related peptide antibody (anti-PTHrP antibody; see International Publication No. WO98-13388)
  • a humanized anti-tissue factor antibody see International Publication No. WO99-51743 and the like are also preferred for use in the present invention.
  • human lymphocytes are sensitized in vitro with a desired antigen or a desired antigen-expressing cell, and the sensitized lymphocytes are then fused with human myeloma cells (e.g., U266) to give desired human antibodies having binding activity to the antigen (see JP KOKOKU 01-59878).
  • human myeloma cells e.g., U266
  • transgenic animals having the entire repertories of human antibody genes may be immunized with an antigen to obtain desired human antibodies (see International Publication Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096 and WO 96/33735).
  • human antibody variable domains may each be expressed as a single-chain antibody (scFv) on the surface of phages by phage display technology, followed by selection of phages binding to the antigen. By analyzing genes of the selected phages, it is possible to determine DNA sequences encoding human antibody variable domains binding to the antigen. Once the DNA sequences of scFv binding to the antigen have been identified, the sequences may be used to construct appropriate expression vectors to obtain human antibodies.
  • scFv single-chain antibody
  • human antibodies produced in transgenic animals and the like are also preferred.
  • the antibody as used herein encompasses antibody fragments including Fab, (Fab′) 2 , Fc, Fc′ and Fd, as well as reshaped antibodies including monovalent or polyvalent single chain antibodies (scFV).
  • physiologically active protein-containing sample or an “antibody-containing sample” is preferably intended to mean a culture medium of mammalian cells (e.g., CHO cells) containing physiologically active protein molecules or antibody molecules produced by culture, which may further be subjected to partial purl cation or other certain treatment(s).
  • mammalian cells e.g., CHO cells
  • physiologically active protein molecules or antibody molecules produced by culture which may further be subjected to partial purl cation or other certain treatment(s).
  • impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • Any substance may be removed as an impurity by the method of the present invention as long as it is not a target protein to be purified.
  • impurities include DNA contaminants, viruses, Protein A (eluted from columns), endotoxins, HCP (host cell-derived proteins), as well as medium components Hy-Fish(FL) and IGF, with DNA contaminants or viruses being preferred.
  • DNA contaminants is intended to mean DNA molecules present in a physiologically active protein-containing sample. Examples include host-derived DNAs and contamination-derived viral DNAs.
  • RNA viruses include retroviruses (e.g., X-MuLV), reoviruses (e.g., Reo 3) and parvoviruses (e.g., MVM).
  • retroviruses e.g., X-MuLV
  • reoviruses e.g., Reo 3
  • parvoviruses e.g., MVM
  • viruses removed by the method of the present invention include, for example, X-MuLV, PRV, Reo 3, MVM, VSV, herpes simplex, CHV, Sindbis, mumps, vaccinia, Measle, Rubella, influenza, herpes zoster, cytomegalo, parainfluenza, EB, HIV, HA, HE, NANB, ATL, ECHO and parvovirus, with X-MuLV, Reo 3, MVM and PRV being preferred.
  • aqueous solution of low conductivity is generally intended to mean an aqueous solution which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m.
  • the isoelectric point of a physiologically active protein refers to the pH value at which the physiologically active protein has no apparent net charge in an aqueous solution.
  • the isoelectric point can be determined in a manner known to those skilled in the art, for example, by means of isoelectric focusing in which a physiologically active protein is electrophoresed in solutions of various pH levels to determine the pH at which the protein will not migrate.
  • a pH equal to or lower than the isoelectric point of a physiologically active protein is preferably a pH below the isoelectric point of the physiologically active protein.
  • the pH is preferably adjusted to a level equal to or lower than the isoelectric point of a physiologically active protein, so that the physiologically active protein is positively charged and the DNA molecules are negatively charged.
  • DNA has very strong negative ion charges resulting from phosphate groups in the backbone (phosphate groups found within strongly acidic phosphodiester bonds in nucleic acids have a pK value of about 1). For this reason, DNA can be negatively charged at any pH and it is possible to use a desired pH in the range of equal to or lower than the isoelectric point of a physiologically active protein. Since the pH level required will vary among different types of physiologically active proteins, those skilled in the art may select a desired pH level in the range of equal to or lower than the isoelectric point of a physiologically active protein in a known manner, for example, by preparing multiple samples with different pHs and measuring their parameters such as % DNA removal and % protein recovery, as described in the Example section below. Such a pH is usually pH 2.0 or higher, preferably pH 3.0 or higher, and particularly preferably pH 4.0 or higher.
  • ETC electrophoretic titration curve
  • a physiologically active protein-containing sample may also be formed into an acidic or alkaline aqueous solution of low conductivity, followed by adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
  • impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • Impurities removed by the method of the present invention are as described above.
  • the term “acidic aqueous solution of low conductivity” is intended to mean an aqueous solution of pH 2.0 to pH 3.9, preferably of pH 2.0 to pH 3.0, which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m.
  • the acidic aqueous solution may be selected from aqueous solutions of hydrochloric acid, citric acid, acetic acid and other acids.
  • the type, conductivity and pH of acidic aqueous solution of low conductivity will vary depending on the type of physiologically active protein or antibody to be purified. Those skilled in the art will readily determine optimal conditions for these parameters in preliminary experiments as described herein.
  • alkaline aqueous solution of low conductivity is intended to mean an aqueous solution usually of pH 7.5 to pH 13, which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m.
  • the pH of this solution will vary depending on the type of physiologically active protein or antibody to be purified.
  • a physiologically active protein-containing sample is formed into an acidic or alkaline aqueous solution of low conductivity
  • the resulting sample is adjusted with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
  • a buffer include Tris-HCl, phosphate, Tris, Na 2 HPO 4 and NaOH.
  • an antibody-containing sample may usually be subjected to affinity chromatography on Protein A or G and eluted with an acidic aqueous solution of low conductivity, followed by adjusting the resulting eluate with a buffer to a desired pH in the range of equal to or lower than the isoelectric point of the physiologically active protein.
  • impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • Impurities removed by the method of the present invention are as described above.
  • the acidic aqueous solution of low conductivity used in this method may be any of those listed above.
  • a buffer include Tris-HCl, phosphate, Tris, Na 2 HPO 4 and NaOH.
  • the solution adjusted to a pH equal to or lower than the isoelectric point of the physiologically active protein in the above step produces particles (i.e., becomes clouded), These particles may be removed by filtration through a filter to ensure efficient removal of impurities such as DNA contaminants.
  • a filter available for filtration include, but are not limited to, a 1.0-0.2 ⁇ m Cellulose Acetate Filter System (Corning) or TFF.
  • these particles may also be removed by centrifugation or any other techniques for efficient particle removal; procedures for removal are not limited to filtration through a filter.
  • the inventors of the present invention estimate that when impurities are DNA molecules, each of these particles is a conjugate formed between physiologically active protein and DNA. They also estimate that when the pH is adjusted below the isoelectric point of a protein, the protein is positively charged and DNA molecules are negatively charged, resulting in conjugation between DNA and protein. Moreover, the conversion into an aqueous solution of low conductivity will further enhance conjugation. Particle removal by filtration results in a small loss of physiologically active protein because it is removed in the form of DNA-physiologically active protein conjugates. However, such a small loss constitutes only a few percent of the total amount of the physiologically active protein; about 90% of the physiologically active protein can be recovered, as will be described in the Example section below.
  • the inventors of the present invention also estimate that Protein A/G column chromatography alone may not be sufficient to ensure effective separation between DNA contaminant and physiologically active protein because DNA-protein conjugates are formed on the column resin.
  • the physiologically active protein thus purified is available for use as a pharmaceutical formulation after further purification by cation-exchange chromatography, anion-exchange chromatography, hydroxyapatite chromatography, or combinations thereof.
  • Quantitative DNA assay may be accomplished by, but not limited to, Threshold Total DNA assay along with DNA extraction prior to the assay.
  • Quantitative virus assay may be accomplished by, but not limited to, TCID 50 (tissue culture infective dose (50%)) assay which is measured by viral infectivity in detection cells, in combination with RT/Q-PCR and Q-PCR which allow determination of the virus amount in fractions.
  • TCID 50 tissue culture infective dose (50%)
  • Example 1 Investigation of Buffer Composition for Protein A Affinity Chromatography in Purifying hPM-1 (Humanized Anti-IL-6 Receptor Antibody)
  • Test Material Antibody-Containing Sample
  • CM culture medium
  • hPM-1 antibody humanized anti-IL-6 receptor antibody
  • CA Cellulose Acetate
  • CORNING Cellulose Acetate
  • HPLC L-6200 Intelligent Pump (HITACHI)
  • hPM-1 is assayed by reversed-phase HPLC on a PLRP-S column (Polymer Laboratories) with a linear gradient.
  • DNA is measured by Threshold Total DNA assay. Prior to the assay, DNA extraction is performed (e.g., using a DNA extracter kit, Wako Pure Chemicals Industries, Ltd.). Likewise, a Threshold Total DNA assay kit (Molecular Devices) is used for the measurement.
  • a DNA extracter kit Wako Pure Chemicals Industries, Ltd.
  • a Threshold Total DNA assay kit (Molecular Devices) is used for the measurement.
  • Each test sample is monitored for particle formation by measuring its absorbance at 660 nm in a spectrophotometer U-2000 (HITACHI).
  • Elution conditions were investigated at various buffer compositions for elution in Protein A affinity chromatography by measuring % recovery of hPM-1 and DNA removal by elution.
  • the above antibody-containing sample was subjected to the column under the conditions indicated in Table 1 below.
  • Protein A resin was equilibrated with the equilibration buffer indicated in Table 1 and then loaded with the above antibody-containing sample, followed by Washing 1, Washing 2 and elution.
  • the elution profile was monitored at A280 nm to isolate a protein peak.
  • C-P Buffer denotes citrate-phosphate buffer.
  • Each elution fraction was adjusted to pH 7.0 with a 300 mM Tris solution, indicating that particles were generated in the fractions eluted with HCl (Elution methods 2 and 3). Further investigation was performed to determine the correlation between particle formation and % recovery of hPM-1 or the amount of residual DNA.
  • each Protein A elution fraction supplemented with NaCl was adjusted to pH 7.0 with a 300 mM Tris solution and then filtered or unfiltered through a 0.22 ⁇ m CA Filter. The filtered and unfiltered samples were measured for % recovery of hPM-1 (filtered samples only) and the amount of residual DNA.
  • the % recovery of hPM-1 was measured for the individual elution methods. As a result, the % recovery was as high as 98.6% in Elution method 1. In contrast, the % recovery ranged from 83.8% to 97.1% in Elution method 2 and from 83.5% to 93.7% in Elution method 3; these variations were estimated to be due to the smallness of examination scale (resin volume: 0.4 ml). When the purification scale was increased, it was confirmed that the % recovery of hPM-1 was stabilized at 90% or more (Elution method 2). Thus, the % recovery of hPM-1 was also found to remain high even in HCl elution.
  • Table 2 summarizes the analysis of the correlation between NaCl concentrations in the HCl eluate and various factors.
  • the % recovery of hPM-1 was 88% at 100 mM NaCl, 86% at 50 mM NaCl and 81% at 0 mM NaCl.
  • the amount of residual DNA was low at 0 mM NaCl in both filtered and unfiltered samples.
  • the filtered sample supplemented with 0 mM NaCl had a very low DNA content of 11 pg DNA/mg hPM-1.
  • a sample containing a humanized anti-PTHrP antibody (a culture medium from CHO cell culture, filtered through 0.45 and 0.2 ⁇ m CA SARTOBRAN P filters (sartorius)) was purified by Protein A affinity column chromatography under the conditions indicated below.
  • the anti-PTHrP antibody was prepared as described in International Publication No. WO98/13388 (isoelectric point: pH 8.3).
  • Resin rProtein A Sepharose Fast Flow Load: direct load of the culture medium (pH 6.6 to pH 7.5) Adjustment of elution fraction: elution fractions are adjusted to various pH levels with a 1 N aqueous Tris solution and then filtered through a 0.2 ⁇ m Cellulose Acetate (hereinafter abbreviated as CA) to remove DNA (the conditions are examined in (1) below).
  • CA Cellulose Acetate
  • the Protein A column was sufficiently equilibrated with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) and then loaded with the above antibody-containing CM. Subsequently, the column was washed with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) to remove unbound impurities, further washed with citrate-phosphate buffer (pH 7.5) to decrease the conductivity, and then eluted with 20 mM aqueous citric acid. The elution profile was monitored at A280 nm to isolate a protein peak. This Protein A elution fraction was used for the following examination of conditions.
  • the Protein A elution fraction was adjusted with a 1.0 M aqueous Tris solution to the following pH levels: 2.7 (unadjusted), 4.0, 4.5, 5,0, 5.5, 6.0, 6.5, 7.0 and 7.5. Subsequently, each sample was allowed to stand for a given period of time, filtered through a 0.22 ⁇ m CA filter, and then adjusted to pH 7 with a 1.0 M aqueous Tris solution, followed by DNA assay. Table 3 lists the examined pH levels and standing periods, along with the amount of residual DNA.
  • the amount of residual DNA was below the detection limit at pH 5.5 and pH 6.0 in all cases where the samples were allowed to stand for 0, 6 and 24 hours. Also, the removal of residual DNA reached a peak around pH 5.5 and pH 6.0, whereas decreased efficiency of DNA, removal was observed at higher and lower pH levels.
  • a sample containing a humanized anti-HM1.24 antigen monoclonal antibody (a culture medium from CHO cell culture) was purified by Protein A affinity column chromatography under the conditions indicated in Table 4 below.
  • the anti-HM1.24 antigen monoclonal antibody was prepared as described in International Publication No. WO98/14580 (isoelectric point: pH 9.0).
  • the Protein A column was sufficiently equilibrated with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) and then loaded with the above antibody-containing CM, Subsequently, the column was washed with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) to remove unbound impurities, further washed with citrate-phosphate buffer (pH 7.5) to decrease the conductivity, and then eluted with 20 mM aqueous citric acid. The elution profile was monitored at A280 nm to isolate a protein peak. This Protein A elation fraction was used for the following investigation of conditions.
  • Experiment 1 indicated that the amount of DNA decreased with decrease in pH in the order of pH 7.5, pH 6.5 and pH 5.5, and that there was a tendency to remove more DNA at 0 hours than at 6 hours.
  • the calculation of % recovery indicated little loss of the humanized anti-HM1.24 antigen monoclonal antibody.
  • G-CSF Granulocyte Colony-Stimulating Factor
  • a G-CSF-containing sample (from CHO cell culture; Chugai Pharmaceutical Co., Ltd.) was used for the following examination of conditions (isoelectric point: pH 5.5-5.7).
  • the optimal pH for filtration through a filter was investigated.
  • the G-CSF-containing sample was diluted in an acidic solution of low conductivity (2.5 mM aqueous HCl) and further formed into an acidic aqueous solution of low conductivity using 20% hydrochloric acid, followed by addition of sample DNA.
  • CM culture medium
  • hPM-1 antibody humanized IL-6 receptor antibody
  • the virus-supplemented samples prepared in 5.1 were purified by rProtein Column Chromatography. Detailed conditions are as shown below.
  • the elution fractions obtained in 5.2 were adjusted to pH 3.2 ⁇ 0.1 with 1 mol/L hydrochloric acid and held at a room temperature of 15 ⁇ 5° C. for 30 minutes or longer. Subsequently, each elution fraction was adjusted to pH 7.2 ⁇ 0.1 with a 300 mmol/L Tris solution, 40.0 mL of which was then filtered under a pressure of 0.03 ⁇ 0.01 MPa using a filtration unit equipped with a glass fiber filter (Millipore) (0.2 ⁇ m, PALL) connected to the primary side and a BioInert (0.2 ⁇ m, PALL) (a PALL filter holder equipped with a ⁇ 15 mm adjuster) connected to the secondary side.
  • a glass fiber filter 0.2 ⁇ m, PALL
  • BioInert 0.2 ⁇ m, PALL
  • TCID 50 assay All the samples collected were measured by TCID 50 assay. In the clearance capacity test for X-MuLV and MVM, these viruses were detected not only by TCID assay which was measured by viral infectivity in detection cells, but also by RT/Q-PCR and Q-PCR which allowed determination of the virus amount in fractions.
  • the purification process of the present invention achieves very high LRVs (Logarithmic Reduction Values) for all the tested viruses and this examination confirms that the viruses were removed to a level below the assay limit of detection after low pH treatment and filtration.
  • LRVs Logarithmic Reduction Values
  • the method of the present invention enables efficient removal of impurities such as DNA contaminants and viruses in a very simple manner, and is significantly advantageous in purifying physiologically active proteins, especially antibodies.
  • the method achieves an extremely low DNA concentration (e.g., 22.5 pg/ml) when impurities are DNA molecules, while it achieves an extremely low virus titer (e.g., 1.03 (expressed in Log 10 /mL), as measured by TCID 50 assay) when impurities are viruses.
  • the method of the present invention also enables cost reduction and has great significance in this field.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Engineering & Computer Science (AREA)
  • Water Supply & Treatment (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A method is provided for purifying physiologically active proteins, especially antibodies, in order to remove impurities such as DNA contaminants and viruses with minimal loss of physiologically active proteins. The physiologically active protein is introduced into an aqueous solution of low conductivity at a pH of below the isoelectric point of the physiologically active protein to precipitate impurities as particles. The particles are removed, leaving a purified physiologically active protein.

Description

    TECHNICAL FIELD
  • The present invention relates to a method for purifying proteins, more specifically to a method for removing impurities such as DNA contaminants from a sample containing a physiologically active protein such as antibody molecules.
  • BACKGROUND ART
  • Advances in gene recombinant technology have enabled a stable supply of various protein formulations. In particular, a variety of recombinant antibody drugs, which are more selective than normal drugs, have been developed and entered clinical trial in recent years.
  • In these recombinantly-produced physiologically active protein-containing formulations, there is a need to remove host DNA and impurities (e.g., DNA contaminants) associated with viral contamination. Under present World Health Organization (WHO) criteria, the amount of DNA in biological drugs should not exceed 100 pg DNA/dose. To meet this criteria, in general, an aqueous medium containing host cell-derived physiologically active proteins is treated by anion-exchange chromatography, hydroxyapatite chromatography or a combination thereof, for the purpose of removing DNA.
  • In particular, in a case where a physiologically active protein is an antibody produced recombinantly in mammalian host cells, the aqueous medium is treated by affinity column chromatography on Protein A or G before being purified by various types of chromatography, based on the binding property of Protein A or Protein G to IgG Fc chain.
  • By way of example, in JP KOHYO 5-504579, an antibody-containing aqueous medium obtained from mammalian cell culture is subjected to Protein A/G column chromatography to adsorb antibody molecules onto the column, followed by elution with an acidic solution (about 0.1 M citric acid, pH 3.0-3.5) to release the antibody molecules. The resulting acidic eluate is subjected sequentially to ion-exchange column chromatography and size exclusion column chromatography to give the purified antibody molecules.
  • However, these individual chromatographic processes and combinations thereof are time-, labor- and cost-consuming, as well as being complicated. Moreover, they fail to provide stable results.
  • Thus, the object of the present invention is to provide a simpler and less expensive method for purifying physiologically active proteins, especially antibodies, which can ensure removal of impurities such as DNA contaminants and viruses, and which can minimize a loss of physiologically active proteins.
  • DISCLOSURE OF THE INVENTION
  • As a result of extensive and intensive efforts made to overcome these problems, the inventors of the present invention have made the surprising finding that impurities such as DNA contaminants and viruses can be efficiently removed from a physiologically active protein-containing sample without using complicated chromatographic processes when the sample is formed into an aqueous solution of low conductivity at a pH below the isoelectric point of the physiologically active protein and then filtrated through a filter to remove the resulting particles. This finding led to the completion of the present invention.
  • Namely, the present invention provides the following.
  • (1) A method for removing impurities in a physiologically active protein-containing sample, which comprises the steps of
  • 1) forming the physiologically active protein-containing sample into an aqueous solution of low conductivity having a pH equal to or lower than the isoelectric point of the physiologically active protein; and
  • 2) removing the resulting particles.
  • (2) The method according to (1) above, wherein the aqueous solution of low conductivity has a conductivity of 0 to 100 mM, as expressed in molarity.
    (3) The method according to (1) or (2) above, wherein the aqueous solution of low conductivity has an ionic strength of 0 to 0.2.
    (4) The method according to any one of (1) to (3) above, wherein the aqueous solution of low conductivity has a conductivity of 0 to 300 mS/m.
    (5) The method according to any one of (1) to (4) above, wherein the solution is selected from aqueous solutions of hydrochloric acid, citric acid and acetic acid.
    (6) The method according to any one of (1) to (5) above, wherein the pH of the aqueous solution is equal to or lower than the isoelectric point of the physiologically active protein and equal to or higher than pH 2.0.
    (7) The method according to any one of (1) to (6) above, wherein the impurities are DNA contaminants.
    (8) The method according to any one of (1) to (6) above, wherein the impurities are viruses.
    (9) The method according to (7) above, wherein the physiologically active protein-containing sample has the DNA contaminants at a DNA concentration of 22.5 pg/ml or less after the treatment of removal of DNA contaminants.
    (10) The method according to any one of (1) to (9) above, wherein the physiologically active protein is an antibody.
    (11) The method according to (10) above, wherein the antibody is an IgG antibody.
    (12) The method according to (10) or (11) above, wherein the antibody is a humanized monoclonal antibody.
    (13) The method according to (12) above, wherein the antibody is a humanized anti-IL-6 receptor antibody.
    (14) The method according to (12) above, wherein the antibody is a humanized anti-HM1.24 antigen monoclonal antibody.
    (15) The method according to (12) above, wherein the antibody is a humanized anti-parathyroid hormone-related peptide antibody (anti-PTHrP antibody).
    (16) The method according to any one of (1) to (9) above, wherein the physiologically active protein is granulocyte colony-stimulating factor.
    (17) The method according to any one of (1) to (16) above, wherein the particles are removed by filtration through a filter.
    (18) The method according to (1) above, wherein step 1) is accomplished by forming the physiologically active protein-containing sample into an acidic or alkaline aqueous solution of low conductivity, and adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
    (19) The method according to (1) above,
  • wherein the physiologically active protein is an antibody, and
  • wherein step 1) is accomplished by subjecting the antibody-containing sample to affinity chromatography on Protein A or G, eluting the sample with an acidic aqueous solution of low conductivity, and adjusting the resulting eluate with a buffer to a pH equal to or lower than the isoelectric point of the antibody.
  • (20) The method according to (18) or (19) above, wherein the buffer is an aqueous solution of Tris.
    (21) A purified physiologically active protein obtainable by the method according to any one of (1) to (20) above.
    (22) A method for manufacturing a medical protein formulation, which comprises a purification step in which the method according to any one of (1) to (20) above is used.
  • BEST MODE FOR CARRYING OUT THE INVENTION
  • Examples of a physiologically active protein contained in a sample to be purified by the method of the present invention include, but are not limited to, hematopoietic factors such as granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), erythropoietin (EPO) and thrombopoietin, cytokines such as interferons, IL-1 and IL-6, monoclonal antibodies, tissue plasminogen activator (TPA), urokinase, serum albumin, blood coagulation factor VIII, leptin, insulin, and stem cell growth factor (SCF). Among these proteins, preferred are G-CSF and antibodies including monoclonal antibodies , and more preferred are monoclonal antibodies. In an embodiment of the present invention using Protein A/G affinity chromatography, monoclonal antibodies are preferred for purification. Antibodies are categorized into IgG, IgA, IgE, IgD and IgM classes, with IgG antibodies being preferred.
  • The term “physiologically active protein” is intended to mean a protein having substantially the same biological activities as a corresponding physiologically active protein of mammalian (especially human) origin. Such a protein may either be native or genetically recombinant, preferably genetically recombinant. Genetically recombinant physiologically active proteins may be prepared by production in bacterial cells such as E. coli; yeast cells; or animal-derived cultured cells such as Chinese hamster ovary (CHO) cells, C127 cells or COS cells. The proteins thus prepared are isolated and purified in various manners before use. Such genetically recombinant proteins encompass those having the same amino acid sequence as the corresponding native protein, as well as those comprising deletion, substitution or addition of one or more amino acids in the amino acid sequence, but retaining the biological activities mentioned above. Further, such proteins include those chemically modified with PEG, etc.
  • When a physiologically active protein is a glycoprotein, it may have sugar chains of any origin, preferably of mammalian origin. Mammalian origins include, for example, Chinese hamster ovary (CHO) cells, BHK cells, COS cells and human-derived cells, with CHO cells being most preferred.
  • When a physiologically active protein is EPO, it may be prepared in any manner, for example, by obtaining from human urine in various manners or by producing with genetic engineering techniques in bacterial cells such as E. coli, yeast cells, Chinese hamster ovary (CHO) cells, BHK cells, COS cells, human-derived cells or the like (e.g., as described in JP KOKAI 61-12288). EPO thus prepared is extracted, isolated, and purified in various manners before use. In addition, EPO may be chemically modified with PEG, etc. (see International Publication No. WO90/12874). EPO as used herein further includes those originally unglycosylated but chemically modified with PEG, etc. Likewise, EPO analogs are also included, which are modified to have at least one additional site for N-linked or O-linked glycosylation in the amino acid sequence of EPO (see, e.g., JP KOKAI 08-151398, JP KOHYO 08-506023). Instead of increasing the number of glycosylation sites, EPO analogs may also be modified to have an increased content of sugar chains such as sialic acid for an increased amount of sugar chains.
  • When a physiologically active protein is G-CSF, any G-CSF can be used as long as it is highly purified. G-CSF as used herein may be prepared in any manner, for example, by obtaining from cultured human tumor cell lines or by producing with genetic engineering techniques in bacterial cells such as E. coli; yeast cells; or animal-derived cultured cells such as Chinese hamster ovary (CHO) cells, C127 cells or COS cells. G-CSF thus prepared is extracted, isolated, and purified in various manners before use. Preferred are those produced recombinantly in E. coli cells, yeast cells or CHO cells. The most preferred are those produced recombinantly in CHO cells. In addition, G-CSF may be chemically modified with PEG, etc. (see International Publication No. WO90/12874).
  • When a physiologically active protein is a monoclonal antibody, it may be prepared in any manner. In principle, a monoclonal antibody can be produced using known techniques by immunizing a sensitizing antigen in accordance with conventional procedures for immunization, fusing the resulting immunocytes with known parent cells through conventional procedures for cell fusion, and then screening monoclonal antibody-producing cells through conventional procedures for screening.
  • Alternatively, antibody genes are cloned from hybridomas, integrated into appropriate vectors, and then transformed into hosts to produce antibody molecules using gene recombination technology. The genetically recombinant antibodies thus produced may also be used in the present invention (see, e.g., Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). More specifically, cDNA of antibody variable domains (V domains) is synthesized from hybridoma mRNA using reverse transcriptase. Upon obtaining DNA encoding the target antibody V domains, the DNA is ligated to DNA encoding desired antibody constant domains (C domains) and integrated into an expression vector. Alternatively, the DNA encoding the antibody V domains may be integrated into an expression vector carrying the DNA of the antibody C domains. The DNA construct is integrated into an expression vector such that it is expressed under control of an expression regulatory region, e.g., an enhancer or a promoter. Host cells are then transformed with this expression vector for antibody expression.
  • In the present invention, it is possible to use genetically recombinant antibodies (e.g., chimeric antibodies, humanized antibodies) that are artificially modified with a view to attenuating the characteristics as heteroantigen to human. These modified antibodies may be prepared in a known manner. A chimeric antibody is composed of variable domains of heavy and light chains from a non-human mammalian (e.g., mouse) antibody and constant domains of heavy and light chains from a human antibody. To obtain chimeric antibodies, DNAs encoding such mouse antibody variable domains may be ligated to DNAs encoding the human antibody constant domains, and then integrated into an expression vector, followed by transformation into a host for antibody production.
  • Humanized antibodies are also called reshaped human antibodies and are obtained by grafting complementarity determining regions (CDRs) of non-human mammalian (e.g., mouse) antibodies to replace those of human antibodies. Standard gene recombination procedures for this purpose are also known. More specifically, a DNA sequence designed to allow ligation between CDRs of mouse antibody and framework regions (FRs) of human antibody is synthesized by PCR from several oligonucleotides which are prepared to have sections overlapping with one another at the ends. The DNA thus obtained is ligated to DNA encoding human antibody constant domains, and integrated into an expression vector, followed by transformation into a host for antibody production (see European Patent Publication No, EP 239400 and International Publication No. WO 96/02576). The FRs of human antibody, which is ligated via CDRs, are selected such that the complementarity determining regions form a favorable antigen-binding site. If necessary, amino acid substitutions may be made in the framework regions of antibody variable domains such that the complementarity determining regions of reshaped humanized antibody may form an appropriate antigen-binding site (Sato, K. et al., Cancer Res. (1993) 53, 851-856).
  • A humanized anti-IL-6 receptor antibody (hPM-1) can be presented as a preferred example for such reshaped humanized antibodies (see International Publication No. WO92-19759). In addition to this, a humanized anti-HM1.24 antigen monoclonal antibody (see International Publication No. WO98-14580), a humanized anti-parathyroid hormone-related peptide antibody (anti-PTHrP antibody; see International Publication No. WO98-13388), a humanized anti-tissue factor antibody (see International Publication No. WO99-51743) and the like are also preferred for use in the present invention.
  • Procedures for obtaining human antibodies are also known. For example, human lymphocytes are sensitized in vitro with a desired antigen or a desired antigen-expressing cell, and the sensitized lymphocytes are then fused with human myeloma cells (e.g., U266) to give desired human antibodies having binding activity to the antigen (see JP KOKOKU 01-59878). Alternatively, transgenic animals having the entire repertories of human antibody genes may be immunized with an antigen to obtain desired human antibodies (see International Publication Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096 and WO 96/33735). There are additional techniques using human antibody libraries to give human antibodies by panning. For example, human antibody variable domains may each be expressed as a single-chain antibody (scFv) on the surface of phages by phage display technology, followed by selection of phages binding to the antigen. By analyzing genes of the selected phages, it is possible to determine DNA sequences encoding human antibody variable domains binding to the antigen. Once the DNA sequences of scFv binding to the antigen have been identified, the sequences may be used to construct appropriate expression vectors to obtain human antibodies. These techniques are already well known and can be found in WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438 and WO 95/15388.
  • Further, human antibodies produced in transgenic animals and the like are also preferred.
  • Furthermore, the antibody as used herein encompasses antibody fragments including Fab, (Fab′)2, Fc, Fc′ and Fd, as well as reshaped antibodies including monovalent or polyvalent single chain antibodies (scFV).
  • As used herein, the term “physiologically active protein-containing sample” or an “antibody-containing sample” is preferably intended to mean a culture medium of mammalian cells (e.g., CHO cells) containing physiologically active protein molecules or antibody molecules produced by culture, which may further be subjected to partial purl cation or other certain treatment(s).
  • In a preferred embodiment of the present invention, impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • 1) forming the physiologically active protein-containing sample into an aqueous solution of low conductivity at a pH equal to or lower than the isoelectric point of the physiologically active protein; and
  • 2) removing the resulting particles.
  • Any substance may be removed as an impurity by the method of the present invention as long as it is not a target protein to be purified. Examples of such impurities include DNA contaminants, viruses, Protein A (eluted from columns), endotoxins, HCP (host cell-derived proteins), as well as medium components Hy-Fish(FL) and IGF, with DNA contaminants or viruses being preferred. As used herein, the term “DNA contaminants” is intended to mean DNA molecules present in a physiologically active protein-containing sample. Examples include host-derived DNAs and contamination-derived viral DNAs.
  • There is no particular limitation on the type of virus to be removed by the method of the present invention. Any virus, including DNA and RNA viruses, may be removed. Examples of RNA viruses include retroviruses (e.g., X-MuLV), reoviruses (e.g., Reo 3) and parvoviruses (e.g., MVM). Illustrative examples of viruses removed by the method of the present invention include, for example, X-MuLV, PRV, Reo 3, MVM, VSV, herpes simplex, CHV, Sindbis, mumps, vaccinia, Measle, Rubella, influenza, herpes zoster, cytomegalo, parainfluenza, EB, HIV, HA, HE, NANB, ATL, ECHO and parvovirus, with X-MuLV, Reo 3, MVM and PRV being preferred.
  • As used herein, the term “aqueous solution of low conductivity” is generally intended to mean an aqueous solution which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m.
  • The isoelectric point of a physiologically active protein refers to the pH value at which the physiologically active protein has no apparent net charge in an aqueous solution. The isoelectric point can be determined in a manner known to those skilled in the art, for example, by means of isoelectric focusing in which a physiologically active protein is electrophoresed in solutions of various pH levels to determine the pH at which the protein will not migrate. A pH equal to or lower than the isoelectric point of a physiologically active protein is preferably a pH below the isoelectric point of the physiologically active protein.
  • When impurities are DNA molecules in the method of the present invention, the pH is preferably adjusted to a level equal to or lower than the isoelectric point of a physiologically active protein, so that the physiologically active protein is positively charged and the DNA molecules are negatively charged.
  • In general, DNA has very strong negative ion charges resulting from phosphate groups in the backbone (phosphate groups found within strongly acidic phosphodiester bonds in nucleic acids have a pK value of about 1). For this reason, DNA can be negatively charged at any pH and it is possible to use a desired pH in the range of equal to or lower than the isoelectric point of a physiologically active protein. Since the pH level required will vary among different types of physiologically active proteins, those skilled in the art may select a desired pH level in the range of equal to or lower than the isoelectric point of a physiologically active protein in a known manner, for example, by preparing multiple samples with different pHs and measuring their parameters such as % DNA removal and % protein recovery, as described in the Example section below. Such a pH is usually pH 2.0 or higher, preferably pH 3.0 or higher, and particularly preferably pH 4.0 or higher.
  • To confirm whether DNA molecules are negatively charged, known procedures may be used such as those using an electrophoretic titration curve (ETC) (see Ion Exchange Chromatography Principles and Methods, Pharmacia (latterly Amersham Biosciences), pp. 52-56).
  • Moreover, in the method of the present invention, a physiologically active protein-containing sample may also be formed into an acidic or alkaline aqueous solution of low conductivity, followed by adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
  • Thus, in another preferred embodiment of the present invention, impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • 1) forming the physiologically active protein-containing sample into an acidic or alkaline aqueous solution of low conductivity;
  • 2) adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein; and
  • 3) removing the resulting particles.
  • Impurities removed by the method of the present invention are as described above.
  • As used herein, the term “acidic aqueous solution of low conductivity” is intended to mean an aqueous solution of pH 2.0 to pH 3.9, preferably of pH 2.0 to pH 3.0, which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m. The acidic aqueous solution may be selected from aqueous solutions of hydrochloric acid, citric acid, acetic acid and other acids. The type, conductivity and pH of acidic aqueous solution of low conductivity will vary depending on the type of physiologically active protein or antibody to be purified. Those skilled in the art will readily determine optimal conditions for these parameters in preliminary experiments as described herein.
  • Likewise, the term “alkaline aqueous solution of low conductivity” as used herein is intended to mean an aqueous solution usually of pH 7.5 to pH 13, which has a molarity of 0 to 100 mM, preferably 0 to 50 mM, more preferably 0 to 30 mM, or which has an ionic strength of 0 to 0.2, preferably 0 to 0.12, or which has a conductivity of 0 to 300 mS/m, preferably 0 to 200 mS/m, more preferably 0 to 150 mS/m. The pH of this solution will vary depending on the type of physiologically active protein or antibody to be purified.
  • In the method of the present invention, after a physiologically active protein-containing sample is formed into an acidic or alkaline aqueous solution of low conductivity, the resulting sample is adjusted with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein. Examples of a buffer include Tris-HCl, phosphate, Tris, Na2HPO4 and NaOH.
  • Moreover, in the present invention, in certain cases such as where a physiologically active protein is an antibody, an antibody-containing sample may usually be subjected to affinity chromatography on Protein A or G and eluted with an acidic aqueous solution of low conductivity, followed by adjusting the resulting eluate with a buffer to a desired pH in the range of equal to or lower than the isoelectric point of the physiologically active protein.
  • Thus, in yet another preferred embodiment of the present invention, impurities in a physiologically active protein-containing sample are removed by a method comprising the steps of:
  • 1) subjecting an antibody-containing sample to affinity chromatography on Protein A or G and eluting the sample with an acidic aqueous solution of low conductivity;
  • 2) adjusting the resulting eluate with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein; and
  • 3) removing the resulting particles.
  • Impurities removed by the method of the present invention are as described above.
  • The acidic aqueous solution of low conductivity used in this method may be any of those listed above. Examples of a buffer include Tris-HCl, phosphate, Tris, Na2HPO4 and NaOH.
  • In the method of the present invention, the solution adjusted to a pH equal to or lower than the isoelectric point of the physiologically active protein in the above step, in turn, produces particles (i.e., becomes clouded), These particles may be removed by filtration through a filter to ensure efficient removal of impurities such as DNA contaminants. Examples of a filter available for filtration include, but are not limited to, a 1.0-0.2 μm Cellulose Acetate Filter System (Corning) or TFF.
  • Alternatively, these particles may also be removed by centrifugation or any other techniques for efficient particle removal; procedures for removal are not limited to filtration through a filter.
  • Without being bound by any particular theory, the inventors of the present invention estimate that when impurities are DNA molecules, each of these particles is a conjugate formed between physiologically active protein and DNA. They also estimate that when the pH is adjusted below the isoelectric point of a protein, the protein is positively charged and DNA molecules are negatively charged, resulting in conjugation between DNA and protein. Moreover, the conversion into an aqueous solution of low conductivity will further enhance conjugation. Particle removal by filtration results in a small loss of physiologically active protein because it is removed in the form of DNA-physiologically active protein conjugates. However, such a small loss constitutes only a few percent of the total amount of the physiologically active protein; about 90% of the physiologically active protein can be recovered, as will be described in the Example section below.
  • The inventors of the present invention also estimate that Protein A/G column chromatography alone may not be sufficient to ensure effective separation between DNA contaminant and physiologically active protein because DNA-protein conjugates are formed on the column resin. The physiologically active protein thus purified is available for use as a pharmaceutical formulation after further purification by cation-exchange chromatography, anion-exchange chromatography, hydroxyapatite chromatography, or combinations thereof.
  • Quantitative DNA assay may be accomplished by, but not limited to, Threshold Total DNA assay along with DNA extraction prior to the assay.
  • Quantitative virus assay may be accomplished by, but not limited to, TCID50 (tissue culture infective dose (50%)) assay which is measured by viral infectivity in detection cells, in combination with RT/Q-PCR and Q-PCR which allow determination of the virus amount in fractions.
  • The present invention will now be further described in the following examples, which are not intended to limit the scope of the invention. Based on the detailed description, various changes and modifications will be apparent to those skilled in the art, and such changes and modifications fall within the scope of the invention.
  • EXAMPLES Example 1: Investigation of Buffer Composition for Protein A Affinity Chromatography in Purifying hPM-1 (Humanized Anti-IL-6 Receptor Antibody) 1.1. Test Procedures (1) Test Material (Antibody-Containing Sample)
  • A sample containing the culture medium (hereinafter abbreviated as CM) of CHO cells producing hPM-1 antibody (humanized anti-IL-6 receptor antibody), which had been centrifuged to remove the cells and stored at −80° C., was filtered through a 0.22 μm Cellulose Acetate (abbreviated as CA) Filter System (CORNING) and used as a test sample for purification investigation. The hPM-1 antibody was prepared as described in Reference Example 2 of JP KOKAI 08-99902 using the human elongation factor Iα promoter shown in Example 10 of International Publication No. WO92/19759 (isoelectric point: pH 9.0).
  • (2) Instrument Used for Examination For HCl Eluate HPLC: L-6200 Intelligent Pump (HITACHI)
      • L-4200 UV-VIS Detector (HITACHI)
      • D-2500 Chromato-Integrator (HITACHI)
    Column: HR5/2 (Pharmacia), 5 mm I.D.×20 mm H Media: POROS 50A (PerSeptive), 0.4 ml
      • Lot; A250-039, Code; SPECIAL
    For Particles HPLC: Waters PrepLC4000 System (Waters)
      • Waters2000 System Controller (Waters)
      • Waters486 Tunable Absorbance Detector (Waters)
      • Waters741 Data Module (Waters)
    Spectrophotometer: U-2000 (HITACHI) Column: XK26 (Pharmacia), 26 mm I.D.×100 mm H Media: POROS 50A (PerSeptive), 53 ml
      • Lot; A250-039, Code; SPECIAL
    (3) Analysis and Assay
  • hPM-1 assay:
  • hPM-1 is assayed by reversed-phase HPLC on a PLRP-S column (Polymer Laboratories) with a linear gradient.
  • DNA assay:
  • DNA is measured by Threshold Total DNA assay. Prior to the assay, DNA extraction is performed (e.g., using a DNA extracter kit, Wako Pure Chemicals Industries, Ltd.). Likewise, a Threshold Total DNA assay kit (Molecular Devices) is used for the measurement.
  • Turbidimetry:
  • Each test sample is monitored for particle formation by measuring its absorbance at 660 nm in a spectrophotometer U-2000 (HITACHI).
  • 1.2. Investigation of Elution Conditions
  • Elution conditions were investigated at various buffer compositions for elution in Protein A affinity chromatography by measuring % recovery of hPM-1 and DNA removal by elution. The above antibody-containing sample was subjected to the column under the conditions indicated in Table 1 below. Protein A resin was equilibrated with the equilibration buffer indicated in Table 1 and then loaded with the above antibody-containing sample, followed by Washing 1, Washing 2 and elution. The elution profile was monitored at A280 nm to isolate a protein peak. In the table, C-P Buffer denotes citrate-phosphate buffer.
  • TABLE 1
    Elution Elution Elution
    method 1 method 2 method 3
    Equilibration 1M NaCl/ 1M NaCl/ 1M NaCl/
    100 mM 10 mM 100 mM
    C-P Buffer C-P Buffer C-P Buffer
    pH 7.5 pH 7.5 pH 7.5
    Washing 1 1M NaCl/ 1M NaCl/ 1M NaCl/
    100 mM 10 mM 100 mM
    C-P Buffer C-P Buffer C-P Buffer
    pH 7.5 pH 7.5 pH 7.5
    Washing 2 100 mM 10 mM 100 mM
    C-P Buffer C-P Buffer C-P Buffer
    pH 7.5 pH 7.5 pH 7.5
    Elution 100 mM 2.5 mM 2.5 mM
    C-P Buffer HCl pH 2.6 HCl pH 2.6
    pH 2.6
  • No chromatographic difference was observed among Elution methods 1, 2 and 3.
  • Each elution fraction was adjusted to pH 7.0 with a 300 mM Tris solution, indicating that particles were generated in the fractions eluted with HCl (Elution methods 2 and 3). Further investigation was performed to determine the correlation between particle formation and % recovery of hPM-1 or the amount of residual DNA.
  • To examine the particle correlation, the HCl eluate from Elution method 2 was supplemented with NaCl and analyzed for the correlation between NaCl concentrations (0 mM, 50 mM, 100 mM) and various factors. In the analysis of the correlation between NaCl concentrations and various factors, filtered and unfiltered samples were prepared as follows: each Protein A elution fraction supplemented with NaCl was adjusted to pH 7.0 with a 300 mM Tris solution and then filtered or unfiltered through a 0.22 μm CA Filter. The filtered and unfiltered samples were measured for % recovery of hPM-1 (filtered samples only) and the amount of residual DNA.
  • 1.3. % Recovery
  • The % recovery of hPM-1 was measured for the individual elution methods. As a result, the % recovery was as high as 98.6% in Elution method 1. In contrast, the % recovery ranged from 83.8% to 97.1% in Elution method 2 and from 83.5% to 93.7% in Elution method 3; these variations were estimated to be due to the smallness of examination scale (resin volume: 0.4 ml). When the purification scale was increased, it was confirmed that the % recovery of hPM-1 was stabilized at 90% or more (Elution method 2). Thus, the % recovery of hPM-1 was also found to remain high even in HCl elution.
  • 1.4. Correlation Between NaCl Concentrations in the HCl Eluate and Various Factors
  • Table 2 summarizes the analysis of the correlation between NaCl concentrations in the HCl eluate and various factors.
  • TABLE 2
    NaCl concentration 0 mM 50 mM 100 mM
    Turbidity 0.004 0.007 0.011
    (pH unadjusted)
    Turbidity 0.252 0.049 0.020
    (pH adjusted)
    % Recovery of 81 86 88
    hPM-1 (filtered) (%)
    Amount of DNA 98 220 241
    (unfiltered)
    (pg DNA/mg hPM-1)
    Amount of DNA 11 30 250
    (filtered) (pg DNA/
    mg hPM-1)
  • For the filtered samples, the % recovery of hPM-1 was 88% at 100 mM NaCl, 86% at 50 mM NaCl and 81% at 0 mM NaCl. The amount of residual DNA was low at 0 mM NaCl in both filtered and unfiltered samples. In particular, the filtered sample supplemented with 0 mM NaCl had a very low DNA content of 11 pg DNA/mg hPM-1.
  • The pH-adjusted samples with a higher turbidity tend to provide a lower % recovery of hPM-1 and a smaller amount of residual DNA after filtration. This result suggests a high possibility that hPM-1 and DNA both contribute to particle formation. It is estimated that hPM-1 and DNA probably interact with each other to form particles by adjusting the pH to 7.0. In view of achieving a higher % recovery of hPM-1, it is preferable to increase the NaCl concentration in the HCl eluate. In view of decreasing an amount of residual DNA, on the other hand, it is desirable to eliminate NaCl supplementation into the HCl eluate.
  • Example 2: Purification of Humanized Anti-PTHrP Antibody
  • A sample containing a humanized anti-PTHrP antibody (a culture medium from CHO cell culture, filtered through 0.45 and 0.2 μm CA SARTOBRAN P filters (sartorius)) was purified by Protein A affinity column chromatography under the conditions indicated below. The anti-PTHrP antibody was prepared as described in International Publication No. WO98/13388 (isoelectric point: pH 8.3).
  • 2.1. Experimental Conditions
  • Purification apparatus: AKTA Explorer (Amersham Pharmacia Biotech)
  • Column: HRS/5, C10, XK-26 (Amersham Pharmacia Biotech)
  • Resin: rProtein A Sepharose Fast Flow
    Load: direct load of the culture medium (pH 6.6 to pH 7.5)
    Adjustment of elution fraction: elution fractions are adjusted to various pH levels with a 1 N aqueous Tris solution and then filtered through a 0.2 μm Cellulose Acetate (hereinafter abbreviated as CA) to remove DNA (the conditions are examined in (1) below).
  • The Protein A column was sufficiently equilibrated with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) and then loaded with the above antibody-containing CM. Subsequently, the column was washed with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) to remove unbound impurities, further washed with citrate-phosphate buffer (pH 7.5) to decrease the conductivity, and then eluted with 20 mM aqueous citric acid. The elution profile was monitored at A280 nm to isolate a protein peak. This Protein A elution fraction was used for the following examination of conditions.
  • 2.2. Examination of Removal Conditions for Residual DNA in the Eluate
  • To ensure efficient removal of residual DNA, the optimal pH for filtration through a filter was investigated. The Protein A elution fraction was adjusted with a 1.0 M aqueous Tris solution to the following pH levels: 2.7 (unadjusted), 4.0, 4.5, 5,0, 5.5, 6.0, 6.5, 7.0 and 7.5. Subsequently, each sample was allowed to stand for a given period of time, filtered through a 0.22 μm CA filter, and then adjusted to pH 7 with a 1.0 M aqueous Tris solution, followed by DNA assay. Table 3 lists the examined pH levels and standing periods, along with the amount of residual DNA.
  • TABLE 3
    Removal of residual DNA (unit: pg/mL)
    Direct
    pH 7.5 pH 7.0 pH 6.5 pH 6.0 pH 5.5 pH 5.0 pH 4.5 pH 4.0 (pH 2.7)
    0 hr. 984 83.3 53.8 <22.5 <15.0 17.2 54.1 32,052 40,878
    6 hr. 816 51.9 <15.0 <22.5 <15.0 <15.0 44.0 38,172 42,078
    24 hr.  310 46.6 <15.0 <22.5 <15.0 <15.0 39.7 42,528 30,222
    (DNA in the culture medium: 6,637,200 pg/mL; DNA in the unfiltered sample: 25,110 pg/mL)
  • As shown in the table, the amount of residual DNA was below the detection limit at pH 5.5 and pH 6.0 in all cases where the samples were allowed to stand for 0, 6 and 24 hours. Also, the removal of residual DNA reached a peak around pH 5.5 and pH 6.0, whereas decreased efficiency of DNA, removal was observed at higher and lower pH levels.
  • Example 3: Purification of Humanized Anti-HM1.24 Antigen Monoclonal Antibody
  • A sample containing a humanized anti-HM1.24 antigen monoclonal antibody (a culture medium from CHO cell culture) was purified by Protein A affinity column chromatography under the conditions indicated in Table 4 below. The anti-HM1.24 antigen monoclonal antibody was prepared as described in International Publication No. WO98/14580 (isoelectric point: pH 9.0).
  • 3.1. Experimental Conditions
  • Column: rProtein A FF, 5 mL (16 mm ID×25 mm H)
    Flow rate: 5 mL/min (150 cm/h)
    Sample: direct load of the culture medium
  • TABLE 4
    Equilibration 10 mM C-P Buffer, 1M NaCl, pH 7.5
    (20 CV)
    Load Direct load of CM
    Washing 1 10 mM C-P Buffer, 1M NaCl, pH 7.5
    (20 CV)
    Washing 2 10 mM C-P Buffer, pH 7.5
    (20 CV)
    Elution Citric acid, pH 2.5
    (10 CV)
    Washing 3 0.1M NaOH
    (4 CV)
  • The Protein A column was sufficiently equilibrated with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) and then loaded with the above antibody-containing CM, Subsequently, the column was washed with 150 mM NaCl-containing citrate-phosphate buffer (pH 7.5) to remove unbound impurities, further washed with citrate-phosphate buffer (pH 7.5) to decrease the conductivity, and then eluted with 20 mM aqueous citric acid. The elution profile was monitored at A280 nm to isolate a protein peak. This Protein A elation fraction was used for the following investigation of conditions.
  • 3.2. Investigation of Removal Conditions for Residual DNA in the Eluate
  • To ensure efficient removal of residual DNA, the optimal pH for filtration through a filter was investigated. The Protein A elution fraction was adjusted with a 1.0 H aqueous Tris solution to the following pH levels (pH=4.5-7.5). Subsequently, each sample was allowed to stand for a given period of time, filtered through a 0.22 μm CA filter, and then adjusted to pH 7 with a 1.0 M aqueous Tris solution, followed by DNA assay and reversed-phase HPLC for assay of the humanized anti-HM1.24 antigen monoclonal antibody. Table 5 shows the results of DNA assay, while Table 6 shows the yield of the humanized anti-HM1.24 antigen monoclonal antibody.
  • TABLE 5
    Removal of residual DNA (unit: pg/ml)
    Experiment 1
    pH 7.5 pH 6.5 pH 5.5
    0 h 1142 624 113
    6 h 3288 1157 117
    (DNA in the culture medium: 235200 pg/ml)
    Experiment 2
    pH 5.5 pH 5.0 pH 4.5
    0 h 137 67 86
    6 h 94 34 164
    (DNA in the culture medium: 5448000 pg/ml;
    DNA in the unfiltered sample: 4330 pg/ml)
  • TABLE 6
    % Recovery of humanized anti-HM1.24
    antigen monoclonal antibody by filtration
    pH 5.5 pH 5.0 pH 4.5
    0 h 98.1% 89.6% 87.8%
    6 h 89.3% 91.1% 98.6%
  • Although the samples purified by Protein A affinity chromatography were still rich in DNA, Experiment 1 indicated that the amount of DNA decreased with decrease in pH in the order of pH 7.5, pH 6.5 and pH 5.5, and that there was a tendency to remove more DNA at 0 hours than at 6 hours. In Experiment 2, the same experiment was carried out under conditions of pH=4.5, 5.0 and 5.5, indicating that DNA was sufficiently removed to the same extent, regardless of pH and standing period within the tested range. In addition, the calculation of % recovery indicated little loss of the humanized anti-HM1.24 antigen monoclonal antibody.
  • Example 4: Purification of Granulocyte Colony-Stimulating Factor (G-CSF)
  • A G-CSF-containing sample (from CHO cell culture; Chugai Pharmaceutical Co., Ltd.) was used for the following examination of conditions (isoelectric point: pH 5.5-5.7).
  • 4.1. Investigation of Removal Conditions for Residual DNA in the Eluate
  • To ensure efficient removal of residual DNA, the optimal pH for filtration through a filter was investigated. The G-CSF-containing sample was diluted in an acidic solution of low conductivity (2.5 mM aqueous HCl) and further formed into an acidic aqueous solution of low conductivity using 20% hydrochloric acid, followed by addition of sample DNA. The G-CSF-containing sample thus treated was adjusted with a 1.0 M aqueous This solution to the following pH level (pH=4.3 or 6.6) and then filtered through a 0.22 μm CA filter. Subsequently, DNA assay was performed on both filtered and unfiltered fractions. Table 7 shows the results of DNA assay.
  • TABLE 7
    Removal of residual DNA (unit: pg/ml)
    pH for filtration pH 6.6 pH 4.3
    Unfiltered 4.3 × 105 4.3 × 105
    Filtered 2.8 × 104 <90
  • This investigation confirms efficient reduction of DNA in the G-CSF-containing sample rich in DNA when the sample was filtered at pH 4.3; namely, the amount of residual DNA was below the assay limit of detection.
  • Example 5: Effects of Virus Removal on the Purification of hPM-1 (Humanized Anti-IL-6 Receptor Antibody) 5.1 Test Material (Antibody-Containing Sample)
  • Samples containing the culture medium (CM) of CHO cells producing hPM-1 antibody (humanized IL-6 receptor antibody), which had been centrifuged to remove the cells and stored at −80° C., were supplemented with X-MuLV, Reo3 and MVM, respectively, followed by filtration through a 0.45 μm filter (Bottle Top Filter, CORNING) for use as test samples for purification investigation. The hPN-1 antibody was prepared as described in Example 1. The viruses used for examination were each obtained from ATCC (American Type Culture Collection).
  • 5.2 Purification by rProtein Column Chromatography
  • The virus-supplemented samples prepared in 5.1 were purified by rProtein Column Chromatography. Detailed conditions are as shown below.
  • Resin: rProteinA Sepharose Fast Flow
  • Instrument: AKTA explorer100, AKTApurifier
  • Column: XK16/20, XK16/40
  • Resin height: 11.5 cm
  • Elution conditions
      • Equilibration: 1 mol/L NaCl, 20 mmol/L C-P Buffer,
        • pH 7.5±0.2, Conductivity 8.5±0.5 S/m
      • Washing 1: 1 mol/L NaCl, 20 mmol/L C-P Buffer,
        • pH 7.5±0.2, Conductivity 8.5±0.5 S/m
      • Washing 2: 10 mmol/L C-P Buffer,
        • pH 7.7 ±0.2, Conductivity 165±20 mS/m
      • Elution: 2.5 mmol/L HCl,
        • pH 2.7±0.2, Conductivity 107±10 mS/m
    5.3 Low pH Treatment
  • The elution fractions obtained in 5.2 were adjusted to pH 3.2±0.1 with 1 mol/L hydrochloric acid and held at a room temperature of 15±5° C. for 30 minutes or longer. Subsequently, each elution fraction was adjusted to pH 7.2±0.1 with a 300 mmol/L Tris solution, 40.0 mL of which was then filtered under a pressure of 0.03±0.01 MPa using a filtration unit equipped with a glass fiber filter (Millipore) (0.2 μm, PALL) connected to the primary side and a BioInert (0.2 μm, PALL) (a PALL filter holder equipped with a ϕ15 mm adjuster) connected to the secondary side.
  • 5.4 Detection of Viruses
  • All the samples collected were measured by TCID50 assay. In the clearance capacity test for X-MuLV and MVM, these viruses were detected not only by TCID assay which was measured by viral infectivity in detection cells, but also by RT/Q-PCR and Q-PCR which allowed determination of the virus amount in fractions.
  • 5.5 Results
  • The results of detection in 5.4 are shown in the tables below.
  • TABLE 8
    Virus titer (TCID50 assay: Log10/mL)
    Reo3 MVM
    Run 1 Run 2 Run 1 Run 2
    Unfiltered 5.76 5.76 4.80 4.18
    Filtered ≤1.03 ≤1.03 ≤1.03 ≤1.03
  • TABLE 9
    Virus titer (PCR: Log10 Copies/5 μL)
    X-MuLV MVM
    Run 1 Run 2 Run 1 Run 2
    Unfiltered 5.05 4.77 4.18 2.83
    Filtered ≤1.90 ≤1.90 ≤1.30 ≤1.90
  • As shown above, the purification process of the present invention achieves very high LRVs (Logarithmic Reduction Values) for all the tested viruses and this examination confirms that the viruses were removed to a level below the assay limit of detection after low pH treatment and filtration.
  • INDUSTRIAL APPLICABILITY
  • The method of the present invention enables efficient removal of impurities such as DNA contaminants and viruses in a very simple manner, and is significantly advantageous in purifying physiologically active proteins, especially antibodies. The method achieves an extremely low DNA concentration (e.g., 22.5 pg/ml) when impurities are DNA molecules, while it achieves an extremely low virus titer (e.g., 1.03 (expressed in Log10/mL), as measured by TCID50 assay) when impurities are viruses. The method of the present invention also enables cost reduction and has great significance in this field.

Claims (22)

1. A method for removing impurities in a physiologically active protein-containing sample, which comprises the steps of:
1) forming the physiologically active protein-containing sample into an aqueous solution of low conductivity as well as a pH equal to or lower than the isoelectric point of the physiologically active protein; and
2) removing the resulting particles.
2. The method according to claim 1, wherein the aqueous solution of low conductivity has a conductivity of 0 to 100 mM, as expressed in molarity.
3. The method according to claim 1, wherein the aqueous solution of low conductivity has an ionic strength of 0 to 0.2.
4. The method according to claim 1, wherein the aqueous solution of low conductivity has a conductivity of 0 to 300 mS/m.
5. The method according to claim 1, wherein the aqueous solution is selected from aqueous solutions of hydrochloric acid, citric acid and acetic acid.
6. The method according to claim 1 wherein the pH of the aqueous solution is equal to or lower than the isoelectric point of the physiologically active protein and equal to or higher than pH 2.0.
7. The method according to claim 1, wherein the impurities are DNA contaminants
8. The method according to claim 1 wherein the impurities are viruses.
9. The method according to claim 7, wherein the physiologically active protein-containing sample has the DNA contaminants at a DNA concentration of 22.5 pg/ml or less after the treatment of removal of DNA contaminants
10. The method according to claim 1, wherein the physiologically active protein is an antibody.
11. The method according to claim 10, wherein the antibody is an IgG antibody.
12. The method according to claim 10, wherein the antibody is a humanized monoclonal antibody.
13. The method according to claim 12, wherein the antibody is a humanized anti-IL-6 receptor antibody.
14. The method according to claim 12, wherein the antibody is a humanized anti-HM1.24 antigen monoclonal antibody.
15. The method according to claim 12, wherein the antibody is a humanized anti-parathyroid hormone-related peptide antibody (anti-PTHrP antibody).
16. The method according to claim 1, wherein the physiologically active protein is granulocyte colony-stimulating factor.
17. The method according to claim 1, wherein the particles are removed by filtration through a filter.
18. The method according to claim 1, wherein step 1) is accomplished by forming the physiologically active protein-containing sample into an acidic or alkaline aqueous solution of low conductivity, and adjusting the resulting sample with a buffer to a pH equal to or lower than the isoelectric point of the physiologically active protein.
19. The method according to claim 1,
wherein the physiologically active protein is an antibody, and
wherein step 1) is accomplished by subjecting the antibody-containing sample to affinity chromatography on Protein A or G, eluting the sample with an acidic aqueous solution of low conductivity, and adjusting the resulting eluate with a buffer to a pH equal to or lower than the isoelectric point of the antibody.
20. The method according to claim 18, wherein the buffer is an aqueous solution of Tris.
21. A purified physiologically active protein obtainable by the method according to claim 1.
22. A method for manufacturing a medical protein formulation, which comprises a purification step in which the method according to claim 1 is used.
US17/562,536 2002-09-11 2021-12-27 Protein purification method Pending US20220185843A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/562,536 US20220185843A1 (en) 2002-09-11 2021-12-27 Protein purification method

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
JP2002265609 2002-09-11
JP265609/2002 2002-09-11
PCT/JP2003/011642 WO2004024752A1 (en) 2002-09-11 2003-09-11 Method of purifying protein
US10/527,455 US8420789B2 (en) 2002-09-11 2003-09-11 Method for removing DNA contaminants from a protein-containing sample
US13/847,691 US8809509B2 (en) 2002-09-11 2013-03-20 Protein purification method
US14/329,487 US10654888B2 (en) 2002-09-11 2014-07-11 Method for removing viruses in a physiologically-active protein-containing sample
US16/844,063 US20200231624A1 (en) 2002-09-11 2020-04-09 Protein purification method
US17/562,536 US20220185843A1 (en) 2002-09-11 2021-12-27 Protein purification method

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/844,063 Continuation US20200231624A1 (en) 2002-09-11 2020-04-09 Protein purification method

Publications (1)

Publication Number Publication Date
US20220185843A1 true US20220185843A1 (en) 2022-06-16

Family

ID=31986593

Family Applications (5)

Application Number Title Priority Date Filing Date
US10/527,455 Expired - Lifetime US8420789B2 (en) 2002-09-11 2003-09-11 Method for removing DNA contaminants from a protein-containing sample
US13/847,691 Expired - Lifetime US8809509B2 (en) 2002-09-11 2013-03-20 Protein purification method
US14/329,487 Expired - Lifetime US10654888B2 (en) 2002-09-11 2014-07-11 Method for removing viruses in a physiologically-active protein-containing sample
US16/844,063 Abandoned US20200231624A1 (en) 2002-09-11 2020-04-09 Protein purification method
US17/562,536 Pending US20220185843A1 (en) 2002-09-11 2021-12-27 Protein purification method

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US10/527,455 Expired - Lifetime US8420789B2 (en) 2002-09-11 2003-09-11 Method for removing DNA contaminants from a protein-containing sample
US13/847,691 Expired - Lifetime US8809509B2 (en) 2002-09-11 2013-03-20 Protein purification method
US14/329,487 Expired - Lifetime US10654888B2 (en) 2002-09-11 2014-07-11 Method for removing viruses in a physiologically-active protein-containing sample
US16/844,063 Abandoned US20200231624A1 (en) 2002-09-11 2020-04-09 Protein purification method

Country Status (13)

Country Link
US (5) US8420789B2 (en)
EP (3) EP2261230B1 (en)
JP (2) JP5525118B2 (en)
KR (1) KR101119448B1 (en)
CN (1) CN100522989C (en)
AU (1) AU2003262087B2 (en)
CA (1) CA2497364C (en)
DK (2) DK2261230T3 (en)
ES (2) ES2558303T3 (en)
HK (1) HK1077836A1 (en)
HU (1) HUE027134T2 (en)
SI (2) SI1561756T1 (en)
WO (1) WO2004024752A1 (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE547119T1 (en) 1997-03-21 2012-03-15 Chugai Pharmaceutical Co Ltd PREVENTIVE OR THERAPEUTIC AGENT CONTAINING AN IL-6 ANTAGONIST AS AN ACTIVE INGREDIENTS FOR SENSITIZED T CELL-MEDIATED DISEASES
UA80091C2 (en) 2001-04-02 2007-08-27 Chugai Pharmaceutical Co Ltd Remedies for infant chronic arthritis-relating diseases and still's disease which contain an interleukin-6 (il-6) antagonist
AU2003211990A1 (en) 2002-02-14 2003-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
EP2261230B1 (en) * 2002-09-11 2017-05-10 Chugai Seiyaku Kabushiki Kaisha Protein purification method
GB2401040A (en) 2003-04-28 2004-11-03 Chugai Pharmaceutical Co Ltd Method for treating interleukin-6 related diseases
EP3269738A1 (en) 2004-03-24 2018-01-17 Chugai Seiyaku Kabushiki Kaisha Subtypes of humanized antibody against interleukin-6 receptor
US8088900B2 (en) 2006-03-30 2012-01-03 Novo Nordisk A/S Two-phase precipitation of proteins
WO2008025748A1 (en) 2006-08-28 2008-03-06 Ares Trading S.A. Process for the purification of fc-containing proteins
AU2007291282B2 (en) * 2006-08-28 2011-10-20 Ares Trading S.A. Process for the purification of Fc-fusion proteins
WO2008036688A2 (en) * 2006-09-18 2008-03-27 Xencor, Inc. Optimized antibodies that target hm1.24
US20080071067A1 (en) * 2006-09-20 2008-03-20 Avigenics, Inc. Methods of purifying proteins from egg white
CN101679507A (en) * 2007-03-29 2010-03-24 艾博特公司 crystalline anti-human il-12 antibodies
PL3327026T3 (en) 2007-07-09 2020-02-28 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
AU2016213727A1 (en) * 2007-07-09 2016-08-25 Genentech, Inc. Prevention of disulfide bond reduction during recombinant production of polypeptides
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
KR20190045414A (en) * 2007-11-30 2019-05-02 애브비 바이오테크놀로지 리미티드 Protein formulations and methods of making same
PE20091174A1 (en) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd LIQUID FORMULATION WITH HIGH CONCENTRATION OF ANTIBODY CONTENT
MX2011001506A (en) * 2008-08-14 2011-03-15 Genentech Inc Methods for removing a contaminant using indigenous protein displacement ion exchange membrane chromatography.
GB0818228D0 (en) * 2008-10-06 2008-11-12 Avecia Biolog Ltd Purification process
EP2412817B2 (en) * 2009-03-27 2019-06-05 Asahi Kasei Medical Co., Ltd. Method for removing viruses in high-concentration monoclonal antibody solution
ES2817802T3 (en) * 2009-08-07 2021-04-08 Emd Millipore Corp Methods to purify a target protein from one or more impurities in a sample
BR122022001178B1 (en) 2009-10-26 2022-10-04 F. Hoffmann-La Roche Ag METHOD FOR THE PRODUCTION OF A GLYCOSYLED IMMUNOGLOBULIN AND ITS USE
DK2513134T3 (en) 2009-12-18 2017-12-18 Novartis Ag Washing solution and process for affinity chromatography
MX348739B (en) 2010-03-10 2017-06-27 F Hoffmann-La Roche Ag * Method for purifying immunoglobulin solutions.
WO2012064627A2 (en) 2010-11-08 2012-05-18 Genentech, Inc. Subcutaneously administered anti-il-6 receptor antibody
EP2682168A1 (en) 2012-07-02 2014-01-08 Millipore Corporation Purification of biological molecules
WO2014133741A1 (en) * 2013-02-26 2014-09-04 Emd Millipore Corporation Selective removal of a protein from a mixture of proteins using activated carbon by adjusting solution conditions
DK3016976T3 (en) * 2013-07-01 2020-12-07 Csl Behring Ag Process
MX363403B (en) 2013-07-04 2019-03-22 Hoffmann La Roche Interference-suppressed immunoassay to detect anti-drug antibodies in serum samples.
AU2014329403B2 (en) * 2013-10-03 2020-10-08 Takeda Vaccines, Inc. Methods of detection and removal of rhabdoviruses from cell lines
CN105407905B (en) 2014-01-03 2020-03-03 生物辐射实验室股份有限公司 Removal of impurities from protein A eluate
JP6521351B2 (en) 2014-10-14 2019-05-29 テックプロジェクトサービス株式会社 Virus inactivation and sampling device
JPWO2016121701A1 (en) 2015-01-26 2017-11-09 株式会社カネカ Affinity separation matrix for protein purification containing immunoglobulin κ chain variable region
EP3252158A4 (en) 2015-01-26 2018-07-18 Kaneka Corporation Mutant immunoglobulin kappa chain variable region-binding peptide
SI3337817T1 (en) * 2015-08-21 2021-12-31 F. Hoffmann - La Roche Ag Method for the reduction of host cell proteins in affinity chromatography
CA2992420A1 (en) 2015-08-21 2017-03-02 F. Hoffmann-La Roche Ag Method for the reduction of host cell proteins in affinity chromatography
WO2017032610A1 (en) * 2015-08-21 2017-03-02 F. Hoffmann-La Roche Ag Affinity chromatography purification with low conductivity wash buffer
CN106496302B (en) * 2015-09-08 2021-12-10 三生国健药业(上海)股份有限公司 Method for purifying protein by ion exchange chromatography
RU2018124771A (en) * 2015-12-09 2020-01-13 Басф Се METHOD FOR PROTEIN CLEANING FROM FERMENTATION SOLIDS UNDER DESORBING CONDITIONS
EP3373739B1 (en) * 2016-02-19 2019-03-06 Coöperatie Avebe U.A. Coagulated protein for human food
US11484591B2 (en) 2016-02-22 2022-11-01 Ohio State Innovation Foundation Chemoprevention using controlled-release formulations of anti-interleukin 6 agents, synthetic vitamin A analogues or metabolites, and estradiol metabolites
WO2017195638A1 (en) 2016-05-09 2017-11-16 株式会社カネカ Method for refining antibody or antibody fragment containing κ-chain variable region
CN105837667A (en) * 2016-05-18 2016-08-10 北京天坛生物制品股份有限公司 Method for removing residual hose cell DNA in recombinant Hansenula polymorpha hepatitis B surface antigen
CN110603057A (en) 2017-03-17 2019-12-20 俄亥俄州创新基金会 Nanoparticles for delivery of chemopreventive agents
AU2018392334A1 (en) * 2017-12-22 2020-05-28 Regeneron Pharmaceuticals, Inc. System and method for characterizing drug product impurities
US11603527B2 (en) * 2017-12-27 2023-03-14 Global Life Sciences Solutions Usa Llc Method and kit for viral vector isolation
CN107937425A (en) * 2017-12-27 2018-04-20 北京四环生物制药有限公司 The production method of long-acting recombinant human erythropoietin
CN110272491B (en) * 2018-03-13 2023-01-24 江苏恒瑞医药股份有限公司 Purification process of anti-PD-1 antibody
WO2019183334A1 (en) 2018-03-21 2019-09-26 Waters Technologies Corporation Non-antibody high-affinity-based sample preparation, sorbents, devices and methods
EP3947737A2 (en) 2019-04-02 2022-02-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of predicting and preventing cancer in patients having premalignant lesions
WO2022155142A1 (en) * 2021-01-12 2022-07-21 Repligen Corporation Offline and inline determination of concentration of metabolites in cell culture fluid

Family Cites Families (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4124576A (en) 1976-12-03 1978-11-07 Coval M L Method of producing intravenously injectable gamma globulin
JPS58201994A (en) 1982-05-21 1983-11-25 Hideaki Hagiwara Method for producing antigen-specific human immunoglobulin
JP2648301B2 (en) 1983-12-27 1997-08-27 ジエネテイツクス・インスチチユ−ト・インコ−ポレ−テツド Vector containing auxiliary DNA for transformation of eukaryotic cells
FI852545L (en) 1984-06-29 1985-12-30 Ortho Diagnostic Systems Inc SANDWICHANALYS AV ANTIKROPPSLEKTIN.
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DE3640513A1 (en) 1986-11-27 1988-06-09 Biotest Pharma Gmbh METHOD FOR THE PRODUCTION OF A VIRUS-SAFE, STORAGE-STABLE AND INTRAVENOES COMPATIBLE IMMUNGLOBULIN-G-PRAEPARATES
DE3641115A1 (en) 1986-12-02 1988-06-16 Lentia Gmbh METHOD FOR PRODUCING AN INTRAVENOUS APPLICABLE AND STABLE IN LIQUID FORM IMMUNOGLOBULINS
EP0287513B1 (en) * 1987-04-11 1992-12-02 Ciba-Geigy Ag An isoelectric focusing process and a means for carrying out said process
KR930008447B1 (en) * 1987-10-23 1993-09-04 쉐링 코포레이션 Method of purifying protein
ES2063783T3 (en) * 1988-06-03 1995-01-16 Chugai Pharmaceutical Co Ltd HUMAN CRYSTALLINE FACTOR OF STIMULATION OF GRANULOCYTE COLONIES AND PROCEDURE FOR ITS PREPARATION.
US5166322A (en) 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
AU646822B2 (en) 1989-10-13 1994-03-10 Kirin-Amgen Inc. Erythropoietin isoforms
US5177194A (en) 1990-02-01 1993-01-05 Baxter International, Inc. Process for purifying immune serum globulins
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
EP0585287B1 (en) 1990-07-10 1999-10-13 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
EP0814159B1 (en) 1990-08-29 2005-07-27 GenPharm International, Inc. Transgenic mice capable of producing heterologous antibodies
GB9022547D0 (en) 1990-10-17 1990-11-28 Wellcome Found Purified immunoglobulin
US5110910A (en) 1991-03-25 1992-05-05 Miles Inc. Virucidal euglobulin precipitation
RU2139351C1 (en) 1991-04-25 1999-10-10 Чугаи Сейяку Кабусики Кайся H- and l-chains of monoclonal antibody pm-1 (monat) to human il-6r receptor and their v-region, modified monat, its h- and l-chains and their v-regions, cdr-sequence, dna-sequence
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
PT1024191E (en) 1991-12-02 2008-12-22 Medical Res Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
EP0656941B1 (en) 1992-03-24 2005-06-01 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
JPH07509137A (en) 1992-07-24 1995-10-12 セル ジェネシス,インク. Production of xenoantibodies
DE4302163A1 (en) 1993-01-27 1994-07-28 Thomae Gmbh Dr K Process for the purification of proteins and peptides
US5328989A (en) 1993-03-05 1994-07-12 Schering-Plough Purification of human interleukin-10 from a cell culture medium
US5840297A (en) 1993-03-19 1998-11-24 Johns Hopkins University Vaccine comprising anti-idiotypic antibody to chlamydia GLXA and process
EP0754225A4 (en) 1993-04-26 2001-01-31 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
GB9313509D0 (en) 1993-06-30 1993-08-11 Medical Res Council Chemisynthetic libraries
IL192290A0 (en) 1993-08-17 2008-12-29 Kirin Amgen Inc Erythropoietin analogs
AU690171B2 (en) 1993-12-03 1998-04-23 Medical Research Council Recombinant binding proteins and peptides
US5429746A (en) * 1994-02-22 1995-07-04 Smith Kline Beecham Corporation Antibody purification
US8017121B2 (en) 1994-06-30 2011-09-13 Chugai Seiyaku Kabushika Kaisha Chronic rheumatoid arthritis therapy containing IL-6 antagonist as effective component
JPH089902A (en) 1994-07-04 1996-01-16 Shiseido Co Ltd Low allergy processed food
US5994524A (en) 1994-07-13 1999-11-30 Chugai Seiyaku Kabushiki Kaisha Polynucleotides which encode reshaped IL-8-specific antibodies and methods to produce the same
JP3630453B2 (en) 1994-09-30 2005-03-16 中外製薬株式会社 A therapeutic agent for immature myeloma cells comprising an IL-6 receptor antibody as an active ingredient
ATE390933T1 (en) 1995-04-27 2008-04-15 Amgen Fremont Inc HUMAN ANTIBODIES AGAINST IL-8 DERIVED FROM IMMUNIZED XENOMICES
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
FI952196A0 (en) * 1995-05-08 1995-05-08 Suomen Punainen Risti Veripalv Immunoglobulin production
EP0840624B1 (en) * 1995-07-14 2007-07-25 CAF - DCF cvba - scrl Apparatus for inactivating viral contaminants in blood products
AU3276297A (en) 1996-06-27 1998-01-14 Chugai Seiyaku Kabushiki Kaisha Remedies for myeloma to be used together with nitrogen mustard antitumor agents
TW491855B (en) 1996-08-07 2002-06-21 Csl Ltd Purification of immunoglobulins
CN100335501C (en) 1996-09-26 2007-09-05 中外制药株式会社 Antibody against human parathormone related peptides
UA76934C2 (en) * 1996-10-04 2006-10-16 Chugai Pharmaceutical Co Ltd Reconstructed human anti-hm 1.24 antibody, coding dna, vector, host cell, method for production of reconstructed human antibody, pharmaceutical composition and drug for treating myeloma containing reconstructed human anti-hm 1.24 antibody
PT950067E (en) 1996-11-27 2007-12-06 Genentech Inc Affinity purification of polypeptide on protein a matrix
US5886154A (en) * 1997-06-20 1999-03-23 Lebing; Wytold R. Chromatographic method for high yield purification and viral inactivation of antibodies
US6096872A (en) * 1997-10-14 2000-08-01 Ortho Diagnostic Systems, Inc. Viral clearance process
US5972613A (en) * 1997-12-09 1999-10-26 The Perkin-Elmer Corporation Methods of nucleic acid isolation
UA64742C2 (en) 1997-12-24 2004-03-15 Альфа Терапевтик Корпорейшн Process for producing intravenously-administrable gamma globulin solution and product manufactured by this process
PL343322A1 (en) 1998-04-03 2001-08-13 Chugai Pharmaceutical Co Ltd Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
AU753468B2 (en) 1998-06-09 2002-10-17 Csl Behring Ag Process for producing immunoglobulins for intravenous administration and other immunoglobulin products
JP2003516111A (en) 1999-02-22 2003-05-13 バリアジェニックス インコーポレーテッド Genetic sequence variants with utility in determining treatment for disease
JP2000319294A (en) 1999-05-11 2000-11-21 Asahi Chem Ind Co Ltd Purification of solution of polymer originating from organism
JP2000351799A (en) 1999-06-08 2000-12-19 Jcr Pharmaceuticals Co Ltd Production of fibronectin solution
US6355243B1 (en) 1999-11-13 2002-03-12 Bayer Corporation Method of thrombolysis by local delivery of active plasmin
EP2311492B1 (en) 2000-08-11 2017-10-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilized preparations
ES2184594B1 (en) 2001-01-17 2004-01-01 Probitas Pharma Sa PROCEDURE FOR THE PRODUCTION OF GAMMAGLOBULINA G HUMANA INACTIVADA OF VIRUS.
JP2002265909A (en) 2001-03-07 2002-09-18 Nippon Synthetic Chem Ind Co Ltd:The Pressure sensitive adhesive dispersed in nonaqueous medium
ES2869895T3 (en) 2001-03-09 2021-10-26 Chugai Pharmaceutical Co Ltd Protein purification method
JP4187941B2 (en) 2001-03-14 2008-11-26 リンテック株式会社 Boron-containing polyorganosilsesquioxane and adhesive composition
AU784808B2 (en) 2001-04-02 2006-06-29 Kedrion Melville Inc. Prion and viral clearance process
KR20040023630A (en) 2001-06-26 2004-03-18 아브게닉스, 인크. Antibodies to opgl
EP1463942B2 (en) * 2001-12-21 2012-06-20 Immunex Corporation Methods for purifying protein
AU2003211990A1 (en) 2002-02-14 2003-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing solution pharmaceuticals
EP2261230B1 (en) 2002-09-11 2017-05-10 Chugai Seiyaku Kabushiki Kaisha Protein purification method
AU2005282241B2 (en) 2004-09-08 2011-03-03 Chelsea Therapeutics, Inc. Quinazoline derivatives as metabolically inert antifolate compounds.
KR101140483B1 (en) 2007-05-02 2012-07-11 에프. 호프만-라 로슈 아게 Method for stabilizing a protein
JP5504579B2 (en) 2008-05-12 2014-05-28 積水ハウス株式会社 Residential floor detachable structure and home refurbishment method
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
NZ621655A (en) 2009-10-26 2015-08-28 Nestec Sa Assays for the detection of anti-tnf drugs and autoantibodies
US8623377B2 (en) 2011-06-29 2014-01-07 University Of Maryland, Baltimore Joint-homing peptides and uses thereof
CN105392916B (en) 2013-03-11 2019-03-08 Sio2医药产品公司 Coat packaging materials
US9387246B2 (en) 2013-09-03 2016-07-12 L. Douglas Graham Treatment methods for rheumatoid arthritis

Also Published As

Publication number Publication date
CN100522989C (en) 2009-08-05
EP2261230B1 (en) 2017-05-10
EP1561756B1 (en) 2015-12-23
SI1561756T1 (en) 2016-02-29
HK1077836A1 (en) 2006-02-24
SI2261230T1 (en) 2017-08-31
CA2497364C (en) 2013-03-19
JP2011006489A (en) 2011-01-13
US20140323695A1 (en) 2014-10-30
HUE027134T2 (en) 2016-10-28
JPWO2004024752A1 (en) 2006-01-05
AU2003262087A1 (en) 2004-04-30
KR20050038042A (en) 2005-04-25
EP2261230A1 (en) 2010-12-15
CA2497364A1 (en) 2004-03-25
AU2003262087B2 (en) 2010-11-11
ES2626268T3 (en) 2017-07-24
KR101119448B1 (en) 2012-03-15
DK2261230T3 (en) 2017-06-12
EP1561756A4 (en) 2006-08-23
EP3225625A1 (en) 2017-10-04
US20060142549A1 (en) 2006-06-29
US8420789B2 (en) 2013-04-16
US20130225796A1 (en) 2013-08-29
US20200231624A1 (en) 2020-07-23
US8809509B2 (en) 2014-08-19
CN1681837A (en) 2005-10-12
WO2004024752A1 (en) 2004-03-25
JP5525118B2 (en) 2014-06-18
ES2558303T3 (en) 2016-02-03
EP1561756A1 (en) 2005-08-10
DK1561756T3 (en) 2016-02-15
US10654888B2 (en) 2020-05-19
JP5687469B2 (en) 2015-03-18

Similar Documents

Publication Publication Date Title
US20220185843A1 (en) Protein purification method
US7927815B2 (en) Protein purification method
JP6783767B2 (en) How to Purify Recombinant Protein
US20120282654A1 (en) Antibody purification
US20240158437A1 (en) Process of purification of protein
WO2021072210A1 (en) Methods of purifying ranibizumab or a ranibizumab variant

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TAKEDA, KOZO;OCHI, NORIMICHI;ISHII, KIMIE;AND OTHERS;REEL/FRAME:058483/0333

Effective date: 20050921

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION