US20140005367A1 - High concentration antibody-containing liquid formulation - Google Patents

High concentration antibody-containing liquid formulation Download PDF

Info

Publication number
US20140005367A1
US20140005367A1 US14/017,013 US201314017013A US2014005367A1 US 20140005367 A1 US20140005367 A1 US 20140005367A1 US 201314017013 A US201314017013 A US 201314017013A US 2014005367 A1 US2014005367 A1 US 2014005367A1
Authority
US
United States
Prior art keywords
antibody
formulation
arginine
methionine
antibodies
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/017,013
Inventor
Toshiyuki Morichika
Daisuke Kameoka
Yoshimi Imaeda
Terutoshi Maeda
Oliver Boris Stauch
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
Hoffmann La Roche Inc
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=40824372&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140005367(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 US14/017,013 priority Critical patent/US20140005367A1/en
Publication of US20140005367A1 publication Critical patent/US20140005367A1/en
Priority to US14/963,414 priority patent/US20160090419A1/en
Priority to US16/390,197 priority patent/US11008394B2/en
Priority to US17/242,199 priority patent/US11359026B2/en
Assigned to HOFFMANN-LA ROCHE INC. reassignment HOFFMANN-LA ROCHE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: F. HOFFMANN-LA ROCHE AG
Priority to US17/752,978 priority patent/US11584798B2/en
Priority to US18/162,368 priority patent/US11767363B2/en
Priority to US18/335,789 priority patent/US20230340134A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to an antibody-containing formulation, and particularly, to a stable liquid formulation containing a high concentration of an antibody.
  • an antibody-containing formulation for subcutaneous injection, since a dose of an antibody per administration is large (about 100 mg to 200 mg) and an amount of an injection solution is generally limited in subcutaneous injection, it is necessary to increase a concentration of an antibody in a liquid to be administered.
  • high concentration formulations are used, which are prepared by the lyophilization-concentration technique, in which a lyophilized formulation is reconstituted in water having a volume smaller than that before lyophilization.
  • Solutions containing a high concentration of an antibody tend to form solutions having a high viscosity due to macromolecular properties of proteins, and due to the intermolecular interactions of proteins. Further, in cases where a protein is stored in a form of a solution having a high concentration, problematic degradation occurs, which includes a generation of insoluble and/or soluble aggregates; and it is necessary to prevent such degradation. Especially, in antibody formulations, associations are likely to be formed and insoluble aggregates are likely to be generated in a liquid state. In cases where a liquid formulation is stored for a long time, a problem exists in that a bioactivity of antibody molecules is lost due to deamidation of amino acid residues such as aspargine residues.
  • An object of the present invention is to provide a high concentration antibody-containing liquid formulation, in which dimerization and deamidation during long-term storage are inhibited, and which stable and suitable for use in subcutaneous administration.
  • the present inventors conducted intensive study with a view to attaining the above object, and as a result, discovered that a stable high concentration antibody-containing liquid formulation can be provided by adding an amino acid, arginine or a salt thereof, as a stabilizer, to thereby complete the present invention.
  • the present invention provides the following:
  • a stable antibody-containing liquid formulation characterized’ y comprising arginine and methionine.
  • a stable liquid formulation containing an anti-IL-6 receptor antibody characterized by comprising either arginine or methionine.
  • a method for inhibiting dimerization of molecules of an antibody in a liquid formulation containing the antibody, comprising adding a and methionine to the liquid formulation.
  • a liquid formulation containing a high concentration of an antibody is provided, with which reformulation by concentration by lyophilization is not necessary, and hence does not require reconstitution.
  • the antibody-containing liquid formulation according to the present invention can he stored in a liquid state for a long time. Since the antibody-containing liquid formulation according to the present invention can be produced by a process not including a lyophilization step, addition of a sugar or the like as a cryoprotective agent is not necessary.
  • FIG. 1 shows a typical chromatogram of Example 1
  • FIG. 2 shows evaluation results of the gel permeation chromatography (SEC) in Example 1.
  • FIG. 3 shows evaluation results of the gel permeation chromatography (SEC) in Example 1,
  • FIG. 4 shows a typical chromatogram of Example 2.
  • FIG. 5 shows evaluation results of the ion exchange chromatography (IEC) in Example 2.
  • Example 6 shows evaluation results of the ion exchange chromatography (IEC) in Example 2.
  • FIG. 7 shows evaluation results of the gel permeation chromatography (SEC) in Example 3.
  • FIG. 8 shows evaluation results of the ion exchange chromatography (IEC) in Example 3.
  • antibody-containing liquid formulation means a liquid formulation containing an antibody as an active component, which is prepared such that it can be administered to an animal such as human, and which is preferably produced by a process not including a lyophilization step.
  • the antibody-containing liquid formulation according to the present invention is a liquid pharmaceutical formulation containing an antibody at a high concentration, which preferably has an antibody concentration of not less than 50 mg/mL, more preferably not less than 100 mg/mL, still more preferably not less than 120 mg/mL, and yet more preferably not less than 150 mg/mL.
  • a liquid formulation containing antibody at a concentration of 120 mg/mL or higher, or preferably 150 mg/mL or higher has not been developed for commercial use. Namely, the present invention allows for the first time to put to use a liquid formulation containing antibody at this high concentration.
  • the highest concentration of antibody in the liquid formulation according to the present invention may be typically 300 mg/mL, preferably 250 mg/mL and more preferably 200 mg/mL. Therefore, the antibody-containing liquid formulation according to the present invention preferably has an antibody concentration of from 50 to 300 mg/mL, more preferably from 100 to 300 mg/mL, still more preferably from 120 to 250 mg/mL, and yet more preferably from 150 to 200 mg/mL.
  • the antibody to be used in the present invention is not restricted as long as it binds to a desired antigen.
  • the antibody can be either a polyclonal antibody or a monoclonal antibody, although a monoclonal antibody is preferred because an antibody having uniform properties can be produced stably.
  • a monoclonal antibody which can be used in the present invention includes not only monoclonal antibodies originated from an animal such as human, mouse, rat, hamster, rabbit, sheep, camel or monkey, but also includes artificially modified recombinant antibodies such as chimeric antibody, humanized antibody and bispecific antibody.
  • the immunoglobulin class of the antibody is not restricted, and can be any of the classes including IgGs such as IgG1, IgG2, IgG3 and IgG4, IgA, IgD, IgE and IgM. Among these classes, IgG and IgM are preferred.
  • the antibody which can be used in the present invention includes not only whole antibodies, but also antibody fragments such as Fv, Fab and F(ab) 2 ; and low molecular weight antibodies such as single chain Fv (scFv, sc(Fv), diabodies such as scPv duller) having one or more specificities, prepared by binding the variable regions of an antibody through a linker such as a peptide linker.
  • a linker such as a peptide linker
  • a hybridoma producing a monoclonal antibody can be prepared as follows basically utilizing a known technique. That is, the hybridoma can be prepared by immunizing an animal with a desired antigen or cells expressing the desired antigen as a sensitizing antigen by a standard method; fusing the obtained immunocytes with known parent cells by a standard cell-fusion method; and screening a monoclonal antibody-producing cell (hybridoma) by a standard screening method. Preparation of a hybridoma can be carried out by, for example, the method according to the method by Milstein et al (Kohlen G. and Milstein, C., Methods Enzymol. (1981) 73: 3-46). In cases where the immunogenicity of the antigen is low, the antigen can be bound to an antigenic macromolecule such as albumin, and the resulting conjugate can be used as an immunogen.
  • Recombinant antibodies can be employed, which are prepared by the genetic recombination technique in which an antigen gene is cloned from a hybridoma, incorporating the gene into an appropriate vector, introducing the vector into a host, and making the host produce the, antibody (see, for example, Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). More specifically, a cDNA encoding the variable region (V region) in the antibody is synthesized from the mRNA of a hybridoma using a reverse transcriptase.
  • a DNA encoding the V region -f the desired antibody is obtained, the DNA is then ligated to a DNA encoding the constant region (C region) of a desired antibody, and the resulting ligated DNA is introduced into an expression vector.
  • a DNA encoding the V region of the antibody can be incorporated into an expression vector containing the DNA encoding the C region of the antibody.
  • the DNA is incorporated into the expression vector such that the DNA is expressed under the control of an expression-controlling region such as enhancer or promoter.
  • Host cells are then transformed with the resulting expression vector, and the antibody can be expressed by the host cells.
  • recombinant antibodies artificially modified for the purpose of reducing the heteroantigenicity to human such as chimeric antibodies and humanized antibodies
  • modified antibodies can be produced by known methods.
  • chimeric antibody is an antibody comprising variable regions in the heavy chain and light chain in an antibody of an animal other than human, such as mouse, and constant regions in the heavy chain and light chain in an antibody of human, and can be obtained by ligating a DNA encoding the variable region in the mouse antibody with a DNA encoding the constant region in the human antibody, incorporating the obtained DNA into an expression vector, introducing the expression vector into a host, and making the host produce the antibody.
  • Humanized antibody is also called reshaped human antibody, and is obtained by transplanting the CDR (complementarity determining region) of, for example, a mouse antibody to the complementarity determining region of a human antibody.
  • a standard genetic recombination technique for preparing the humanized antibody is also known. Specifically, a DNA designed such that the CDR of the mouse antibody and the framework region (FR) of the human antibody are ligated is synthesized by PCR method from several oligonucleotides prepared so as to have overlapping regions at their terminals. The obtained DNA is ligated to a DNA encoding the constant region of a human antibody, and the resulting DNA is introduced into an expression vector.
  • the expression vector is introduced into a host, and the host is made to produce the humanized antibody (see EP 239400 A and WO 96/02576).
  • the FR of the human antibody to be ligated through CDR one of which complementarity determining region forms a good antigen-binding site is selected.
  • an amino acid(s) in the complementarity determining region can be substituted so that the complementarity determining region of the reshaped human antibody forms an appropriate antigen-binding site (Sato, K. el al., Cancer Res. (1993) 53, 851-856).
  • a desired human antibody having a binding activity to a desired antigen can be obtained by sensitizing, in vitro, human lymphocytes with the desired antigen or with the cells expressing the desired antigen; fusing the sensitized lymphocytes with human myeloma cells, for example, U266 cells; and obtaining the antibody from the cells (see JP 1-59878 B).
  • the desired human antibody can also be obtained by imminuzing a transgenic animal having all repertories of human antibody genes with the antigen (see WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096 and WO 96/33735). Further, a technique by which a human antibody is obtained by panning using a human antibody library is also known. For example, a variable region of a human body is expressed in the form of a single chain antibody (scFv) on the surface of a phage by use of a phage display method, and the phage which binds to the antigen can be selected.
  • scFv single chain antibody
  • the DNA sequence coding for the variable region of the human antibody which binds to the antigen can be determined. If the DNA sequence of the scFv which binds to the antigen is determined, an appropriate expression vector containing the sequence is constructed, and the humanized antibody can be obtained.
  • WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236,WO 93/19172, WO 95/01438 and WO 95/15388 can be referred to.
  • an antibody gene is once isolated, and the gene is introduced into an appropriate host so as to prepare the antibody
  • appropriate combinations of the host and expression vector can be used.
  • animal cells in cases where eukaryotic cells are used as the host, animal cells, plant cells and fungal cells can be used.
  • eukaryotic cells include (1) mammalian cells, for example, CHO, COS, myeloma, BHK (baby hamster kidney), Hela and Vero, (2) amphibian cells, for example, Xenopus oocytes and (3) insect cells, for example, sf9, sf21 and Tn5.
  • Known plant cells include cells originated from plants belonging to the genus Nicotiana , for example, Nicotiana tabacum , and the cells can be subjected to callus culture.
  • Known fungal cells include the cells originated from yeasts, for example, those belonging to the genus Saccharomyces such as Saccharomyces cerevisiae ; and filamentous bacteria, for example, those belonging to the genus Aspergillus such as Aspergillus niger . In cases where prokaryotic cells are used, there are production systems using bacterial cells.
  • Known bacterial cells include E. coli cells and Bacillus subtilis cells. The antibody is obtained by introducing a desired antibody gene into these cells by transformation, and culturing the transformed cells in vitro.
  • Antibodies in the form of antibody fragments, low molecular weight antibodies and modified antibodies can also be employed as the antibody in the present invention.
  • the antibody fragments and low molecular weight antibodies include Fab. F(ab′) 2 , Fv, and single chain Fv (scFv, sc(Fv) 2 and the like) having one or more specificities, prepared by ligating the Fvs in the H-chain and IL-chain through an appropriate linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A. (1988) 85, 5879-5883).
  • an antibody is treated with papain or pepsin to generate antibody fragments, or a gene encoding these antibody fragments is constructed, and the gene is expressed in appropriate host cells after introducing the gene into an expression vector (see, for example, Co, M. S. et al., T. Immunol. (1994)152, 2968-2976; Better, M. and Horwitz A. H., Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E., Methods (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol, (1986)121, 663-669 ; Bird, R. E. and Walker, B. W., Trends Biotechnol. (1991) 9, 132-137.
  • Antibodies bound to various molecules such as polyethylene glycol (PEG) can also be used as modified antibodies.
  • the term “antibody” used in the present invention also includes these modified antibodies. These modified antibodies can be obtained by chemically modifying an obtained antibody. Methods for carrying out the modifications are established in the art.
  • Examples of the antibody contained in the formulation according to the present invention include, but not limited to, anti-tissue factor antibodies, anti-IL-6 receptor antibodies, anti-IL-6 antibodies, HM1.24 antigen monoclonal antibodies, anti-parathyroid hormone-related peptide antibodies (anti-PTHP antibodies), anti-glypican-3 antibodies, anti-ganglioside GM3 antibodies, anti-TPO receptor antagonist antibodies, factor VIII-substituting antibodies, anti-CD3 antibodies, anti-CD20 antibodies, anti-GPIIb/IIIa antibodies, anti-TNF antibodies, anti-CD25 antibodies, anti-EGFR antibodies, anti-Her2/neu antibodies, anti-RSV antibodies, anti-CD33 antibodies, and-CD52 antibodies, anti-IgE antibodies, anti-CD11a antibodies, anti-VEGE antibodies, anti-VLA4 antibodies, anti-AXL antibodies, and so on.
  • IL-6 receptor antibodies include humanized anti-interleukin (IL-6) receptor antibodies (hPM-1 or MRA) (see WO 92-19759), humanized anti-HM1.24 antigen monoclonal antibodies (see WO 98-14580), humanized anti-parathyroid hormone-related peptide antibodies (anti-PTHrP antibodies) (see WO 98-13388), humanized anti-tissue factor antibodies (see WO 99-51743) and anti-glypican.-3 humanized IgG IK antibodies (see PCT/JP05/013103),
  • the humanized antibodies especially preferred in the present invention are humanized anti-IL-6 receptor antibodies,
  • human IgM antibodies anti-ganglioside GM3 recombinant human IgM antibodies (see WO 05-05636) and the like are preferred.
  • anti-TPO receptor antagonist diabodies see WO 02-33072
  • anti-CD47 agonist diabodies see WO 01-66737
  • the present inventors studied the effects of various additives by conducting heat acceleration tests and light acceleration tests. As a result, it was found that in solutions in which a high concentration of antibody was dissolved a buffer solution containing the amino acid arginine, the amount of generated dimer was smaller than that in solutions to which arginine was not added. From these results, it was found that arginine is effective as a stabilizer for inhibiting dimerization.
  • a stable antibody formulation can be provided, in which dimerization of the antibody is reduced and deamidation of the antibody is prevented. Therefore, a first aspect of the present invention is characterized by adding arginine to a solution, whereby dimerization or deamidation of the antibody molecules is inhibited in the resulting antibody-containing liquid formulation. Accordingly, an embodiment as a stable antibody-containing liquid formulation is characterized in that it contains an antibody and arginine in a buffer solution. Further, as described above, an antibody-containing liquid formulation of the present invention can additionally contain methionine in the solution, with a synergistic effect being obtained by use of arginine and methionine in combination.
  • a second aspect of the present invention is characterized by adding arginine and methionine to a solution, whereby dimerization, in particular, of the antibody molecules is inhibited in the resulting antibody-containing liquid formulation.
  • an embodiment as a stable antibody-containing liquid formulation is characterized in that it contains an antibody, arginine and methionine in a buffer solution,
  • any of the arginine compound per se, derivatives thereof and salts thereof can be used. L-arginine and salts thereof are preferred.
  • the methionine used in the present invention any of the methionine compound per se, derivatives thereof and salts thereof can be used. L-methionine and salts thereof are preferred.
  • the concentration of arginine is preferably 50 to 1500 mM, more preferably 100 to 1000 mM, still more preferably 200 to 700 mM.
  • the total concentration of arginine and methionine is preferably 50 to 1200 mM, for example, preferably, the arginine concentration is 40 to 1000 mM and the methionine concentration is 10 to 200 mM; more preferably, the arginine concentration is 50 to 700 mM and the methionine concentration is 10 to 100 mM; and still more preferably, the arginine concentration is 100 to 300 mM, and the methionine concentration is 10 to 50 mM.
  • the buffer solution is prepared using a buffering agent which is a substance for maintaining a pH of the solution.
  • a pH of the formulation is preferably 4 to 8, more preferably 5.0 to 7.5, still more preferably 5.5 to 7,2, and still more preferably 6.0 to 6.5.
  • a buffering agent which can be used in the present invention is one which can adjust the pH in this range and which is pharmaceutically acceptable.
  • Such a buffering agent is known by those skilled in the art, and examples thereof include inorganic salts such as phosphoric acid salts (sodium or potassium) and sodium hydrogen carbonate organic acid salts such as citric acid salts (sodium or potassium), sodium acetate and sodium succinate; and acids such as phosphoric acid, carbonic acid, citric acid, succinic acid, malic acid and gluconic acid.
  • inorganic salts such as phosphoric acid salts (sodium or potassium) and sodium hydrogen carbonate organic acid salts such as citric acid salts (sodium or potassium), sodium acetate and sodium succinate
  • acids such as phosphoric acid, carbonic acid, citric acid, succinic acid, malic acid and gluconic acid.
  • Tris buffers Good's buffers such as MES, MOPS and HEPES, histidine (e.g., histidine hydrochloric acid salt) and glycine can also be used.
  • the buffer is preferably a histidine buffer or glycine buffer, and a histidine buffer is especially preferred.
  • concentration of the buffer solution is generally 1 to 500 mM, preferably 5 to 100 mM, still more preferably 10 to 20 mM.
  • the buffer solution contains histidine at a concentration of preferably 5 to 25 mM, more preferably 10 to 20 mM.
  • the “stable” high concentration antibody-containing liquid formulation according to the present invention significant change is not observed when it is stored at a refrigeration temperature (2 to 8° C.) for at least 12 months, preferably for 2 years, and more preferably for 3 years; or when it is stored at room temperature (22 to 28° C.) for at least 3 months, preferably 6 months, and more preferably 1 year.
  • sum amount of dimers and degradation products in the formulation when it is stored at 5° C. for 2 years is 5.0% or lower, preferably 2% or lower, and more preferably 1.5% or lower; or sum amount of dimers and degradation products in the formulation when it is stored at 25° C. for 6 months is 5.0% or lower, preferably 2% or lower, and more preferably 1.5% or longer.
  • the formulation according to the present invention can further contain a surfactant.
  • Typical examples of the surfactant include nonionic surfactants, for example, sorbitan fatty acid esters such as sorbitan monocaprylate, sorbitan monolaurate and sorbitan monopalmitate; glycerin fatty acid esters such as glycerol monocaprylate, glycerol monomyristate and glycerol monostearate, polyglycerol fatty acid esters such as decaglyceryl monostearate, decaglyceryl distearate and decaglyceryl monolinoleate; polyoxyethylene sorbitan fatty acid esters such as polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate, polyoxyethylenesorbitan monostearate, polyoxyethylene sorbitan monopalmitate, polyoxyethylene sorbitan trioleate and polyoxyethylene sorbitan tristearate; polyoxyethylene sorbitol fatty acid esters such as polyoxyethylene sorbitol t
  • Preferred surfactants are polyoxyethylene sorbitan fatty acid esters and polyoxyethylene polyoxypropylene alkyl ethers, and especially preferred are polysorbates 20, 21, 40, 60, 65, 80, 81 and 85, and Pluronic type surfactants, and most preferred are polysorbates 20 and 80, and Phironic F-68 (Poloxamer 188),
  • the amount of the surfactant( )to be added to the antibody formulation according to the present invention is generally 0.0001 to 10% (w/v), preferably 0.001 to 5%, more preferably 0.005 to 3%.
  • the formulation according to the present invention is preferably substantially composed of the following components:
  • substantially composed of herein means that a component other than the components usually added to formulations is not contained, the components usually added to formulations being the optional additive components described below, such as suspending agents, solubilizing agents, isotonic agents, preservatives, adsorption inhibitors, diluents, vehicles, pH-adjusters, soothing agents, sulfur--containing reducing agents and antioxidants.
  • arginine and/or methionine, and additional other amino acid(s) as an optional additional component(s)
  • additional other amino acid(s) e.g., tryptophan
  • the formulation contains (b-i) arginine; (b-2) arginine and methionine; and (b-3) methionine; respectively, as an amino acid additive(s), and further include the cases where the formulation additionally contains other amino acid(s).
  • Preferred example of the other amino acid(s) is tryptophan.
  • any of the tryptophan compound per se, derivatives thereof and salts thereof can be used. L-tryptophan and salts thereof are preferred.
  • a suspending agent such as sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium bicarbonate, sodium bicarbonate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulfate, sodium sulf
  • suspending agent examples include methyl cellulose, polysorbate 80, hydroxyethyl cellulose, gum arabic, powdered tragacanth, sodiumcarboxymethylcellulose and polyoxyethylene sorbitan monolaurate.
  • solubilizing agent examples include, polyoxyethylene hydrogenated castor oil, polysorbate 80, nicotinamide, polyoxyethylene sorbitan. monolaurate, macrogol and castor oil fatty acid ethyl ester.
  • Examples of the isotonic agent include sodium chloride, potassium chloride and calcium chloride.
  • preservative examples include methyl p-hydroxybenzoate, ethyl p-hydroxybenzoate, sorbic acid, phenol, cresol and chlorocresol.
  • adsorption inhibitor examples include human serum albumin, lecithin, dextran, ethylencoxide--propylene oxide copolynner, hydroxypropylcellulose, methyl cellulose, polyoxyethylene hydrogenated castor oil and polyethylene glycol.
  • sulfur-containing reducing agent examples include the compounds having a sulfhydryl group(s), such as N-acetylcysteine, N-acetyl homocysteine, thioctic acid, thiodiglycol, thioethanolamine, thioglycerol, thiosorbitol, thioglycolic acid and salts thereof, sodium thiosulfate, glutathione and C 1 -C 7 thioalkanes.
  • a sulfhydryl group(s) such as N-acetylcysteine, N-acetyl homocysteine, thioctic acid, thiodiglycol, thioethanolamine, thioglycerol, thiosorbitol, thioglycolic acid and salts thereof, sodium thiosulfate, glutathione and C 1 -C 7 thioalkanes.
  • antioxidants examples include erythorbic acid, dibutylhydroxytoluene, butylated hydroxyanisole, ⁇ -tocopherol, tocopherol acetate, L-ascorbic acid and salts thereof, L-ascorbyl palmitate, L-ascorbyl stearate, sodium hydrogen sulfite, sodium sulfite, triamyl gallate, propyl gallate, and chelating agents such as disodium ethyleriediaminetetraacetate (EDTA) sodium pyrophosphate and sodium metaphosphate.
  • EDTA disodium ethyleriediaminetetraacetate
  • the antibody-containing liquid formulation according to the present invention usually administered through a parenteral route, for example, by injection (subcutaneous, intravenous, intramuscular injections or the like), percutaneous, transmucosal, transnasal or pulmonary administration, but it can also be administered orally.
  • a parenteral route for example, by injection (subcutaneous, intravenous, intramuscular injections or the like), percutaneous, transmucosal, transnasal or pulmonary administration, but it can also be administered orally.
  • subcutaneous injection the dose of antibody per administration large (about 100 to 200 mg) while the amount of the injection solution is limited, so that the formulation according to the present invention is especially suited for subcutaneous injection.
  • the osmotic pressure ratio of the antibody-containing liquid formulation according to the present invention is preferably about 0.5 to 4, more preferably about 0.7 to 2, and still more preferably about 1.
  • the viscosity of the antibody-containing liquid formulation according to the present invention is preferably about 2 to 15 mPa ⁇ s, more preferably about 4 to 10 mPa ⁇ s. It should be noted that the viscosity described herein is measured by a rotation viscometer method using a cone-plate type viscometer, in accordance with 2.53 Viscosity Determination / General Tests , the Japanese Pharmacopoeia, 15th edition.
  • a stable liquid formulation can be obtained, in which dimerization and deamidation of the antibody during long-term storaize are small, by adding to the formulation arginine alone, or arginine and methionine, or methionine alone.
  • a method for inhibiting deamidation in antibody-containing liquid formulations comprising adding to the formulation arginine or a salt thereof.
  • a method for inhibiting dimerization of antibody in antibody-containing liquid formulations comprising adding to the formulation arginine and methionine.
  • the antibody is preferably anti--IL-6 receptor antibody, which is a humanized antibody or human antibody.
  • the humanized anti-IL-6 receptor antibody was the humanized antibody prepared in accordance with the method described in Reference Example 2 in JP 8-99902 A using the human elongation factor La promoter described in Example 10 in WO 92/19759. This antibody will occasionally be referred to as “MRA” in the tables in Examples.
  • Liquid formulations containing anti-IL-6 receptor humanized antibody were evaluated for an influence on stabilization of the formulations obtained by use of a combination of arginine and methionine.
  • evaluation samples numbered A1 to A9 were prepared. Prescriptions for the evaluation samples were as follows:
  • each sample was subjected to a heat acceleration test (stored at 40° C. for 3 months and at 25° C. for 6 months, respectively).
  • the purity of the antibody before and after the heat acceleration test was evaluated by gel permeation chromatography (SEC).
  • SEC gel permeation chromatography
  • the sample was used as the solution to he measured as it was.
  • LMW Low Molecular Weight Degradation Products
  • FIG 1 A typical chromatography is shown in FIG 1 .
  • the evaluation results obtained by the gel permeation chromatography (SEC) are shown in Table 1 and FIGS. 2 and 3 .
  • the amount of dimer the samples (Sample Nos. A2 to A6) to which arginine was added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was smaller than that in the sample (Sample No. A1) to which arginine was not added and accordingly, the inhibitory effect of arginine against dimerization was confirmed. It was also confirmed that the amount of dirtier was reduced proportionally to the amount of the arginine added. On the other hand, the amount of dimer in the samples (Sample Nos.
  • Liquid formulations containing and-IL-6 receptor humanized antibody were evaluated for influence on the deamidation by arginine.
  • evaluation samples numbered A10 to A15 and numbered A16 to A18, containing different amounts of arginine and methionine, respectively, were prepared.
  • each sample was subjected to a heat acceleration test (stored at 40° C. for 3 months and at 25° C. for 6 months, respectively).
  • the purities of the antibody before and after the heat acceleration test were evaluated by ion-exchange chromatography (IEC).
  • the analytical conditions were as follows:
  • MRA Pre indicates the total of the peaks of the substances each eluted after a retention time shorter than that of the main component, and a plurality of degradation products, mainly deamidation products of human ed anti-IL-6 receptor antibody, was included. When the production amount of this Pre peak was small, inhibition of deamidation of the antibody is indicated.
  • Total Area of All Peaks Grand Total of Total Area of MRA Pre Peaks+Peak Area of MRA Main+Peak Area of MAR Sub-1+Peak Area of MAR Sub-2+Peak Area of MAR Sub-3+Peak Area of MAR R-1+Total Area of 1Q(H)-MRA Peaks+Total Area of 2Q(H)-MRA Peaks+Peak Area of Others
  • Amount of MRA Pre (%) (Total Area of MRA Pre Peaks/Total Area of All Peaks) ⁇ 100
  • MRA Pre indicates the total of the peaks of the substances appealing earlier than that of the main component.
  • Evaluation results of the ion-exchange chromatography are shown Table and FIGS. 5 and 6 .
  • the amount of Pre peaks in the samples (Sample Nos. A11 to A15) to which arginine was added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was smaller than that in the sample (Sample No. A10) to which arginine was not added; and accordingly, the inhibitory effect of arginine against the generation of Pre peaks was confirmed. It was also confirmed that the amount of Pre peaks was reduced proportionally to an amount of arginine added. On the other hand, the amount of Pre peaks in the samples (Sample Nos.
  • liquid formulations containing receptor humanized antibody were evaluated for influence on stabilization of the formulations obtained by use of a combination of arginine and methionine.
  • each sample was subjected to a light acceleration test (total illuminance 1,200,000 lux and total near-ultraviolet radiation energy: 200 W ⁇ h/m 2 ).
  • the purities of the antibody before and after the light acceleration test were evaluated by gel permeation chromatography (SEC) and ion exchange chromatography (IEC) as in Examples 1 and 2.
  • the evaluation results by the gel permeation chromatography (SEC) are shown in Table 3 and FIG. 7 .
  • the amount of dimer in the samples (Sample Nos. A20 to A24) to which arginine was added, after the light acceleration test was smaller than that in the sample (Sample No. A19) to which arginine was not added; and accordingly, the inhibitory effect of arginine against dimerization was confirmed. It was also confirmed that the amount of dimer was reduced proportionally to an amount of arginine added.
  • the amount of dimer n the samples (Sample Nos.
  • the amount of Pre peak in the samples (Sample Nos. A20 to A24) to which arginine was added, after the light acceleration test was smaller than that in the sample (Sample No. A19) to which arginine was not added; and accordingly, the inhibitory effect of arginine against formation of Pre peak was confirmed. Further, it was confirmed that as the amount of arginine increases, the production amount of Pre peak decreases proportionately.
  • the amount of dimer after the light acceleration test in the samples (Sample Nos. A25 to A27) to which methionine was further added to arginine (100 mM) was smaller than that in the sample (Sample No.

Abstract

The problem to be solved is to provide an antibody-containing formulation which is stable and suited for subcutaneous administration, wherein dimerization and deamidation is prevented during long-term storage.
The present application is directed to a stable antibody-containing liquid formulation characterized by containing arginine and methionine.

Description

    TECHNICAL FIELD
  • The present invention relates to an antibody-containing formulation, and particularly, to a stable liquid formulation containing a high concentration of an antibody.
  • BACKGROUND ART
  • In recent years, various antibody formulations have been developed and used in practice. Many such antibody formulations are used in intravenous injection. However, due to needs of a clinical site, there is an increasing demand for development of an antibody-containing formulation that can be administered as a self-injectable subcutaneous injection.
  • In designing an antibody-containing formulation for subcutaneous injection, since a dose of an antibody per administration is large (about 100 mg to 200 mg) and an amount of an injection solution is generally limited in subcutaneous injection, it is necessary to increase a concentration of an antibody in a liquid to be administered. In view of this, in many cases, high concentration formulations are used, which are prepared by the lyophilization-concentration technique, in which a lyophilized formulation is reconstituted in water having a volume smaller than that before lyophilization. However, a strong demand exists for a liquid formulation which does not require reconstitution, and which is easy to handle. Although an increase in a viscosity of a formulation due to addition of a cryoprotective agent such as a sugar in the production process of the lyophilized formulation is not preferred for formulations for subcutaneous injection, it is surmised that this problem could be avoided if the formulation were a liquid formulation.
  • Solutions containing a high concentration of an antibody tend to form solutions having a high viscosity due to macromolecular properties of proteins, and due to the intermolecular interactions of proteins. Further, in cases where a protein is stored in a form of a solution having a high concentration, problematic degradation occurs, which includes a generation of insoluble and/or soluble aggregates; and it is necessary to prevent such degradation. Especially, in antibody formulations, associations are likely to be formed and insoluble aggregates are likely to be generated in a liquid state. In cases where a liquid formulation is stored for a long time, a problem exists in that a bioactivity of antibody molecules is lost due to deamidation of amino acid residues such as aspargine residues.
  • There have been proposed various ideas for providing a stabilized formulation, in which loss of an active component is small even after the formulation is stored for a long period of time. Such formulations are produced by dissolving an active component and various additives in a buffer solution. However, for liquid formulations containing a high concentration of an antibody, there does not yet exist a technology that is sufficient to prevent dimerization and deamidation.
  • A need to provide a high concentration antibody-containing formulation exists, in which dimerization and deamidation during long-term storage are inhibited, and which is both stable and suitable for use in subcutaneous administration.
  • DISCLOSURE OF THE INVENTION Problem to be Solved by the Invention
  • An object of the present invention is to provide a high concentration antibody-containing liquid formulation, in which dimerization and deamidation during long-term storage are inhibited, and which stable and suitable for use in subcutaneous administration.
  • Means for Solving the Problem
  • The present inventors conducted intensive study with a view to attaining the above object, and as a result, discovered that a stable high concentration antibody-containing liquid formulation can be provided by adding an amino acid, arginine or a salt thereof, as a stabilizer, to thereby complete the present invention.
  • That is, the present invention provides the following:
  • (1) A stable antibody-containing liquid formulation, characterized’ y comprising arginine and methionine.
  • (2) The formulation of (1) further comprising a histidine buffering agent.
  • (3) The formulation of (1) or (2) further comprising a surfactant.
  • (4) The formulation according to (1) to (3) containing the antibody in an amount of at least 50 mg/ml.
  • (5) The formulation according to (1) to (3) containing the antibody in an amount of a east 100 mg/ml.
  • (6) The formulation according to (1) to (3) containing the antibody in an amount of at least 120 mg/ml.
  • (7) The formulation according to (1) to (6) wherein the antibody is an anti-IL-6 receptor antibody.
  • (8) A stable liquid formulation containing an anti-IL-6 receptor antibody, characterized by comprising either arginine or methionine.
  • (9) The formulation according to (1) to (8) wherein the antibody is a humanized antibody or human antibody.
  • (10) The formulation according to (1) to (9) further comprising tryptophan.
  • (11) The formulation according to (1) to (10) having the pH in the range from 4 to 8.
  • (12) The formulation according to (1) to (11) wherein the arginine is present amount of from 50 to 1500 mM.
  • (13) The formulation according to (1) to (12) having a viscosity of from 2 to 15 mPa·s.
  • (14) The formulation according to (1) to (13), which is stable at 22-28° C. for at least 6 months.
  • (15) The formulation according to (1) to (13), characterized in that dimerization of antibody molecules is inhibited.
  • (16) The formulation according to (1) to (13), characterized in that deamidation of antibody molecules is inhibited.
  • (17) The formulation according to (1) to (13), which is for subcutaneous administration.
  • (18) The formulation according to (1) to (13) which has not been subjected to lyophilization during preparation of the formulation.
  • (19) A method for inhibiting deamidation of molecules of an antibody in a liquid formulation containing the antibody, comprising adding arginine to the liquid formulation.
  • (20) A method for inhibiting dimerization of molecules of an antibody in a liquid formulation containing the antibody, comprising adding a and methionine to the liquid formulation.
  • Advantages of the Invention
  • By the present invention, a liquid formulation containing a high concentration of an antibody is provided, with which reformulation by concentration by lyophilization is not necessary, and hence does not require reconstitution. The antibody-containing liquid formulation according to the present invention can he stored in a liquid state for a long time. Since the antibody-containing liquid formulation according to the present invention can be produced by a process not including a lyophilization step, addition of a sugar or the like as a cryoprotective agent is not necessary.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a typical chromatogram of Example 1,
  • FIG. 2 shows evaluation results of the gel permeation chromatography (SEC) in Example 1.
  • FIG. 3 shows evaluation results of the gel permeation chromatography (SEC) in Example 1,
  • FIG. 4 shows a typical chromatogram of Example 2.
  • FIG. 5 shows evaluation results of the ion exchange chromatography (IEC) in Example 2.
  • 6 shows evaluation results of the ion exchange chromatography (IEC) in Example 2.
  • FIG. 7 shows evaluation results of the gel permeation chromatography (SEC) in Example 3.
  • FIG. 8 shows evaluation results of the ion exchange chromatography (IEC) in Example 3.
  • BEST MODE FOR CARRYING OUT THE INVENTION
  • The present invention will now be described in detail.
  • In the present invention, “antibody-containing liquid formulation” means a liquid formulation containing an antibody as an active component, which is prepared such that it can be administered to an animal such as human, and which is preferably produced by a process not including a lyophilization step.
  • The antibody-containing liquid formulation according to the present invention is a liquid pharmaceutical formulation containing an antibody at a high concentration, which preferably has an antibody concentration of not less than 50 mg/mL, more preferably not less than 100 mg/mL, still more preferably not less than 120 mg/mL, and yet more preferably not less than 150 mg/mL. It should be noted that a liquid formulation containing antibody at a concentration of 120 mg/mL or higher, or preferably 150 mg/mL or higher, has not been developed for commercial use. Namely, the present invention allows for the first time to put to use a liquid formulation containing antibody at this high concentration.
  • Further, considering the manufacturing process, the highest concentration of antibody in the liquid formulation according to the present invention may be typically 300 mg/mL, preferably 250 mg/mL and more preferably 200 mg/mL. Therefore, the antibody-containing liquid formulation according to the present invention preferably has an antibody concentration of from 50 to 300 mg/mL, more preferably from 100 to 300 mg/mL, still more preferably from 120 to 250 mg/mL, and yet more preferably from 150 to 200 mg/mL.
  • The antibody to be used in the present invention is not restricted as long as it binds to a desired antigen. The antibody can be either a polyclonal antibody or a monoclonal antibody, although a monoclonal antibody is preferred because an antibody having uniform properties can be produced stably.
  • A monoclonal antibody which can be used in the present invention includes not only monoclonal antibodies originated from an animal such as human, mouse, rat, hamster, rabbit, sheep, camel or monkey, but also includes artificially modified recombinant antibodies such as chimeric antibody, humanized antibody and bispecific antibody. The immunoglobulin class of the antibody is not restricted, and can be any of the classes including IgGs such as IgG1, IgG2, IgG3 and IgG4, IgA, IgD, IgE and IgM. Among these classes, IgG and IgM are preferred.
  • The antibody which can be used in the present invention includes not only whole antibodies, but also antibody fragments such as Fv, Fab and F(ab)2; and low molecular weight antibodies such as single chain Fv (scFv, sc(Fv), diabodies such as scPv duller) having one or more specificities, prepared by binding the variable regions of an antibody through a linker such as a peptide linker.
  • The above-described antibodies which can be used in the present invention can be prepare(by methods well known to those skilled in the art.
  • A hybridoma producing a monoclonal antibody can be prepared as follows basically utilizing a known technique. That is, the hybridoma can be prepared by immunizing an animal with a desired antigen or cells expressing the desired antigen as a sensitizing antigen by a standard method; fusing the obtained immunocytes with known parent cells by a standard cell-fusion method; and screening a monoclonal antibody-producing cell (hybridoma) by a standard screening method. Preparation of a hybridoma can be carried out by, for example, the method according to the method by Milstein et al (Kohlen G. and Milstein, C., Methods Enzymol. (1981) 73: 3-46). In cases where the immunogenicity of the antigen is low, the antigen can be bound to an antigenic macromolecule such as albumin, and the resulting conjugate can be used as an immunogen.
  • Recombinant antibodies can be employed, which are prepared by the genetic recombination technique in which an antigen gene is cloned from a hybridoma, incorporating the gene into an appropriate vector, introducing the vector into a host, and making the host produce the, antibody (see, for example, Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). More specifically, a cDNA encoding the variable region (V region) in the antibody is synthesized from the mRNA of a hybridoma using a reverse transcriptase. If a DNA encoding the V region -f the desired antibody is obtained, the DNA is then ligated to a DNA encoding the constant region (C region) of a desired antibody, and the resulting ligated DNA is introduced into an expression vector. Alternatively, a DNA encoding the V region of the antibody can be incorporated into an expression vector containing the DNA encoding the C region of the antibody. The DNA is incorporated into the expression vector such that the DNA is expressed under the control of an expression-controlling region such as enhancer or promoter. Host cells are then transformed with the resulting expression vector, and the antibody can be expressed by the host cells.
  • In the present invention, recombinant antibodies artificially modified for the purpose of reducing the heteroantigenicity to human, such as chimeric antibodies and humanized antibodies can be used. These modified antibodies can be produced by known methods. chimeric antibody is an antibody comprising variable regions in the heavy chain and light chain in an antibody of an animal other than human, such as mouse, and constant regions in the heavy chain and light chain in an antibody of human, and can be obtained by ligating a DNA encoding the variable region in the mouse antibody with a DNA encoding the constant region in the human antibody, incorporating the obtained DNA into an expression vector, introducing the expression vector into a host, and making the host produce the antibody.
  • Humanized antibody is also called reshaped human antibody, and is obtained by transplanting the CDR (complementarity determining region) of, for example, a mouse antibody to the complementarity determining region of a human antibody. A standard genetic recombination technique for preparing the humanized antibody is also known. Specifically, a DNA designed such that the CDR of the mouse antibody and the framework region (FR) of the human antibody are ligated is synthesized by PCR method from several oligonucleotides prepared so as to have overlapping regions at their terminals. The obtained DNA is ligated to a DNA encoding the constant region of a human antibody, and the resulting DNA is introduced into an expression vector. The expression vector is introduced into a host, and the host is made to produce the humanized antibody (see EP 239400 A and WO 96/02576). As the FR of the human antibody to be ligated through CDR, one of which complementarity determining region forms a good antigen-binding site is selected. As required, an amino acid(s) in the complementarity determining region can be substituted so that the complementarity determining region of the reshaped human antibody forms an appropriate antigen-binding site (Sato, K. el al., Cancer Res. (1993) 53, 851-856).
  • Methods for obtaining a human antibody are known in the art. For example, a desired human antibody having a binding activity to a desired antigen can be obtained by sensitizing, in vitro, human lymphocytes with the desired antigen or with the cells expressing the desired antigen; fusing the sensitized lymphocytes with human myeloma cells, for example, U266 cells; and obtaining the antibody from the cells (see JP 1-59878 B). The desired human antibody can also be obtained by imminuzing a transgenic animal having all repertories of human antibody genes with the antigen (see WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096 and WO 96/33735). Further, a technique by which a human antibody is obtained by panning using a human antibody library is also known. For example, a variable region of a human body is expressed in the form of a single chain antibody (scFv) on the surface of a phage by use of a phage display method, and the phage which binds to the antigen can be selected. By analyzing the gene of the selected phage, the DNA sequence coding for the variable region of the human antibody which binds to the antigen can be determined. If the DNA sequence of the scFv which binds to the antigen is determined, an appropriate expression vector containing the sequence is constructed, and the humanized antibody can be obtained. These methods are well known, and WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236,WO 93/19172, WO 95/01438 and WO 95/15388 can be referred to.
  • In cases where an antibody gene is once isolated, and the gene is introduced into an appropriate host so as to prepare the antibody, appropriate combinations of the host and expression vector can be used. In cases where eukaryotic cells are used as the host, animal cells, plant cells and fungal cells can be used. Known animal cells include (1) mammalian cells, for example, CHO, COS, myeloma, BHK (baby hamster kidney), Hela and Vero, (2) amphibian cells, for example, Xenopus oocytes and (3) insect cells, for example, sf9, sf21 and Tn5. Known plant cells include cells originated from plants belonging to the genus Nicotiana, for example, Nicotiana tabacum, and the cells can be subjected to callus culture. Known fungal cells include the cells originated from yeasts, for example, those belonging to the genus Saccharomyces such as Saccharomyces cerevisiae; and filamentous bacteria, for example, those belonging to the genus Aspergillus such as Aspergillus niger. In cases where prokaryotic cells are used, there are production systems using bacterial cells. Known bacterial cells include E. coli cells and Bacillus subtilis cells. The antibody is obtained by introducing a desired antibody gene into these cells by transformation, and culturing the transformed cells in vitro.
  • Antibodies in the form of antibody fragments, low molecular weight antibodies and modified antibodies can also be employed as the antibody in the present invention. Examples of the antibody fragments and low molecular weight antibodies include Fab. F(ab′)2, Fv, and single chain Fv (scFv, sc(Fv)2 and the like) having one or more specificities, prepared by ligating the Fvs in the H-chain and IL-chain through an appropriate linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. U.S.A. (1988) 85, 5879-5883). Specifically, an antibody is treated with papain or pepsin to generate antibody fragments, or a gene encoding these antibody fragments is constructed, and the gene is expressed in appropriate host cells after introducing the gene into an expression vector (see, for example, Co, M. S. et al., T. Immunol. (1994)152, 2968-2976; Better, M. and Horwitz A. H., Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra, A., Methods Enzymol. (1989) 178, 497-515; Lamoyi, E., Methods (1986) 121, 652-663; Rousseaux, J. et al., Methods Enzymol, (1986)121, 663-669 ; Bird, R. E. and Walker, B. W., Trends Biotechnol. (1991) 9, 132-137.
  • Antibodies bound to various molecules such as polyethylene glycol (PEG) can also be used as modified antibodies. The term “antibody” used in the present invention also includes these modified antibodies. These modified antibodies can be obtained by chemically modifying an obtained antibody. Methods for carrying out the modifications are established in the art.
  • Examples of the antibody contained in the formulation according to the present invention include, but not limited to, anti-tissue factor antibodies, anti-IL-6 receptor antibodies, anti-IL-6 antibodies, HM1.24 antigen monoclonal antibodies, anti-parathyroid hormone-related peptide antibodies (anti-PTHP antibodies), anti-glypican-3 antibodies, anti-ganglioside GM3 antibodies, anti-TPO receptor antagonist antibodies, factor VIII-substituting antibodies, anti-CD3 antibodies, anti-CD20 antibodies, anti-GPIIb/IIIa antibodies, anti-TNF antibodies, anti-CD25 antibodies, anti-EGFR antibodies, anti-Her2/neu antibodies, anti-RSV antibodies, anti-CD33 antibodies, and-CD52 antibodies, anti-IgE antibodies, anti-CD11a antibodies, anti-VEGE antibodies, anti-VLA4 antibodies, anti-AXL antibodies, and so on.
  • Preferred examples of the reshaped human antibodies used in the present invention include humanized anti-interleukin (IL-6) receptor antibodies (hPM-1 or MRA) (see WO 92-19759), humanized anti-HM1.24 antigen monoclonal antibodies (see WO 98-14580), humanized anti-parathyroid hormone-related peptide antibodies (anti-PTHrP antibodies) (see WO 98-13388), humanized anti-tissue factor antibodies (see WO 99-51743) and anti-glypican.-3 humanized IgG IK antibodies (see PCT/JP05/013103), The humanized antibodies especially preferred in the present invention are humanized anti-IL-6 receptor antibodies,
  • As the human IgM antibodies, anti-ganglioside GM3 recombinant human IgM antibodies (see WO 05-05636) and the like are preferred.
  • As the low molecular weight antibodies, anti-TPO receptor antagonist diabodies (see WO 02-33072), anti-CD47 agonist diabodies (see WO 01-66737) and the like are preferred.
  • To evaluate the shelf stability of the high concentration antibody-containing liquid formulation, the present inventors studied the effects of various additives by conducting heat acceleration tests and light acceleration tests. As a result, it was found that in solutions in which a high concentration of antibody was dissolved a buffer solution containing the amino acid arginine, the amount of generated dimer was smaller than that in solutions to which arginine was not added. From these results, it was found that arginine is effective as a stabilizer for inhibiting dimerization. Further, in solutions in which a high concentration of antibody was dissolved in a buffer solution containing arginine and methionine, the inhibitory effect against dimerization was observed at a total concentration of arginine and methionine which is lower than the concentration of arginine alone needed for attaining the same inhibitory effect. From these results, it was found that a synergistic effect is obtained by the addition of arginine and methionine in combination. Further, it was found that deamidation of the antibody molecules is inhibited by the addition of arginine. These results are exemplified as test results obtained for a sample containing a humanized anti-IL-6 receptor antibody at a concentration of 180 mg/ml.
  • Thus, by adding arginine as a stabilizer, a stable antibody formulation can be provided, in which dimerization of the antibody is reduced and deamidation of the antibody is prevented. Therefore, a first aspect of the present invention is characterized by adding arginine to a solution, whereby dimerization or deamidation of the antibody molecules is inhibited in the resulting antibody-containing liquid formulation. Accordingly, an embodiment as a stable antibody-containing liquid formulation is characterized in that it contains an antibody and arginine in a buffer solution. Further, as described above, an antibody-containing liquid formulation of the present invention can additionally contain methionine in the solution, with a synergistic effect being obtained by use of arginine and methionine in combination. Therefore, a second aspect of the present invention is characterized by adding arginine and methionine to a solution, whereby dimerization, in particular, of the antibody molecules is inhibited in the resulting antibody-containing liquid formulation. Accordingly, an embodiment as a stable antibody-containing liquid formulation is characterized in that it contains an antibody, arginine and methionine in a buffer solution,
  • As the arginine used in the present invention, any of the arginine compound per se, derivatives thereof and salts thereof can be used. L-arginine and salts thereof are preferred. As the methionine used in the present invention, any of the methionine compound per se, derivatives thereof and salts thereof can be used. L-methionine and salts thereof are preferred.
  • In cases :here the antibody-containing liquid formulation according to the present invention contains arginine and does not contain methionine, the concentration of arginine is preferably 50 to 1500 mM, more preferably 100 to 1000 mM, still more preferably 200 to 700 mM. In cases where the antibody-containing liquid formulation according to the present invention contains arginine and methionine, the total concentration of arginine and methionine: is preferably 50 to 1200 mM, for example, preferably, the arginine concentration is 40 to 1000 mM and the methionine concentration is 10 to 200 mM; more preferably, the arginine concentration is 50 to 700 mM and the methionine concentration is 10 to 100 mM; and still more preferably, the arginine concentration is 100 to 300 mM, and the methionine concentration is 10 to 50 mM.
  • The buffer solution is prepared using a buffering agent which is a substance for maintaining a pH of the solution. In a high concentration antibody-containing liquid formulation according to the present invention, a pH of the formulation is preferably 4 to 8, more preferably 5.0 to 7.5, still more preferably 5.5 to 7,2, and still more preferably 6.0 to 6.5. A buffering agent which can be used in the present invention is one which can adjust the pH in this range and which is pharmaceutically acceptable. Such a buffering agent is known by those skilled in the art, and examples thereof include inorganic salts such as phosphoric acid salts (sodium or potassium) and sodium hydrogen carbonate organic acid salts such as citric acid salts (sodium or potassium), sodium acetate and sodium succinate; and acids such as phosphoric acid, carbonic acid, citric acid, succinic acid, malic acid and gluconic acid. Further, Tris buffers, Good's buffers such as MES, MOPS and HEPES, histidine (e.g., histidine hydrochloric acid salt) and glycine can also be used. In the high concentration antibody-containing liquid formulation according to the present invention, the buffer is preferably a histidine buffer or glycine buffer, and a histidine buffer is especially preferred. The concentration of the buffer solution is generally 1 to 500 mM, preferably 5 to 100 mM, still more preferably 10 to 20 mM. In cases where a histidine buffer is used, the buffer solution contains histidine at a concentration of preferably 5 to 25 mM, more preferably 10 to 20 mM.
  • For the “stable” high concentration antibody-containing liquid formulation according to the present invention, significant change is not observed when it is stored at a refrigeration temperature (2 to 8° C.) for at least 12 months, preferably for 2 years, and more preferably for 3 years; or when it is stored at room temperature (22 to 28° C.) for at least 3 months, preferably 6 months, and more preferably 1 year. For example, sum amount of dimers and degradation products in the formulation when it is stored at 5° C. for 2 years is 5.0% or lower, preferably 2% or lower, and more preferably 1.5% or lower; or sum amount of dimers and degradation products in the formulation when it is stored at 25° C. for 6 months is 5.0% or lower, preferably 2% or lower, and more preferably 1.5% or longer.
  • The formulation according to the present invention can further contain a surfactant.
  • Typical examples of the surfactant include nonionic surfactants, for example, sorbitan fatty acid esters such as sorbitan monocaprylate, sorbitan monolaurate and sorbitan monopalmitate; glycerin fatty acid esters such as glycerol monocaprylate, glycerol monomyristate and glycerol monostearate, polyglycerol fatty acid esters such as decaglyceryl monostearate, decaglyceryl distearate and decaglyceryl monolinoleate; polyoxyethylene sorbitan fatty acid esters such as polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate, polyoxyethylenesorbitan monostearate, polyoxyethylene sorbitan monopalmitate, polyoxyethylene sorbitan trioleate and polyoxyethylene sorbitan tristearate; polyoxyethylene sorbitol fatty acid esters such as polyoxyethylene sorbitol tetrastea qte and polyoxyethylene sorbitol tetra oleate; polyoxyethylene glycerin fatty acid esters such as polyoxyethylene glyceryl monostearate; polyethylene glycol fatty acid esters such as polyethylene glycol distearate; polyoxyethylene alkyl ethers such as polyoxyethylene lauryl ether; polyoxyethylene polyoxypropylene alkyl ethers such as polyoxyethylene polyoxypropylene glycol ether, polyoxyethylene polyoxypropylene propyl ether and polyoxyethylene polyoxypropylene cetyl ether; polyoxyethylene alkyl phenyl ethers such as polyoxyethylene nonylphenyl ether; polyoxyethylene hardened castor oils such as polyoxyethylene castor oil and polyoxyethylene hardened castor oil (polyoxyethylene hydrogenated castor oil); polyoxyethylene bees wax derivatives such as polyoxyethylene sorbitol bees wax; polyoxyethylene lanolin derivatives such as polyoxyethylene lanolin; surfactants having an HLB of 6 to 18 such as polyoxyethylene fatty acid amides, for example, polyoxyethylene octadecana ide: anionic surfactants, for example, alkyl sulfate salts having a C10-C18 alkyl group, such as sodium cetyl sulfate, sodium lauryl sulfate and sodium oleyl sulfate; polyoxyethylerie alkyl ether sulfate salts in which the average number of moles of the added ethylene oxide units is 2 to 4 and the number of carbon atoms of the alkyl group is 10 to 18, such as polyoxyethylene sodium lauryl sulfate; alkyl sulfosuccinate salts having a C8-C18 alkyl group, such as sodium lauryl sulfosuccinate; natural surfactants such as lecithin and glycerophospholipos; sphingophospholipids such as sphingomyelin; and sucrose esters of C12-C18 fatty acids. These surfactants can be added to the formulation of the present invention individually, or two or more of these surfactants can be added in combination.
  • Preferred surfactants are polyoxyethylene sorbitan fatty acid esters and polyoxyethylene polyoxypropylene alkyl ethers, and especially preferred are polysorbates 20, 21, 40, 60, 65, 80, 81 and 85, and Pluronic type surfactants, and most preferred are polysorbates 20 and 80, and Phironic F-68 (Poloxamer 188),
  • The amount of the surfactant( )to be added to the antibody formulation according to the present invention is generally 0.0001 to 10% (w/v), preferably 0.001 to 5%, more preferably 0.005 to 3%.
  • In another aspect of the present invention, the formulation according to the present invention is preferably substantially composed of the following components:
    • A) anti-IL-6 receptor antibody;
    • B) arginine and/or methionine, and additional other amino acid(s) (e.g., tryptophan) as an optional additional component(s);
    • C) buffering agent(s); and
    • D) surfactant(s).
  • The term “substantially composed of” herein means that a component other than the components usually added to formulations is not contained, the components usually added to formulations being the optional additive components described below, such as suspending agents, solubilizing agents, isotonic agents, preservatives, adsorption inhibitors, diluents, vehicles, pH-adjusters, soothing agents, sulfur--containing reducing agents and antioxidants.
  • The above-described “B) arginine and/or methionine, and additional other amino acid(s) (e.g., tryptophan) as an optional additional component(s)” is meant to include the cases where the formulation contains (b-i) arginine; (b-2) arginine and methionine; and (b-3) methionine; respectively, as an amino acid additive(s), and further include the cases where the formulation additionally contains other amino acid(s). Preferred example of the other amino acid(s) is tryptophan. As the tryptophan, any of the tryptophan compound per se, derivatives thereof and salts thereof can be used. L-tryptophan and salts thereof are preferred.
  • As required, a suspending agent, solubilizing agent, isotonic agent, preservative, adsorption inhibitor, diluent, vehicle, pH-adjuster, soothing agent, sulfur-containing reducing agent, antioxidant and the like can be added to the formulation according to the present invention.
  • Examples of the suspending agent include methyl cellulose, polysorbate 80, hydroxyethyl cellulose, gum arabic, powdered tragacanth, sodiumcarboxymethylcellulose and polyoxyethylene sorbitan monolaurate.
  • Examples of the solubilizing agent include, polyoxyethylene hydrogenated castor oil, polysorbate 80, nicotinamide, polyoxyethylene sorbitan. monolaurate, macrogol and castor oil fatty acid ethyl ester.
  • Examples of the isotonic agent include sodium chloride, potassium chloride and calcium chloride.
  • Examples of the preservative include methyl p-hydroxybenzoate, ethyl p-hydroxybenzoate, sorbic acid, phenol, cresol and chlorocresol.
  • Examples of the adsorption inhibitor include human serum albumin, lecithin, dextran, ethylencoxide--propylene oxide copolynner, hydroxypropylcellulose, methyl cellulose, polyoxyethylene hydrogenated castor oil and polyethylene glycol.
  • Examples of the sulfur-containing reducing agent include the compounds having a sulfhydryl group(s), such as N-acetylcysteine, N-acetyl homocysteine, thioctic acid, thiodiglycol, thioethanolamine, thioglycerol, thiosorbitol, thioglycolic acid and salts thereof, sodium thiosulfate, glutathione and C1-C7 thioalkanes.
  • Examples of the antioxidant include erythorbic acid, dibutylhydroxytoluene, butylated hydroxyanisole, α-tocopherol, tocopherol acetate, L-ascorbic acid and salts thereof, L-ascorbyl palmitate, L-ascorbyl stearate, sodium hydrogen sulfite, sodium sulfite, triamyl gallate, propyl gallate, and chelating agents such as disodium ethyleriediaminetetraacetate (EDTA) sodium pyrophosphate and sodium metaphosphate.
  • The antibody-containing liquid formulation according to the present invention usually administered through a parenteral route, for example, by injection (subcutaneous, intravenous, intramuscular injections or the like), percutaneous, transmucosal, transnasal or pulmonary administration, but it can also be administered orally. In subcutaneous injection, the dose of antibody per administration large (about 100 to 200 mg) while the amount of the injection solution is limited, so that the formulation according to the present invention is especially suited for subcutaneous injection.
  • The osmotic pressure ratio of the antibody-containing liquid formulation according to the present invention is preferably about 0.5 to 4, more preferably about 0.7 to 2, and still more preferably about 1.
  • The viscosity of the antibody-containing liquid formulation according to the present invention is preferably about 2 to 15 mPa·s, more preferably about 4 to 10 mPa·s. It should be noted that the viscosity described herein is measured by a rotation viscometer method using a cone-plate type viscometer, in accordance with 2.53 Viscosity Determination / General Tests , the Japanese Pharmacopoeia, 15th edition.
  • As can be seen from the results of the examples described below, according to the present invention, a stable liquid formulation can be obtained, in which dimerization and deamidation of the antibody during long-term storaize are small, by adding to the formulation arginine alone, or arginine and methionine, or methionine alone.
  • As another aspect of the present invention, a method for inhibiting deamidation in antibody-containing liquid formulations is provided, the method comprising adding to the formulation arginine or a salt thereof.
  • As still another aspect of the present invention, a method for inhibiting dimerization of antibody in antibody-containing liquid formulations is provided, the method comprising adding to the formulation arginine and methionine.
  • In the above-described two methods, the antibody is preferably anti--IL-6 receptor antibody, which is a humanized antibody or human antibody.
  • The present invention will now be described in more detail by way of the examples given below however, the scope of the present invention is not restricted thereto.
  • EXAMPLES
  • Antibody Sample
  • The humanized anti-IL-6 receptor antibody was the humanized antibody prepared in accordance with the method described in Reference Example 2 in JP 8-99902 A using the human elongation factor La promoter described in Example 10 in WO 92/19759. This antibody will occasionally be referred to as “MRA” in the tables in Examples.
  • Example 1
  • Stabilizing Effects by Combination of Arginine and Methionine
  • Liquid formulations containing anti-IL-6 receptor humanized antibody were evaluated for an influence on stabilization of the formulations obtained by use of a combination of arginine and methionine.
  • In this study, to evaluate the effects by the combination of arginine and methionine, evaluation samples numbered A1 to A9 were prepared. Prescriptions for the evaluation samples were as follows:
  • TABLE 1-1
    [Prescriptions]
    Sample Antibody Arg Met Polysorbate 80 Histidine buffer
    No. mg/mL mM mM mg/mL mM pH
    A1 180 0.5 20 6.0
    A2 180  50 0.5 20 6.0
    A3 180 100 0.5 20 6.0
    A4 180 150 0.5 20 6.0
    A5 180 200 0.5 20 6.0
    A6 180 300 0.5 20 6.0
    A7 180 100 10 0.5 20 6.0
    A8 180 100 30 0.5 20 6.0
    A9 180 100 50 0.5 20 6.0
  • To evaluate stability of the liquid formulations, each sample was subjected to a heat acceleration test (stored at 40° C. for 3 months and at 25° C. for 6 months, respectively). The purity of the antibody before and after the heat acceleration test was evaluated by gel permeation chromatography (SEC). The analytical conditions were as follows:
  • [Gel Permeation Chromatography]
  • The sample was used as the solution to he measured as it was.
  • One microliter of the solution to be measured was subjected to liquid chromatography, and the peak areas of the peaks of dimer, monomer and low molecular weight degradation products (LMW) were measured by an automatic analytical method, and the amounts thereof (%) were determined.
  • TABLE 1-2
    Analytical Conditions
    Column: TSKgel G3000SWx1 7.8 mm I.D. × 30 cm (TOSOH)
    Mobile Phase: phosphate buffer, pH 7.0 (50 mmol/L phosphate buffer, pH
    7.0, containing 300 mmol/L of sodium chloride and 0.05% sodium azide)
    Amount of Injected Sample: about 180 μg in terms of humanized anti-IL-6
    receptor antibody
    Flow Rate: 1 mL/min
    Detection Wavelength: 280 nm
  • [Formula 1]
  • Calculation Equation

  • Total Area of All Peaks=Peak Area of Monomer+Peak Area of Dinner+Peak Area of Low Molecular Weight Degradation Products (LMW)

  • Amount of Miller (%)=(Peak Area of Dimer/Total Area of All Peaks)×100

  • Amount of Low Molecular Weight Degradation Products (LMW) (%)=(Peak Area of Low Molecular Weight Degradation Products/Total Area of All Peaks)×100
  • A typical chromatography is shown in FIG 1.
  • The evaluation results obtained by the gel permeation chromatography (SEC) are shown in Table 1 and FIGS. 2 and 3. As shown, the amount of dimer the samples (Sample Nos. A2 to A6) to which arginine was added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was smaller than that in the sample (Sample No. A1) to which arginine was not added and accordingly, the inhibitory effect of arginine against dimerization was confirmed. It was also confirmed that the amount of dirtier was reduced proportionally to the amount of the arginine added. On the other hand, the amount of dimer in the samples (Sample Nos. A7 to A9) to which arginine (100 mM) and methionine were added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was smaller than that in the samples (Sample Nos. A3 and A4) containing 150 mM of arginine, which concentration was about the same as the total concentration of the stabilizers; and the amount of dimer was about the same as in the sample (Sample No. A6) having an arginine concentration of 300 mM. These results are considered to indicate that a synergistic effect in the inhibition of dimerization is obtained by combining arginine and methionine.
  • Influence of arginine and methionine on the amount f low molecular weight degradation products was not observed.
  • TABLE 1
    40° C.-3 months 25° C.-6 months
    Dimer (%) LMW (%) Dimer (%) LMW (%)
    A1 2.70 1.25 1.88 0.48
    A2 2.19 1.24 1.41 0.47
    A3 2.00 1.34 1.33 0.49
    A4 1.85 1.38 1.19 0.49
    A5 1.62 1.37 1.09 0.49
    A6 1.53 1.46 0.99 0.50
    A7 1.58 1.29 1.11 0.45
    A8 1.52 1.21 1.07 0.47
    A9 1.48 1.32 1.03 0.47
  • Example 2
  • Inhibitory Effect by Arginine against Deamidation
  • Liquid formulations containing and-IL-6 receptor humanized antibody were evaluated for influence on the deamidation by arginine.
  • In this study, evaluation samples numbered A10 to A15 and numbered A16 to A18, containing different amounts of arginine and methionine, respectively, were prepared.
  • Prescriptions for the evaluation samples were as follows:
  • TABLE 2-1
    [Prescriptions]
    Sample Antibody Arg Met Polysorbate 80 Histidine buffer
    No. mg/mL mM mM mg/mL mM pH
    A10 180 0.5 20 6.0
    A11 180  50 0.5 20 6.0
    A12 180 100 0.5 20 6.0
    A13 180 150 0.5 20 6.0
    A14 180 200 0.5 20 6.0
    A15 180 300 0.5 20 6.0
    A16 180 10 0.5 20 6.0
    A17 180 30 0.5 20 6.0
    A18 180 50 0.5 20 6.0
  • To evaluate the stability of the liquid formulations, each sample was subjected to a heat acceleration test (stored at 40° C. for 3 months and at 25° C. for 6 months, respectively). The purities of the antibody before and after the heat acceleration test were evaluated by ion-exchange chromatography (IEC). The analytical conditions were as follows:
  • [Ion-Exchange Chromatography]
  • To each sample, purified water was added to adjust the amount of the humanized anti-IL-6 receptor antibody to about 1 mg in 1 mL of the sample, and the resulting sample was used as the sample to be measured.
  • Thirty microliters of the sample solution was subjected to liquid chromatography, and the peak areas of the peaks of MRA Pre, MRA Main, MRA Sub-1,MRA Sub-2, MRA R- 1, 1Q(H)-MRA, 2Q(H)-MRA and other related substances (Others) were measured by an automatic analytical method, and the amounts thereof (%) were determined by an area percentage method.
  • MRA Pre indicates the total of the peaks of the substances each eluted after a retention time shorter than that of the main component, and a plurality of degradation products, mainly deamidation products of human ed anti-IL-6 receptor antibody, was included. When the production amount of this Pre peak was small, inhibition of deamidation of the antibody is indicated.
  • TABLE 2-2
    Analytical Conditions
    Column: ProPac WCX-10 4 × 250 mm (DIONEX)
    Mobile Phase: Solution A: 25 mmol/L MES buffer solution, pH 6.1
    Mobile Phase: Solution B: 25 mmol/L MES buffer solution, pH 6.1
    (containing 250 mmol/L of sodium chloride)
    Amount of Injected Sample: about 30 μg in terms of humanized anti-IL-6
    receptor antibody
    Flow Rate: 0.5 mL/min
    Detection Wavelength: 280 nm
  • [Formula 2]
  • Calculation Equation

  • Total Area of All Peaks=Grand Total of Total Area of MRA Pre Peaks+Peak Area of MRA Main+Peak Area of MAR Sub-1+Peak Area of MAR Sub-2+Peak Area of MAR Sub-3+Peak Area of MAR R-1+Total Area of 1Q(H)-MRA Peaks+Total Area of 2Q(H)-MRA Peaks+Peak Area of Others

  • Amount of MRA Pre (%)=(Total Area of MRA Pre Peaks/Total Area of All Peaks)×100
  • A typical chromatography is shown in FIG. 4. MRA Pre indicates the total of the peaks of the substances appealing earlier than that of the main component.
  • Evaluation results of the ion-exchange chromatography are shown Table and FIGS. 5 and 6. As shown, the amount of Pre peaks in the samples (Sample Nos. A11 to A15) to which arginine was added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was smaller than that in the sample (Sample No. A10) to which arginine was not added; and accordingly, the inhibitory effect of arginine against the generation of Pre peaks was confirmed. It was also confirmed that the amount of Pre peaks was reduced proportionally to an amount of arginine added. On the other hand, the amount of Pre peaks in the samples (Sample Nos. A16 to A18) to which methionine was added, after the acceleration at 40° C. for 3 months and at 25° C. for 6 months, respectively, was similar to the sample (Sample No. A10) to which arginine was not added; and accordingly, influence of the addition of methionine was not observed.
  • TABLE 2
    Pre peak (%)
    40° C.-3 months 25° C.-6 months
    A10 56.2 32.3
    A11 51.3 30.3
    A12 50.7 29.3
    A13 49.0 28.7
    A14 47.8 28.5
    A15 47.0 27.9
    A16 55.7 31.2
    A17 55.0 31.2
    A18 55.3 31.4
  • Example 3
  • Stabilizing Effects by Combination of Arginine and Methionine (2)
  • As in Example 1, liquid formulations containing receptor humanized antibody were evaluated for influence on stabilization of the formulations obtained by use of a combination of arginine and methionine.
  • In this study, to evaluate effects of the combination of arginine and evaluation samples numbered A19 to A27 were prepared. Prescriptions for the evaluation samples were as follows:
  • TABLE 3-1
    [Prescriptions]
    Sample Antibody Arg Met Polysorbate 80 Histidine buffer
    No. mg/mL mM mM mg/mL mM pH
    A19 180 0.5 20 6.0
    A20 180  50 0.5 20 6.0
    A21 180 100 0.5 20 6.0
    A22 180 150 0.5 20 6.0
    A23 180 200 0.5 20 6.0
    A24 180 300 0.5 20 6.0
    A25 180 100 10 0.5 20 6.0
    A26 180 100 30 0.5 20 6.0
    A27 180 100 50 0.5 20 6.0
  • To evaluate the stability of the liquid formulations, each sample was subjected to a light acceleration test (total illuminance 1,200,000 lux and total near-ultraviolet radiation energy: 200 W·h/m2). The purities of the antibody before and after the light acceleration test were evaluated by gel permeation chromatography (SEC) and ion exchange chromatography (IEC) as in Examples 1 and 2.
  • The evaluation results by the gel permeation chromatography (SEC) are shown in Table 3 and FIG. 7. As shown, the amount of dimer in the samples (Sample Nos. A20 to A24) to which arginine was added, after the light acceleration test was smaller than that in the sample (Sample No. A19) to which arginine was not added; and accordingly, the inhibitory effect of arginine against dimerization was confirmed. It was also confirmed that the amount of dimer was reduced proportionally to an amount of arginine added. On the other hand, the amount of dimer n the samples (Sample Nos. A25 to A27) to which arginine (100 mM) and methionine were added, after the light acceleration test was smaller than that in the sample (Sample No. A22) containing 150 mM of arginine, which concentration was about the same as the total concentration of the stabilizers; and the amount of dimer was smaller than in the samples (Sample Nos. A23 and A24) having arginine concentrations of 200 mM and 300 mM, respectively. These results are thought to indicate that a synergistic effect in the inhibition of dimerization is obtained by combining arginine: and methionine.
  • Influence of arginine and methionine on the amount of low molecular weight degradation products was not observed.
  • TABLE 3
    1,200,000 lux +
    200 W · h/m2
    Dimer (%) LMW (%)
    A19 6.95 0.22
    A20 6.75 0.24
    A21 5.78 0.21
    A22 5.08 0.19
    A23 4.73 0.18
    A24 4.13 0.18
    A25 5.27 0.19
    A26 4.05 0.17
    A27 3.84 0.16
  • Next, the evaluation results by the ion exchange chromatography (IEC) are shown it Table 4 and FIG. 8.
  • As shown, the amount of Pre peak in the samples (Sample Nos. A20 to A24) to which arginine was added, after the light acceleration test was smaller than that in the sample (Sample No. A19) to which arginine was not added; and accordingly, the inhibitory effect of arginine against formation of Pre peak was confirmed. Further, it was confirmed that as the amount of arginine increases, the production amount of Pre peak decreases proportionately. On the other hand, the amount of dimer after the light acceleration test in the samples (Sample Nos. A25 to A27) to which methionine was further added to arginine (100 mM) was smaller than that in the sample (Sample No. A22) containing 150 nM of arginine, which concentration was about the same as the total concentration of the stabilizers; and it was smaller than in the samples (Sample Nos. A23 and A24) having arginine concentrations of 200 mM and 300 mM, respectively. These results are thought to indicate that a synergistic effect in the inhibition of formation of Pre peak by the combination of arginine and methionine.
  • TABLE 4
    Pre peak (%)
    1,200,000 lux +
    200 W · h/m2
    A19 39.2
    A20 38.6
    A21 36.7
    A22 35.7
    A23 34.9
    A24 34.9
    A25 36.8
    A26 35.0
    A27 33.8

Claims (19)

1. A method for inhibiting dimerization of antibody molecules in a liquid formulation comprising the antibody, comprising adding arginine and methionine to the liquid formulation.
2. The method of claim 1 wherein the antibody comprises humanized anti-IL-6 receptor IgG1 antibody.
3. A method for inhibiting dimerization of humanized anti-IL-6 receptor IgG1 antibody in a liquid formulation comprising 100 to 300 mg/mL of the antibody, comprising adding 50 to 300 mM arginine and 10 to 50 mM methionine to the formulation.
4. The method of claim 3 wherein the antibody comprises the humanized anti-IL-6 receptor IgG1 antibody MRA.
5. The method of claim 3 comprising adding 100 mM arginine to the formulation.
6. The method of claim 3 further comprising adding 0.005 to 3% surfactant, and histidine buffer, pH 5.0 to 7.5, to the formulation.
7. The method of claim 6 wherein the surfactant comprises polysorbate 80.
8. The method of claim 6 wherein the pH of the formulation is 5.5 to 6.5.
9. The method of claim 6 wherein the pH of the formulation is 6.0.
10. The method of claim 3 wherein the formulation comprises 150 to 200 mg/mL of the antibody.
11. The method of claim 10 wherein the formulation comprises 180 mg/mL of the antibody.
12. The method of claim 3 wherein the formulation is for subcutaneous administration.
13. The method of claim 3 wherein the formulation has not been subjected to lyophilization during preparation thereof.
14. The method of claim 3 wherein the formulation has a viscosity of from 2 to 15 mPa.
15. The method of claim 3 wherein the formulation is stable at 22-28° C. for at least 6 months.
16. A method for preparing a stable liquid formulation suitable for subcutaneous administration comprising a humanized anti-IL-6 receptor IgG1 antibody, wherein the method comprises combining 150 to 200 mg/mL of the antibody with 50 to 300 mM arginine and 10 to 50 mM methionine.
17. The method of claim 16 wherein the formulation further comprises 0.005 to 3% surfactant, and histidine buffer, pH 5.5 to 6.5.
18. A stable liquid formulation comprising 150 to 200 mg/mL humanized anti-IL-6 receptor IgG1 antibody, 50 to 100 mM arginine, 10 to 50 mM methionine, 0.005 to 3% surfactant, and histidine buffer, pH 5.5 to 6.5.
19. The formulation of claim 18 wherein the antibody comprises the humanized anti-IL-6 receptor IgG1 antibody MRA.
US14/017,013 2007-12-26 2013-09-03 High concentration antibody-containing liquid formulation Abandoned US20140005367A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US14/017,013 US20140005367A1 (en) 2007-12-26 2013-09-03 High concentration antibody-containing liquid formulation
US14/963,414 US20160090419A1 (en) 2007-12-27 2015-12-09 High concentration antibody-containing liquid formulation
US16/390,197 US11008394B2 (en) 2007-12-27 2019-04-22 High concentration antibody-containing liquid formulation
US17/242,199 US11359026B2 (en) 2007-12-27 2021-04-27 High concentration antibody-containing liquid formulation
US17/752,978 US11584798B2 (en) 2007-12-27 2022-05-25 High concentration antibody-containing liquid formulation
US18/162,368 US11767363B2 (en) 2007-12-27 2023-01-31 High concentration antibody-containing liquid formulation
US18/335,789 US20230340134A1 (en) 2007-12-27 2023-06-15 High concentration antibody-containing liquid formulation

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2007336310 2007-12-27
JP336310/2007 2007-12-27
PCT/JP2008/073798 WO2009084659A1 (en) 2007-12-27 2008-12-26 Solution preparation containing antibody at high concentration
US81093810A 2010-06-28 2010-06-28
US14/017,013 US20140005367A1 (en) 2007-12-26 2013-09-03 High concentration antibody-containing liquid formulation

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
PCT/JP2008/073798 Division WO2009084659A1 (en) 2007-12-26 2008-12-26 Solution preparation containing antibody at high concentration
US12/810,938 Division US8568720B2 (en) 2007-12-27 2008-12-26 High concentration antibody-containing liquid formulation
US81093810A Division 2007-12-26 2010-06-28

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/963,414 Division US20160090419A1 (en) 2007-12-27 2015-12-09 High concentration antibody-containing liquid formulation

Publications (1)

Publication Number Publication Date
US20140005367A1 true US20140005367A1 (en) 2014-01-02

Family

ID=40824372

Family Applications (8)

Application Number Title Priority Date Filing Date
US12/810,938 Active 2029-11-05 US8568720B2 (en) 2007-12-27 2008-12-26 High concentration antibody-containing liquid formulation
US14/017,013 Abandoned US20140005367A1 (en) 2007-12-26 2013-09-03 High concentration antibody-containing liquid formulation
US14/963,414 Abandoned US20160090419A1 (en) 2007-12-27 2015-12-09 High concentration antibody-containing liquid formulation
US16/390,197 Active US11008394B2 (en) 2007-12-27 2019-04-22 High concentration antibody-containing liquid formulation
US17/242,199 Active US11359026B2 (en) 2007-12-27 2021-04-27 High concentration antibody-containing liquid formulation
US17/752,978 Active US11584798B2 (en) 2007-12-27 2022-05-25 High concentration antibody-containing liquid formulation
US18/162,368 Active US11767363B2 (en) 2007-12-27 2023-01-31 High concentration antibody-containing liquid formulation
US18/335,789 Pending US20230340134A1 (en) 2007-12-27 2023-06-15 High concentration antibody-containing liquid formulation

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/810,938 Active 2029-11-05 US8568720B2 (en) 2007-12-27 2008-12-26 High concentration antibody-containing liquid formulation

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/963,414 Abandoned US20160090419A1 (en) 2007-12-27 2015-12-09 High concentration antibody-containing liquid formulation
US16/390,197 Active US11008394B2 (en) 2007-12-27 2019-04-22 High concentration antibody-containing liquid formulation
US17/242,199 Active US11359026B2 (en) 2007-12-27 2021-04-27 High concentration antibody-containing liquid formulation
US17/752,978 Active US11584798B2 (en) 2007-12-27 2022-05-25 High concentration antibody-containing liquid formulation
US18/162,368 Active US11767363B2 (en) 2007-12-27 2023-01-31 High concentration antibody-containing liquid formulation
US18/335,789 Pending US20230340134A1 (en) 2007-12-27 2023-06-15 High concentration antibody-containing liquid formulation

Country Status (31)

Country Link
US (8) US8568720B2 (en)
EP (1) EP2238985B2 (en)
JP (7) JP4937358B2 (en)
KR (1) KR101083616B1 (en)
CN (2) CN101883588B (en)
AR (2) AR069969A1 (en)
AU (1) AU2008344292B2 (en)
BR (1) BRPI0818903B8 (en)
CA (1) CA2708627C (en)
CL (1) CL2008003910A1 (en)
CO (1) CO6450630A2 (en)
CR (1) CR11594A (en)
CY (1) CY1113616T1 (en)
DK (1) DK2238985T4 (en)
EC (1) ECSP10010370A (en)
ES (1) ES2389881T5 (en)
HR (1) HRP20120903T4 (en)
IL (2) IL206548A (en)
MA (1) MA31934B1 (en)
MX (1) MX2010004399A (en)
MY (1) MY159450A (en)
NZ (1) NZ586378A (en)
PE (1) PE20091174A1 (en)
PL (1) PL2238985T5 (en)
PT (1) PT2238985E (en)
RU (2) RU2497544C2 (en)
SG (1) SG2013049325A (en)
SI (1) SI2238985T2 (en)
TW (1) TWI375566B (en)
UA (1) UA104134C2 (en)
WO (1) WO2009084659A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9017677B2 (en) 1997-03-21 2015-04-28 Chugai Seiyaku Kabushiki Kaisha Methods of treating a disease mediated by sensitized T cells
US9255145B2 (en) 2001-04-02 2016-02-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for chronic arthritides diseases of childhood-related diseases
WO2016109822A1 (en) * 2014-12-31 2016-07-07 Novelmed Therapeutics, Inc. Formulation of aglycosylated therapeutic antibodies
US9539263B2 (en) 2010-11-08 2017-01-10 Genentech, Inc. Subcutaneously administered anti-IL-6 receptor antibody for treatment of systemic sclerosis
US9926366B2 (en) 2012-10-04 2018-03-27 Novelmed Therapeutics, Inc. Methods of treating a hemolytic disorder comprising administering anti-properdin antibodies
US9968676B2 (en) 2009-07-31 2018-05-15 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10744201B2 (en) 2003-04-28 2020-08-18 Chugai Seiyaku Kabushiki Kaisha Method for treating rheumatoid arthritis with a human IL-6 receptor antibody and methotrexate
US11008394B2 (en) 2007-12-27 2021-05-18 Chugai Seiyaku Kabushiki Kaisha High concentration antibody-containing liquid formulation
US11021728B2 (en) 2009-10-26 2021-06-01 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2536709T3 (en) 2002-02-14 2015-05-27 Chugai Seiyaku Kabushiki Kaisha Use of acetic acid to eliminate problems induced by the Fe ion in anti-HM1.24 or anti-IL6R antibody formulations
EP1728801A4 (en) 2004-03-24 2009-10-21 Chugai Pharmaceutical Co Ltd Subtype of humanized antibody against interleukin-6 receptor
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
EP3050963B1 (en) 2005-03-31 2019-09-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
WO2007074880A1 (en) * 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
WO2007114319A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
CN105177091A (en) 2006-03-31 2015-12-23 中外制药株式会社 Antibody modification method for purifying bispecific antibody
EP2202245B1 (en) 2007-09-26 2016-08-24 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
AU2013202276B2 (en) * 2009-12-21 2016-03-24 Genentech, Inc. Antibody formulation
CN107095846A (en) * 2009-12-21 2017-08-29 霍夫曼-拉罗奇有限公司 Antibody formulations
JO3417B1 (en) 2010-01-08 2019-10-20 Regeneron Pharma Stabilized formulations containing anti-interleukin-6 receptor (il-6r) antibodies
PT2624865T (en) * 2010-10-06 2018-11-05 Regeneron Pharma Stabilized formulations containing anti-interleukin-4 receptor (il-4r) antibodies
TWI452135B (en) 2010-11-17 2014-09-11 中外製藥股份有限公司 A multiple specific antigen-binding molecule that replaces the function of Factor VIII in blood coagulation
EP2508525A1 (en) 2011-04-05 2012-10-10 Bayer Pharma Aktiengesellschaft Substituted 2,3-dihydroimidazo[1,2-c]quinazoline salts
US20140023655A1 (en) * 2011-04-07 2014-01-23 Glaxosmithkline Llc Formulations with reduced viscosity
BR112013025845A2 (en) * 2011-04-07 2018-09-04 Glaxosmithkline Llc low viscosity formulations
UY34105A (en) * 2011-06-03 2012-07-31 Lg Life Sciences Ltd STABLE LIQUID FORMULATION OF ETANERCEPT
EP2735315B1 (en) 2011-07-19 2019-10-02 Chugai Seiyaku Kabushiki Kaisha Stable protein-containing preparation containing argininamide or valinamide
AR087305A1 (en) 2011-07-28 2014-03-12 Regeneron Pharma STABILIZED FORMULATIONS CONTAINING ANTI-PCSK9 ANTIBODIES, PREPARATION METHOD AND KIT
EP2750784A1 (en) 2011-09-01 2014-07-09 Chugai Seiyaku Kabushiki Kaisha Method for preparing a composition comprising highly concentrated antibodies by ultrafiltration
KR102276161B1 (en) 2011-10-25 2021-07-14 프로테나 바이오사이언시즈 리미티드 Antibody formulations and methods
KR101759694B1 (en) 2011-10-28 2017-07-19 인테그리티 바이오, 아이엔씨. Protein formulations containing amino acids
AU2013255413C1 (en) 2012-03-07 2016-03-24 Cadila Healthcare Limited Pharmaceutical formulations of TNF-alpha antibodies
NZ725654A (en) 2012-05-18 2018-04-27 Genentech Inc High-concentration monoclonal antibody formulations
JP6224586B2 (en) * 2012-07-10 2017-11-01 武田薬品工業株式会社 Injectable preparation
US8883979B2 (en) 2012-08-31 2014-11-11 Bayer Healthcare Llc Anti-prolactin receptor antibody formulations
TWI698253B (en) 2012-09-07 2020-07-11 美商柯赫勒斯生物科學有限公司 Stable aqueous formulations of adalimumab
MX2015004668A (en) * 2012-10-25 2015-07-23 Medimmune Llc Stable, low viscosity antibody formulation.
CA2913687C (en) 2013-07-04 2022-12-13 F. Hoffmann-La Roche Ag Interference-suppressed immunoassay to detect anti-drug antibodies in serum samples
SG10201913950XA (en) 2013-09-11 2020-03-30 Eagle Biologics Inc Liquid protein formulations containing viscosity-lowering agents
DK3050896T3 (en) 2013-09-27 2021-07-19 Chugai Pharmaceutical Co Ltd Process for the preparation of a polypeptide heteromultimer
TWI694836B (en) * 2014-05-16 2020-06-01 英商葛蘭素史克智慧財產管理有限公司 Antibody formulation
US11357857B2 (en) 2014-06-20 2022-06-14 Comera Life Sciences, Inc. Excipient compounds for protein processing
US20160074515A1 (en) 2014-06-20 2016-03-17 Reform Biologics, Llc Viscosity-reducing excipient compounds for protein formulations
US10478498B2 (en) 2014-06-20 2019-11-19 Reform Biologics, Llc Excipient compounds for biopolymer formulations
KR20170057339A (en) * 2014-09-15 2017-05-24 제넨테크, 인크. Antibody formulations
TWI701435B (en) 2014-09-26 2020-08-11 日商中外製藥股份有限公司 Method to determine the reactivity of FVIII
TWI700300B (en) 2014-09-26 2020-08-01 日商中外製藥股份有限公司 Antibodies that neutralize substances with the function of FVIII coagulation factor (FVIII)
AU2015325055B2 (en) 2014-10-01 2021-02-25 Eagle Biologics, Inc. Polysaccharide and nucleic acid formulations containing viscosity-lowering agents
PL3224341T3 (en) 2014-11-25 2021-11-08 Corning Incorporated Cell culture media extending materials and methods
AR103173A1 (en) * 2014-12-22 2017-04-19 Novarits Ag PHARMACEUTICAL PRODUCTS AND STABLE LIQUID COMPOSITIONS OF ANTIBODIES IL-17
JP6731953B2 (en) 2015-02-11 2020-07-29 ジーエムエーエックス バイオファーム エルエルシー. Stable pharmaceutical solution formulation of GLP-1R antibody fusion protein
AR103726A1 (en) 2015-02-27 2017-05-31 Merck Sharp & Dohme HUMAN ANTI-PD-1 MONOCLONAL ANTIBODY CRYSTALS
US11142587B2 (en) 2015-04-01 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Method for producing polypeptide hetero-oligomer
MA42657A (en) 2015-08-18 2018-06-27 Regeneron Pharma ANTI-PCSK9 ANTIBODY INHIBITORS FOR THE TREATMENT OF PATIENTS WITH HYPERLIPIDEMIA SUBJECTING LIPOPROTEIN APHERESIS
TW201726111A (en) * 2015-09-30 2017-08-01 持田製藥股份有限公司 Liquid pharmaceutical agent comprising high concentration antibody
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
TWI787161B (en) * 2015-12-18 2022-12-21 日商安斯泰來製藥股份有限公司 Pharmaceutical composition comprising anti-human tslp receptor antibody
WO2017115773A1 (en) 2015-12-28 2017-07-06 中外製薬株式会社 Method for promoting efficiency of purification of fc region-containing polypeptide
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
JP7104629B2 (en) * 2016-02-24 2022-07-21 ビステラ, インコーポレイテッド Preparation of antibody molecule against influenza virus
WO2017184880A1 (en) 2016-04-20 2017-10-26 Coherus Biosciences, Inc. A method of filling a container with no headspace
AU2017255077A1 (en) * 2016-04-28 2018-10-04 Chugai Seiyaku Kabushiki Kaisha Antibody-containing preparation
TWI826351B (en) * 2016-05-31 2023-12-21 大陸商鴻運華寧(杭州)生物醫藥有限公司 R antibodies, their pharmaceutical compositions and uses
CN109661240B (en) 2016-07-05 2022-11-29 赛诺菲 Antibody formulations
RU2766233C2 (en) 2016-09-06 2022-02-10 Чугаи Сейяку Кабусики Кайся Methods for use of bispecific antibody that recognizes clotting factor ix and/or activated clotting factor ix and clotting factor x and/or activated clotting factor x
US10961314B2 (en) 2016-09-27 2021-03-30 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition comprising an anti-IL-6 receptor antibody
HUE054858T2 (en) 2016-10-31 2021-10-28 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
WO2018116198A1 (en) 2016-12-23 2018-06-28 Serum Institute Of India Private Limited Improved methods for enhancing antibody productivity in mammalian cell culture and minimizing aggregation during downstream, formulation processes and stable antibody formulations obtained thereof
US11203636B2 (en) 2017-02-01 2021-12-21 Yale University Treatment of existing left ventricular heart failure
US11608357B2 (en) 2018-08-28 2023-03-21 Arecor Limited Stabilized antibody protein solutions
GB201703063D0 (en) 2017-02-24 2017-04-12 Arecor Ltd Stabilized antibody protein solutions
TW202228779A (en) 2017-03-01 2022-08-01 英商梅迪繆思有限公司 Anti-rsv monoclonal antibody formulation
EP3372241A1 (en) 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
EP3372242A1 (en) 2017-03-06 2018-09-12 Ares Trading S.A. Liquid pharmaceutical composition
WO2018170405A1 (en) 2017-03-17 2018-09-20 Ohio State Innovation Foundation Nanoparticles for delivery of chemopreventive agents
WO2018179138A1 (en) * 2017-03-29 2018-10-04 持田製薬株式会社 Antibody-containing liquid preparation
JP7179717B2 (en) 2017-03-31 2022-11-29 Meiji Seikaファルマ株式会社 Aqueous formulation, aqueous formulation containing syringe, antibody protein disaggregation agent, and antibody protein disaggregation method
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
KR20200003107A (en) * 2017-05-02 2020-01-08 머크 샤프 앤드 돔 코포레이션 Preparation of Anti-LAG3 Antibodies, and Co-Formulations of Anti-LAG3 Antibodies and Anti-PD-1 Antibodies
WO2018211517A1 (en) 2017-05-16 2018-11-22 Bhami's Research Laboratory, Pvt. Ltd. High concentration protein formulations with reduced viscosity
KR20200044066A (en) * 2017-08-22 2020-04-28 바이오젠 엠에이 인코포레이티드 Pharmaceutical compositions and dosing regimens containing anti-alpha (V) beta (6) antibodies
WO2019040612A1 (en) * 2017-08-22 2019-02-28 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies
KR20200049764A (en) 2017-09-29 2020-05-08 추가이 세이야쿠 가부시키가이샤 Multispecific antigen-binding molecules having blood coagulation factor viii (fviii) cofactor function-substituting activity and pharmaceutical formulations containing such a molecule as an active ingredient
AU2019205936B2 (en) 2018-01-05 2022-09-15 Novo Nordisk A/S Methods for treating IL-6 mediated inflammation without immunosuppression
CA3099551A1 (en) 2018-05-10 2019-11-14 Regeneron Pharmaceuticals, Inc. High concentration vegf receptor fusion protein containing formulations
CN113164618A (en) 2018-09-12 2021-07-23 希沃尔拜克治疗公司 Methods and compositions for treating diseases with immunostimulatory conjugates
KR20210136063A (en) * 2019-03-11 2021-11-16 바이오젠 엠에이 인코포레이티드 Pharmaceutical composition comprising anti-LINGO-1 antibody
JP2022527972A (en) 2019-04-02 2022-06-07 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル How to predict and prevent cancer in patients with premalignant lesions
US20200347120A1 (en) * 2019-04-30 2020-11-05 Medimmune Limited Dosage regimens for and compositions including anti-rsv antibodies
KR102169901B1 (en) * 2019-05-17 2020-10-26 연세대학교 산학협력단 Methods for providing information about responses to cancer immunotherapy using dna methylation and kits using the same
KR20220035908A (en) 2019-06-19 2022-03-22 실버백 테라퓨틱스, 인크. Anti-Mesothelin Antibodies and Immunoconjugates Thereof
CN112618482A (en) * 2019-09-24 2021-04-09 江苏恒瑞医药股份有限公司 Novel protein formulations
EP4106819A1 (en) 2020-02-21 2022-12-28 Silverback Therapeutics, Inc. Nectin-4 antibody conjugates and uses thereof
US20240025991A1 (en) 2020-03-23 2024-01-25 Genentech, Inc. Method for treating pneumonia, including covid-19 pneumonia, with an il6 antagonist
CN115867577A (en) 2020-03-23 2023-03-28 基因泰克公司 Biomarkers for predicting response to IL-6 antagonists in COVID-19 pneumonia
JP2023518812A (en) 2020-03-23 2023-05-08 ジェネンテック, インコーポレイテッド Combination of tocilizumab and remdesivir to treat COVID19 pneumonia
WO2022005113A1 (en) * 2020-06-29 2022-01-06 한올바이오파마주식회사 Formulation for anti-fcrn antibody
WO2022006327A1 (en) 2020-07-01 2022-01-06 Silverback Therapeutics, Inc. Anti-asgr1 antibody conjugates and uses thereof
KR20220028972A (en) * 2020-08-31 2022-03-08 (주)셀트리온 Stable Pharmaceutical Formulation
US20230357418A1 (en) 2020-09-17 2023-11-09 Genentech, Inc. Results of empacta: a randomized, double-blind, placebo-controlled, multicenter study to evaluate the efficacy and safety of tocilizumab in hospitalized patients with covid-19 pneumonia
US20230399402A1 (en) * 2020-11-03 2023-12-14 La Jolla Institute For Immunology Hla class ii-restricted tcrs against the kras g12>v activating mutation

Family Cites Families (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS55106216A (en) 1979-02-09 1980-08-14 Hitachi Chem Co Ltd Preparation of amino resin
JPS58201994A (en) 1982-05-21 1983-11-25 Hideaki Hagiwara Method for producing antigen-specific human immunoglobulin
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
JPS63453A (en) 1986-06-20 1988-01-05 Tohoku Metal Ind Ltd Oxidation resistant permanent magnet material and its production
US5171840A (en) 1988-01-22 1992-12-15 Tadamitsu Kishimoto Receptor protein for human B cell stimulatory factor-2
US6428979B1 (en) 1988-01-22 2002-08-06 Tadamitsu Kishimoto Receptor protein for human B cell stimulatory factor-2
US5670373A (en) 1988-01-22 1997-09-23 Kishimoto; Tadamitsu Antibody to human interleukin-6 receptor
US5158761A (en) * 1989-04-05 1992-10-27 Toko Yakuhin Kogyo Kabushiki Kaisha Spray gel base and spray gel preparation using thereof
ATE185601T1 (en) 1990-07-10 1999-10-15 Cambridge Antibody Tech METHOD FOR PRODUCING SPECIFIC BONDING PAIRS
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
ES2108048T3 (en) 1990-08-29 1997-12-16 Genpharm Int PRODUCTION AND USE OF LOWER TRANSGENIC ANIMALS CAPABLE OF PRODUCING HETEROLOGICAL ANTIBODIES.
WO1992019759A1 (en) 1991-04-25 1992-11-12 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
ATE181571T1 (en) 1991-09-23 1999-07-15 Medical Res Council METHODS FOR PRODUCING HUMANIZED ANTIBODIES
ATE463573T1 (en) 1991-12-02 2010-04-15 Medimmune Ltd PRODUCTION OF AUTOANTIBODIES ON PHAGE SURFACES BASED ON ANTIBODIES SEGMENT LIBRARIES
EP0746609A4 (en) 1991-12-17 1997-12-17 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
WO1993019172A1 (en) 1992-03-24 1993-09-30 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
AU675661B2 (en) 1992-07-24 1997-02-13 Abgenix, Inc. Generation of xenogeneic antibodies
AU6819494A (en) 1993-04-26 1994-11-21 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
GB9313509D0 (en) 1993-06-30 1993-08-11 Medical Res Council Chemisynthetic libraries
US5888510A (en) 1993-07-21 1999-03-30 Chugai Seiyaku Kabushiki Kaisha Chronic rheumatoid arthritis therapy containing IL-6 antagonist as effective component
AU690171B2 (en) 1993-12-03 1998-04-23 Medical Research Council Recombinant binding proteins and peptides
US8017121B2 (en) 1994-06-30 2011-09-13 Chugai Seiyaku Kabushika Kaisha Chronic rheumatoid arthritis therapy containing IL-6 antagonist as effective component
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
WO1996012503A1 (en) 1994-10-21 1996-05-02 Chugai Seiyaku Kabushiki Kaisha Remedy for diseases caused by il-6 production
AU704723B2 (en) 1994-12-29 1999-04-29 Chugai Seiyaku Kabushiki Kaisha Antitumor agent effect enhancer containing IL-6 antagonist
PT811384E (en) 1995-02-13 2006-11-30 Chugai Pharmaceutical Co Ltd Muscle protein decomposition inhibitor containing il-6 receptor antibody
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
ES2263178T3 (en) 1996-06-27 2006-12-01 Chugai Seiyaku Kabushiki Kaisha REMEDIES FOR MYELOMA TO BE USED TOGETHER WITH NITROGEN MOSTAZA ANTITUMORAL AGENTS.
EP0962467A4 (en) 1996-09-26 2002-10-30 Chugai Pharmaceutical Co Ltd 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
CA2284271C (en) 1997-03-21 2012-05-08 Chugai Seiyaku Kabushiki Kaisha A preventive or therapeutic agent for sensitized t cell-mediated diseases comprising il-6 antagonist as an active ingredient
US6991790B1 (en) 1997-06-13 2006-01-31 Genentech, Inc. Antibody formulation
US20020187150A1 (en) 1997-08-15 2002-12-12 Chugai Seiyaku Kabushiki Kaisha Preventive and/or therapeutic agent for systemic lupus erythematosus comprising anti-IL-6 receptor antibody as an active ingredient
DE69937994T2 (en) 1998-03-17 2008-12-24 Chugai Seiyaku K.K. PROPHYLACTIC OR THERAPEUTIC AGENTS AGAINST INFLAMMATORY DISEASES OF THE DIGESTIVE STRUCTURE CONTAINED ANTAGONISTIC IL-6 RECEPTOR ANTIBODIES
CN1303431A (en) 1998-04-03 2001-07-11 中外制药株式会社 Humanized antibody against human tissue factor (TF) and process for constructing humanized antibody
US6537782B1 (en) 1998-06-01 2003-03-25 Chugai Seiyaku Kabushiki Kaisha Media for culturing animal cells and process for producing protein by using the same
US6406909B1 (en) 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
EP1108435B1 (en) 1998-08-24 2007-01-03 Chugai Seiyaku Kabushiki Kaisha Preventives or remedies for pancreatitis containing anti-il-6 receptor antibodies as the active ingredient
JP2000247903A (en) 1999-03-01 2000-09-12 Chugai Pharmaceut Co Ltd Long-term stabilized pharmaceutical preparation
JP2007204498A (en) * 1999-03-01 2007-08-16 Chugai Pharmaceut Co Ltd Long-term stabilized formulations
US6914128B1 (en) * 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
AU784045B2 (en) * 1999-06-25 2006-01-19 Genentech Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
PL211886B1 (en) 1999-10-04 2012-07-31 Novartis Vaccines & Diagnostic Stabilized liquid polypeptide-containing pharmaceutical compositions
US7163671B2 (en) 2000-02-29 2007-01-16 Chugai Seiyaku Kabushiki Kaisha Long-term stabilized formulations
TWI242043B (en) 2000-03-10 2005-10-21 Chugai Pharmaceutical Co Ltd Polypeptide inducing apoptosis
AU2001278716A1 (en) 2000-08-10 2002-02-25 Chugai Seiyaku Kabushiki Kaisha Method of inhibiting antibody-containing solution from coagulating or becoming turbid
PT1314437E (en) * 2000-08-11 2014-08-29 Chugai Pharmaceutical Co Ltd Stabilized antibody-containing preparations
EP1930024A3 (en) 2000-09-01 2008-08-06 Chugai Seiyaku Kabushiki Kaisha G-CSF solution formulations having long-term stability
US8703126B2 (en) 2000-10-12 2014-04-22 Genentech, Inc. Reduced-viscosity concentrated protein formulations
AU1344102A (en) * 2000-10-12 2002-04-22 Genentech Inc Reduced-viscosity concentrated protein formulations
WO2002033072A1 (en) 2000-10-20 2002-04-25 Chugai Seiyaku Kabushiki Kaisha Degraded tpo agonist antibody
ES2298273T3 (en) 2000-10-25 2008-05-16 Chugai Seiyaku Kabushiki Kaisha PREVENTIVE OR THERAPEUTIC AGENTS AGAINST PSORIASIS CONTAINING AN IL-6 ANTAGONIST AS THEIR ACTIVE INGREDIENT.
WO2002036164A1 (en) 2000-10-27 2002-05-10 Chugai Seiyaku Kabushiki Kaisha Blooe vegf level-lowering agent containing il-6 antagonist as the active ingredient
JP4889187B2 (en) 2000-10-27 2012-03-07 中外製薬株式会社 A blood MMP-3 concentration reducing agent comprising an IL-6 antagonist as an active ingredient
ES2869895T3 (en) 2001-03-09 2021-10-26 Chugai Pharmaceutical Co Ltd Protein purification method
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
US20030138417A1 (en) 2001-11-08 2003-07-24 Kaisheva Elizabet A. Stable liquid pharmaceutical formulation of IgG antibodies
NZ533223A (en) * 2001-11-14 2007-04-27 Centocor Inc Anti-il-6 antibodies, compositions, methods and uses
US20030138416A1 (en) * 2001-12-03 2003-07-24 Jesper Lau Use of glucokinase activator in combination with a glucagon antagonist for treating type 2 diabetes
ES2536709T3 (en) 2002-02-14 2015-05-27 Chugai Seiyaku Kabushiki Kaisha Use of acetic acid to eliminate problems induced by the Fe ion in anti-HM1.24 or anti-IL6R antibody formulations
US20030180287A1 (en) * 2002-02-27 2003-09-25 Immunex Corporation Polypeptide formulation
JP3822137B2 (en) 2002-05-20 2006-09-13 中外製薬株式会社 Additive for animal cell culture medium and method for producing protein using the same
PL376536A1 (en) 2002-08-28 2006-01-09 Immunex Corporation Compositions and methods for treating cardiovascular disease
US8420789B2 (en) 2002-09-11 2013-04-16 Chugai Seiyaku Kabushiki Kaisha Method for removing DNA contaminants from a protein-containing sample
DE10255508A1 (en) 2002-11-27 2004-06-17 Forschungszentrum Jülich GmbH Process for cultivating cells for the production of substances
AU2003293543A1 (en) * 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
US20060165696A1 (en) 2003-02-24 2006-07-27 Chugai Seiyaku Kabushiki Kaisha Keio University Remedy for spinal injury containing interleukin-6 antagonist
PT2335725T (en) 2003-04-04 2017-01-06 Novartis Ag High concentration antibody and protein formulations
US20050158303A1 (en) 2003-04-04 2005-07-21 Genentech, Inc. Methods of treating IgE-mediated disorders comprising the administration of high concentration anti-IgE antibody formulations
GB2401040A (en) 2003-04-28 2004-11-03 Chugai Pharmaceutical Co Ltd Method for treating interleukin-6 related diseases
EP1652925B1 (en) 2003-07-15 2010-12-08 Chugai Seiyaku Kabushiki Kaisha IgM PRODUCTION BY TRANSFORMED CELLS AND METHODS FOR QUANTIFYING SAID IgM PRODUCTION
WO2005037315A1 (en) 2003-10-17 2005-04-28 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for mesothelioma
EP1532983A1 (en) 2003-11-18 2005-05-25 ZLB Bioplasma AG Immunoglobulin preparations having increased stability
DE10355251A1 (en) 2003-11-26 2005-06-23 Merck Patent Gmbh Water-based pharmaceutical preparation for treatment of tumors has active ingredient effective against receptor of endothelial growth factor receptor
AR048210A1 (en) 2003-12-19 2006-04-12 Chugai Pharmaceutical Co Ltd A PREVENTIVE AGENT FOR VASCULITIS.
US8617550B2 (en) 2003-12-19 2013-12-31 Chugai Seiyaku Kabushiki Kaisha Treatment of vasculitis with IL-6 antagonist
EP1712240B1 (en) 2003-12-25 2015-09-09 Kyowa Hakko Kirin Co., Ltd. Stable water-based medicinal preparation containing antibody
JP2007522157A (en) 2004-02-12 2007-08-09 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Highly concentrated liquid formulation of anti-EGFR antibody
WO2005089503A2 (en) * 2004-03-19 2005-09-29 Progenics Pharmaceuticals, Inc. Cd4-igg2 formulations
AR048335A1 (en) 2004-03-24 2006-04-19 Chugai Pharmaceutical Co Ltd THERAPEUTIC AGENTS FOR INTERNAL EAR DISORDERS CONTAINING AN IL-6 ANTAGONIST AS AN ACTIVE INGREDIENT
EP1728801A4 (en) 2004-03-24 2009-10-21 Chugai Pharmaceutical Co Ltd Subtype of humanized antibody against interleukin-6 receptor
WO2006023665A2 (en) * 2004-08-17 2006-03-02 Regeneron Pharmaceuticals, Inc. Il-1 antagonist formulations
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US9714410B2 (en) 2005-01-05 2017-07-25 Chugai Seiyaku Kabushiki Kaisha Cell culture method and utilization of the same
SV2008002394A (en) 2005-01-28 2008-02-08 Wyeth Corp POLYPEPTIDE STABILIZED LIQUID FORMULATIONS REF. AHN- 072SV
NZ623901A (en) 2005-08-03 2015-10-30 Immunogen Inc Immunoconjugate formulations
AR061400A1 (en) 2005-10-14 2008-08-27 Chugai Kabushiki Kaisha AGENTS TO DELETE THE DANE TO TRANSPLANTED ISLOTS AFTER THE ISLOT TRANSPLANT
WO2007046489A1 (en) 2005-10-21 2007-04-26 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for heart disease
AR057582A1 (en) 2005-11-15 2007-12-05 Nat Hospital Organization AGENTS TO DELETE INDUCTION OF CYTOTOXIC T LYMPHOCYTES
AR057941A1 (en) 2005-11-25 2007-12-26 Univ Keio THERAPEUTIC AGENTS FOR PROSTATE CANCER
WO2007074880A1 (en) 2005-12-28 2007-07-05 Chugai Seiyaku Kabushiki Kaisha Antibody-containing stabilizing preparation
AU2007208678B2 (en) 2006-01-27 2013-01-10 Chugai Seiyaku Kabushiki Kaisha Therapeutic agents for diseases involving choroidal neovascularization
WO2007092772A2 (en) * 2006-02-03 2007-08-16 Medimmune, Inc. Protein formulations
BRPI0708677A2 (en) 2006-03-09 2011-06-21 Hoffmann La Roche anti-drug antibody assay
TW200806317A (en) 2006-03-20 2008-02-01 Wyeth Corp Methods for reducing protein aggregation
WO2007116962A1 (en) 2006-04-07 2007-10-18 Osaka University Muscle regeneration promoter
US8945564B2 (en) 2006-04-21 2015-02-03 Novartis Ag Antagonist anti-CD40 antibody pharmaceutical compositions
WO2008016134A1 (en) 2006-08-04 2008-02-07 Norihiro Nishimoto Method for predicting prognosis of rheumatoid arthritis patients
JP2010095445A (en) 2006-12-27 2010-04-30 Tokyo Medical & Dental Univ Therapeutic agent for inflammatory myopathy containing il-6 antagonist as active ingredient
WO2008086395A2 (en) 2007-01-09 2008-07-17 Wyeth Anti-il-13 antibody formulations and uses thereof
RU2450829C2 (en) 2007-01-23 2012-05-20 Синсу Юниверсити Chronic rejection inhibitor
JP5424330B2 (en) 2007-07-26 2014-02-26 国立大学法人大阪大学 A therapeutic agent for ocular inflammatory diseases comprising an interleukin 6 receptor inhibitor as an active ingredient
AU2008304756B8 (en) 2007-09-26 2015-02-12 Chugai Seiyaku Kabushiki Kaisha Anti-IL-6 receptor antibody
RU2490025C2 (en) 2007-10-02 2013-08-20 Чугаи Сейяку Кабусики Кайся Therapeutic agent used for graft-versus-host disease, containing interleukin-6 receptor inhibitor as active ingredient
JP2009092508A (en) 2007-10-09 2009-04-30 Norihiro Nishimoto Method for predicting effect of rheumatic therapeutic agent
AU2008337904B2 (en) 2007-12-15 2014-02-13 F. Hoffmann-La Roche Ag Distinguishing assay
PE20091174A1 (en) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd LIQUID FORMULATION WITH HIGH CONCENTRATION OF ANTIBODY CONTENT
JP5544290B2 (en) 2008-06-05 2014-07-09 独立行政法人国立がん研究センター Nerve infiltration inhibitor
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
WO2011013786A1 (en) 2009-07-31 2011-02-03 Maeda Shin Cancer metastasis inhibitor
KR101436219B1 (en) 2009-10-26 2014-09-01 에프. 호프만-라 로슈 아게 Method for the production of a glycosylated immunoglobulin
TWI505838B (en) 2010-01-20 2015-11-01 Chugai Pharmaceutical Co Ltd Stabilized antibody
WO2011128096A1 (en) 2010-04-16 2011-10-20 Roche Diagnostics Gmbh Polymorphism markers for predicting response to interleukin-6 receptor-inhibiting monoclonal antibody drug treatment
RS63800B1 (en) 2010-05-28 2022-12-30 Chugai Pharmaceutical Co Ltd Antitumor t cell response enhancer
US20130149302A1 (en) 2010-05-28 2013-06-13 Chugai Seiyaku Kabushiki Kaisha Therapeutic agents for pancreatic cancer
CA2801107A1 (en) 2010-06-07 2011-12-15 F. Hoffman-La Roche Ag Gene expression markers for predicting response to interleukin-6 receptor-inhibiting monoclonal antibody drug treatment
KR20220070586A (en) 2010-11-08 2022-05-31 제넨테크, 인크. Subcutaneously administered anti-il-6 receptor antibody
EP2750784A1 (en) 2011-09-01 2014-07-09 Chugai Seiyaku Kabushiki Kaisha Method for preparing a composition comprising highly concentrated antibodies by ultrafiltration
CA2913687C (en) 2013-07-04 2022-12-13 F. Hoffmann-La Roche Ag Interference-suppressed immunoassay to detect anti-drug antibodies in serum samples
US10349940B2 (en) 2015-08-24 2019-07-16 Ethicon Llc Surgical stapler buttress applicator with state indicator
US10961314B2 (en) 2016-09-27 2021-03-30 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition comprising an anti-IL-6 receptor antibody
HUE054858T2 (en) 2016-10-31 2021-10-28 Fresenius Kabi Deutschland Gmbh Liquid pharmaceutical composition
CN109350740B (en) 2017-11-30 2023-06-20 百奥泰生物制药股份有限公司 Liquid preparation of humanized antibody for treating IL-6 related diseases

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9017677B2 (en) 1997-03-21 2015-04-28 Chugai Seiyaku Kabushiki Kaisha Methods of treating a disease mediated by sensitized T cells
US9255145B2 (en) 2001-04-02 2016-02-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic agent for chronic arthritides diseases of childhood-related diseases
US10744201B2 (en) 2003-04-28 2020-08-18 Chugai Seiyaku Kabushiki Kaisha Method for treating rheumatoid arthritis with a human IL-6 receptor antibody and methotrexate
US11767363B2 (en) 2007-12-27 2023-09-26 Chugai Seiyaku Kabushiki Kaisha High concentration antibody-containing liquid formulation
US11584798B2 (en) 2007-12-27 2023-02-21 Hoffmann-La Roche Inc. High concentration antibody-containing liquid formulation
US11359026B2 (en) 2007-12-27 2022-06-14 Chugai Seiyaku Kabushiki Kaisha High concentration antibody-containing liquid formulation
US11008394B2 (en) 2007-12-27 2021-05-18 Chugai Seiyaku Kabushiki Kaisha High concentration antibody-containing liquid formulation
US9968676B2 (en) 2009-07-31 2018-05-15 Genentech, Inc. Subcutaneous anti-HER2 antibody formulations and uses thereof
US10752696B2 (en) 2009-09-11 2020-08-25 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10280227B2 (en) 2009-09-11 2019-05-07 Genentech, Inc. Highly concentrated pharmaceutical formulations
US10377831B2 (en) 2009-09-11 2019-08-13 Genentech, Inc. Highly concentrated pharmaceutical formulations
US11136610B2 (en) 2009-10-26 2021-10-05 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US11021728B2 (en) 2009-10-26 2021-06-01 Hoffmann-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US11377678B2 (en) 2009-10-26 2022-07-05 Hoffman-La Roche Inc. Method for the production of a glycosylated immunoglobulin
US10874677B2 (en) 2010-11-08 2020-12-29 Hoffmann-La Roche Inc. Subcutaneously administered anti-IL-6 receptor antibody
US10231981B2 (en) 2010-11-08 2019-03-19 Chugai Seiyaku Kabushiki Kaisha Subcutaneously administered anti-IL-6 receptor antibody for treatment of juvenile idiopathic arthritis
US9750752B2 (en) 2010-11-08 2017-09-05 Hoffmann-La Roche Inc. Subcutaneously administered anti-IL-6 receptor antibody
US9539263B2 (en) 2010-11-08 2017-01-10 Genentech, Inc. Subcutaneously administered anti-IL-6 receptor antibody for treatment of systemic sclerosis
US11622969B2 (en) 2010-11-08 2023-04-11 Hoffmann-La Roche Inc. Subcutaneously administered anti-IL-6 receptor antibody
US11667720B1 (en) 2010-11-08 2023-06-06 Hoffmann-La Roche Inc. Subcutaneously administered anti-IL-6 receptor antibody
US9926366B2 (en) 2012-10-04 2018-03-27 Novelmed Therapeutics, Inc. Methods of treating a hemolytic disorder comprising administering anti-properdin antibodies
WO2016109822A1 (en) * 2014-12-31 2016-07-07 Novelmed Therapeutics, Inc. Formulation of aglycosylated therapeutic antibodies

Also Published As

Publication number Publication date
US20230340134A1 (en) 2023-10-26
US20210246216A1 (en) 2021-08-12
MY159450A (en) 2017-01-13
JP6259436B2 (en) 2018-01-10
JP5906067B2 (en) 2016-04-20
CR11594A (en) 2010-10-05
AU2008344292A1 (en) 2009-07-09
DK2238985T4 (en) 2022-10-17
KR101083616B1 (en) 2011-11-16
NZ586378A (en) 2012-06-29
HRP20120903T1 (en) 2012-12-31
IL206548A (en) 2015-06-30
US11008394B2 (en) 2021-05-18
JPWO2009084659A1 (en) 2011-05-19
SI2238985T2 (en) 2022-09-30
BRPI0818903A2 (en) 2015-05-12
US11359026B2 (en) 2022-06-14
AR122312A2 (en) 2022-08-31
IL206548A0 (en) 2010-12-30
SI2238985T1 (en) 2012-12-31
CA2708627A1 (en) 2009-07-09
BRPI0818903B1 (en) 2021-04-06
ECSP10010370A (en) 2010-08-31
CA2708627C (en) 2016-10-11
HRP20120903T4 (en) 2022-09-02
EP2238985A4 (en) 2011-01-12
CN106075434B (en) 2020-02-14
WO2009084659A1 (en) 2009-07-09
US20230167183A1 (en) 2023-06-01
ES2389881T5 (en) 2022-10-07
TW200942259A (en) 2009-10-16
JP7256234B2 (en) 2023-04-11
EP2238985B2 (en) 2022-07-27
CN101883588A (en) 2010-11-10
JP2018076334A (en) 2018-05-17
AR069969A1 (en) 2010-03-03
US8568720B2 (en) 2013-10-29
JP6567024B2 (en) 2019-08-28
IL238896A (en) 2016-06-30
US20220281988A1 (en) 2022-09-08
US20100285011A1 (en) 2010-11-11
US11767363B2 (en) 2023-09-26
RU2019128863A (en) 2021-03-16
IL238896A0 (en) 2015-07-30
ES2389881T3 (en) 2012-11-02
CO6450630A2 (en) 2012-05-31
EP2238985B9 (en) 2012-12-26
EP2238985A1 (en) 2010-10-13
TWI375566B (en) 2012-11-01
EP2238985B1 (en) 2012-08-29
CN106075434A (en) 2016-11-09
US11584798B2 (en) 2023-02-21
CN101883588B (en) 2017-05-10
MA31934B1 (en) 2010-12-01
JP2021155453A (en) 2021-10-07
MX2010004399A (en) 2010-08-16
JP2016065079A (en) 2016-04-28
BRPI0818903B8 (en) 2021-05-25
JP4937358B2 (en) 2012-05-23
US20200079857A1 (en) 2020-03-12
RU2010131179A (en) 2012-02-10
AU2008344292B2 (en) 2012-11-08
RU2013137740A (en) 2015-02-20
US20160090419A1 (en) 2016-03-31
PT2238985E (en) 2012-11-28
ES2389881T9 (en) 2013-01-15
KR20100095474A (en) 2010-08-30
RU2497544C2 (en) 2013-11-10
JP2019206559A (en) 2019-12-05
DK2238985T3 (en) 2012-11-19
UA104134C2 (en) 2014-01-10
RU2701181C2 (en) 2019-09-25
CY1113616T1 (en) 2016-06-22
PE20091174A1 (en) 2009-08-03
JP2023055780A (en) 2023-04-18
JP2012072170A (en) 2012-04-12
PL2238985T3 (en) 2013-03-29
SG2013049325A (en) 2015-01-29
CL2008003910A1 (en) 2009-07-03
PL2238985T5 (en) 2023-07-17

Similar Documents

Publication Publication Date Title
US11359026B2 (en) High concentration antibody-containing liquid formulation
US9084777B2 (en) Stabilized antibody-containing formulations
US20050214278A1 (en) Antibody-containing solution pharmaceuticals
RU2795116C2 (en) Liquid composition containing high concentration antibody

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: HOFFMANN-LA ROCHE INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:F. HOFFMANN-LA ROCHE AG;REEL/FRAME:058058/0023

Effective date: 20211105