US20140093496A1 - Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY - Google Patents

Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY Download PDF

Info

Publication number
US20140093496A1
US20140093496A1 US14/001,218 US201214001218A US2014093496A1 US 20140093496 A1 US20140093496 A1 US 20140093496A1 US 201214001218 A US201214001218 A US 201214001218A US 2014093496 A1 US2014093496 A1 US 2014093496A1
Authority
US
United States
Prior art keywords
numbering
substitution
fcγriib
polypeptide
fcγriia
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
US14/001,218
Other languages
English (en)
Inventor
Futa Mimoto
Taichi Kuramochi
Tomoyuki Igawa
Hitoshi Katada
Shojiro Kadono
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46721013&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140093496(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
Assigned to CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment CHUGAI SEIYAKU KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KURAMOCHI, TAICHI, IGAWA, TOMOYUKI, KATADA, Hitoshi, MIMOTO, FUTA, KADONO, SHOJIRO
Publication of US20140093496A1 publication Critical patent/US20140093496A1/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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/303Liver or Pancreas
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • 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
    • C07K2317/72Increased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to polypeptides comprising an IgG Fc region that have maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide by introducing amino acid substitutions into the IgG Fc region; pharmaceutical compositions comprising the polypeptide; therapeutic agents or preventive agents comprising the polypeptide for immunological inflammatory diseases; and methods for producing them.
  • the present invention relates to methods for maintaining or decreasing binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and enhancing Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide; and methods for suppressing antibody production compared with the parent polypeptide in in vivo administration.
  • the present invention also relates to methods for producing a polypeptide having maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide; and methods for producing a polypeptide that suppresses antibody production compared with a parent polypeptide in in vivo administration.
  • Non-patent Documents 1 and 2 Antibodies are drawing attention as pharmaceuticals since they are highly stable in blood and have few side effects. Almost all antibody pharmaceuticals currently on the market are antibodies of the human IgG1 subclass.
  • One of the known functions of IgG class antibodies is antibody-dependent cell-mediated cytotoxicity (hereinafter denoted as ADCC activity) (Non-patent Document 3).
  • ADCC activity antibody-dependent cell-mediated cytotoxicity
  • the antibody Fc region must bind to an Fc ⁇ receptor (hereinafter denoted as Fc ⁇ R) which is an antibody-binding receptor present on the surface of effector cells such as killer cells, natural killer cells, and activated macrophages.
  • Fc ⁇ R an Fc ⁇ receptor
  • Fc ⁇ RIa CD64A
  • Fc ⁇ RIIa CD32A
  • Fc ⁇ RIIb CD32B
  • Fc ⁇ RIIIa CD16A
  • Fc ⁇ RIIIb CD16B isoforms
  • Fc ⁇ R protein family the Fc ⁇ R protein family, and the respective allotypes have also been reported (Non-patent Document 7).
  • Fc ⁇ RIa, Fc ⁇ RIIa, and Fc ⁇ RIIIa are called activating Fc ⁇ R since they have immunologically active functions
  • Fc ⁇ RIIb is called inhibitory Fc ⁇ R since it has immunosuppressive functions (Non-patent Document 8).
  • Non-patent Documents 4, 5, and 6 Various variants having Fc ⁇ R-binding properties, mainly antibodies with mutations introduced into these sites, have been studied so far; and Fc region variants having higher binding activities towards activating Fc ⁇ R have been obtained (Patent Documents 1, 2, 3, and 4).
  • Fc ⁇ R When activating Fc ⁇ R is cross-linked with an immune complex, it phosphorylates immunoreceptor tyrosine-based activating motifs (ITAMs) contained in the intracellular domain or FcR common ⁇ -chain (an interaction partner), activates a signal transducer SYK, and triggers inflammatory immune response by initiating an activation signal cascade (Non-patent Document 9).
  • ITAMs immunoreceptor tyrosine-based activating motifs
  • Fc ⁇ RIIb is the only Fc ⁇ R expressed on B cells (Non-patent Document 10). Interaction of the antibody Fc region with Fc ⁇ RIIb has been reported to suppress the primary immune response of B cells (Non-patent Document 11). Furthermore, it is reported that when Fc ⁇ RIIb on B cells and a B cell receptor (BCR) are cross-linked via an immune complex in blood, B cell activation is suppressed, and antibody production by B cells is suppressed (Non-patent Document 12).
  • BCR B cell receptor
  • ITIM immunoreceptor tyrosine-based inhibitory motif contained in the intracellular domain of Fc ⁇ RIIb is necessary (Non-patent Documents 13 and 14).
  • SH2-containing inositol polyphosphate 5-phosphatase (SHIP) is recruited, transduction of other activating Fc ⁇ R signal cascades is inhibited, and inflammatory immune response is suppressed (Non-patent Document 15).
  • Non-patent Document 16 aggregation of Fc ⁇ RIIb alone has been reported to transiently suppress calcium influx due to BCR cross-linking and B cell proliferation in a BCR-independent manner without inducing apoptosis of IgM-producing B cells.
  • Fc ⁇ RIIb is also expressed on dendritic cells, macrophages, activated neutrophils, mast cells, and basophils. Fc ⁇ RIIb inhibits the functions of activating Fc ⁇ R such as phagocytosis and release of inflammatory cytokines in these cells, and suppresses inflammatory immune responses (Non-patent Document 8).
  • Fc ⁇ RIIb regulatory inadequacy of Fc ⁇ RIIb has been reported to be related to human autoimmnue diseases.
  • the relationship between genetic polymorphism in the transmembrane region and promoter region of Fc ⁇ RIIb, and the frequency of development of systemic lupus erythematosus (SLE) (Non-patent Documents 20, 21, 22, 23, and 24), and decrease of Fc ⁇ RIIb expression on the surface of B cells in SLE patients (Non-patent Document 25 and 26) have been reported.
  • Fc ⁇ RIIb is considered to play the role of controlling autoimmune diseases and inflammatory diseases mainly through involvement with B cells, and it is a promising target molecule for controlling autoimmune diseases and inflammatory diseases.
  • IgG1 mainly used as a commercially available antibody pharmaceutical, is known to bind not only to Fc ⁇ RIIb, but also strongly to activating Fc ⁇ R (Non-patent Document 27). It may be possible to develop antibody pharmaceuticals having greater immunosuppressive properties compared with those of IgG1, by utilizing an Fc region with enhanced Fc ⁇ RIIb binding, or improved Fc ⁇ RIIb-binding selectivity compared with activating Fc ⁇ R. For example, it has been suggested that the use of an antibody having a variable region that binds to BCR and an Fc with enhanced Fc ⁇ RIIb binding may inhibit B cell activation (Non-patent Document 28).
  • an antibody having an Fc with improved Fc ⁇ RIIb-binding activity is suggested to be promising as a therapeutic agent for inflammatory diseases such as autoimmune diseases.
  • Non-patent Documents 32, 33, 34, 35, 36, and 37 show that the use of antibodies with enhanced Fc ⁇ RIIb binding enhances the anti-tumor effect of anti-CD40 antibodies. Accordingly, antibodies with enhanced Fc ⁇ RIIb are expected to have an effect of enhancing agonistic activity of agonist antibodies including antibodies against the anti-TNF receptor family.
  • Non-patent Document 28 Antibodies having an Fc with improved Fc ⁇ RIIb-binding activity have been reported (Non-patent Document 28).
  • Fc ⁇ RIIb-binding activity was improved by adding alterations such as S267E/L328F, G236D/S267E, and S239D/S267E to an antibody Fc region.
  • the antibody introduced with the S267E/L328F mutation most strongly binds to Fc ⁇ RIIb, and maintains the same level of binding to Fc ⁇ RIa and Fc ⁇ RIIa type H as that of a naturally-occurring IgG1.
  • Non-patent Document 8 Even if Fc ⁇ RIIb binding had been enhanced compared with that of IgG1, only the effect of enhancing Fc ⁇ RIIa binding and not the enhancement of Fc ⁇ RIIb binding is considered to have influence on cells such as platelets which express Fc ⁇ RIIa but do not express Fc ⁇ RIIb.
  • the group of patients who were administered bevacizumab, an antibody against VEGF is known to have an increased risk for thromboembolism (Non-patent Document 38). Furthermore, thromboembolism has been observed in a similar manner in clinical development tests of antibodies against the CD40 ligand, and the clinical study was discontinued (Non-patent Document 39).
  • Non-patent Documents 40 and 41 In systemic lupus erythematosus which is an autoimmune disease, platelets are activated via an Fc ⁇ RIIa-dependent mechanism, and platelet activation has been reported to correlate with the severity of symptoms (Non-patent Document 42). Even if Fc ⁇ RIIb binding is enhanced, administering an antibody with enhanced Fc ⁇ RIIa binding to such patients who already have a high risk for developing thromboembolism will increase the risk for developing thromboembolism, thus is extremely dangerous.
  • Non-patent Document 43 antibodies with enhanced Fc ⁇ RIIa binding have been reported to enhance macrophage-mediated antibody dependent cellular phagocytosis (ADCP) (Non-patent Document 43).
  • ADCP antibody dependent cellular phagocytosis
  • peptide fragments derived from those antibodies are also presented as an antigen and the antigenicity may become higher, thereby increasing the risk of production of antibodies against antibodies (anti-antibodies).
  • enhancing Fc ⁇ RIIa binding will increase the risk of production of antibodies against the antibodies, and this will remarkably decrease their value as pharmaceuticals.
  • the value as pharmaceuticals will be considerably reduced when Fc ⁇ RIIa binding is enhanced, which leads to increased risk of thrombus formation via platelet aggregation, higher antigenicity, and increased risk of anti-antibody production.
  • the aforementioned Fc with enhanced Fc ⁇ RIIb binding shows remarkably enhanced type-R Fc ⁇ RIIa binding compared with that of a naturally-occurring IgG1. Therefore, its value as a pharmaceutical for patients carrying type-R Fc ⁇ RIIa is considerably reduced.
  • Types H and R of Fc ⁇ RIIa are observed in Caucasians and African-Americans with approximately the same frequency (Non-patent Documents 44 and 45). Therefore, when this Fc was used for treatment of autoimmune diseases, the number of patients who can safely use it while enjoying its effects as a pharmaceutical will be limited.
  • Fc ⁇ RIIa is expressed on the dendritic cell surface in addition to Fc ⁇ RIIb; therefore, even if binding to inhibitory Fc ⁇ RIIb is enhanced and if binding to activating Fc ⁇ R such as Fc ⁇ RIIa is also enhanced, maturation of dendritic cells may be promoted as a result. More specifically, improving not only the Fc ⁇ RIIb-binding activity but also the ratio of Fc ⁇ RIIb-binding activity relative to Fc ⁇ RIIa-binding activity is considered to be important in providing antibodies with an immunosuppressive action.
  • Non-patent Document 48 cases where amino acid alterations were introduced into the Fc region to increase the Fc ⁇ RIIb-binding selectivity have been reported so far (Non-patent Document 48).
  • all variants said to have improved Fc ⁇ RIIb selectivity as reported in this document showed decreased Fc ⁇ RIIb binding compared with that of a naturally-occurring IgG1. Therefore, it is considered to be difficult for these variants to actually induce an Fc ⁇ RIIb-mediated immunosuppressive reaction more strongly than IgG1.
  • Fc ⁇ RIIb plays an important role in the agonist antibodies mentioned above, enhancing their binding activity is expected to enhance the agonistic activity.
  • Fc ⁇ RIIa binding is similarly enhanced, unintended activities such as ADCC activity and ADCP activity will be exhibited, and this may cause side effects.
  • Fc ⁇ RIIb shares 93% sequence identity in the extracellular region with that of Fc ⁇ RIIa which is one of the activating Fc ⁇ Rs, and they are very similar structurally.
  • Fc ⁇ RIIa H type and R type, in which the amino acid at position 131 is His (type H) or Arg (type R), and yet each of them reacts differently with the antibodies (Non-patent Document 49).
  • Patent Document 5 reports variants with enhanced Fc ⁇ RIIb-binding activity; however, the degree of enhancement is low, and there is a demand for development of variants having properties similar to those described above.
  • An objective of the present invention is to provide polypeptides comprising an IgG Fc region that have maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide through introduction of amino acid substitutions into the IgG Fc region; pharmaceutical compositions comprising the polypeptide; therapeutic agents or preventive agents comprising the polypeptide for immunological inflammatory diseases; and methods for producing them.
  • an objective is to provide a method for maintaining or decreasing binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and for enhancing Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide; and a method for suppressing antibody production in comparison with a parent polypeptide in in vivo administration.
  • an objective is to provide methods for producing a polypeptide having maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, H type and R type, in which the amino acid at position 131 (EU numbering) in Fc ⁇ RIIa is His (type H) or Arg (type R), and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide; and methods for producing a polypeptide with suppressed antibody production in comparison with a parent polypeptide when administered in vivo.
  • the present inventors performed dedicated research on a polypeptide comprising an Fc region having decreased Fc-mediated binding to Fc ⁇ RIIa, and increased binding to Fc ⁇ RIIb in comparison with a parent polypeptide.
  • a polypeptide comprising an antibody Fc region that comprises an alteration produced by substituting Pro at position 238 (EU numbering) with Asp or Leu at position 328 (EU numbering) with Glu enhances Fc ⁇ RIIb-binding activity, and decreases Fc region-mediated binding activity towards both allotypes of Fc ⁇ RIIa, types H and R.
  • polypeptide comprising an antibody Fc region that comprises an alteration of substituting Pro at position 238 (EU numbering) with Asp and several other alterations that enhance Fc ⁇ RIIb-binding activity, and maintains or decreases Fc region-mediated binding activities towards both allotypes of Fc ⁇ RIIa, types H and R.
  • the present invention relates to the following:
  • a polypeptide variant comprising an antibody Fc region with at least one amino acid alteration, which has maintained or decreased binding activities towards Fc ⁇ RIIa (type R) and Fc ⁇ RIIa (type H), and enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide, and wherein the value of [KD value of the polypeptide variant for Fc ⁇ RIIa (type R)]/[KD value of the polypeptide variant for Fc ⁇ RIIb] is 1.2 or more; [2] the polypeptide of [1], wherein the value of [KD value of the polypeptide variant for Fc ⁇ RIIa (type H)]/[KD value of the polypeptide variant for Fc ⁇ RIIb] is 4.2 or more; [3] the polypeptide of [1] or [2], wherein the value of [KD value of the parent polypeptide for Fc ⁇ RIIb]/[KD value of the polypeptide variant for Fc ⁇ RIIb] is 1.6 or more;
  • the present invention also relates to methods for treating or preventing immunological inflammatory diseases, which comprise the step of administering to a subject a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention. Furthermore, the present invention relates to kits for use in the therapeutic methods or preventive methods of the present invention, which comprise a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention, or a pharmaceutical composition of the present invention. The present invention also relates to use of a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention in the production of therapeutic agents or preventive agents for immunological inflammatory diseases.
  • the present invention relates to a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention for use in a therapeutic method or a preventive method of the present invention.
  • the present invention also relates to methods for suppressing activation of B cells, mast cells, dendritic cells, and/or basophils, which comprise the step of administering to a subject a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention.
  • the present invention relates to kits for use in the inhibition method of the present invention, which comprises a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention, or a pharmaceutical composition of the present invention.
  • the present invention relates to use of a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention in the production of agents that suppress activation of B cells, mast cells, dendritic cells, and/or basophils.
  • the present invention relates to polypeptides of the present invention or polypeptides produced by the production methods of the present invention for use in the inhibitory methods of the present invention.
  • the present invention relates to methods for treating diseases with deficiency of biologically essential proteins, which comprises the step of administering to a subject a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention.
  • the present invention relates to kits for use in the therapeutic method of the present invention, which comprises a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention, or a pharmaceutical composition of the present invention.
  • the present invention relates to use of a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention in the production of therapeutic agents for diseases with deficiency of biologically essential proteins.
  • the present invention also relates to a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention for use in a therapeutic method of the present invention.
  • the present invention relates to methods for inhibiting viruses, which comprises the step of administering to a subject a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention.
  • the present invention relates to kits for use in the inhibition method of the present invention, which comprises a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention, or a pharmaceutical composition of the present invention.
  • the present invention relates to use of a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention in the production of an antiviral agent.
  • the present invention relates to a polypeptide of the present invention or a polypeptide produced by the production methods of the present invention for use in the inhibition method of the present invention.
  • Polypeptides comprising an Fc region having maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, types R and H, and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide are provided by the present invention.
  • polypeptides with enhanced binding selectivity for Fc ⁇ RIIb than for both allotypes of Fc ⁇ RIIa types H and R
  • an antibody Fc with the property of selective Fc ⁇ RIIb binding, it may be possible to suppress anti-antibody production through the Fc ⁇ RIIb-mediated immunosuppressive action.
  • FIG. 1 shows comparison of Fc ⁇ RIa binding and Fc ⁇ RIIb binding. Binding of the antibody with substitution of Pro at position 238 (EU numbering) with Asp, and binding of the antibody with substitution of Leu at position 328 (EU numbering) with Glu have been labeled. “Mutation A” refers to an alteration produced by substituting Pro at position 238 (EU numbering) with Asp and “mutation B” refers to an alteration produced by substituting Leu at position 328 (EU numbering) with Glu.
  • FIG. 2 shows comparison of Fc ⁇ RIIa type H binding and Fc ⁇ RIIb binding. Binding of the antibody with substitution of Pro at position 238 (EU numbering) with Asp, and binding of the antibody with substitution of Leu at position 328 (EU numbering) with Glu have been labeled. “Mutation A” refers to an alteration produced by substituting Pro at position 238 (EU numbering) with Asp, and “mutation B” refers to an alteration produced by substituting Leu at position 328 (EU numbering) with Glu.
  • FIG. 3 shows comparison of Fc ⁇ RIIa type R binding and Fc ⁇ RIIb binding. Binding of the antibody with substitution of Pro at position 238 (EU numbering) with Asp, and binding of the antibody with substitution of Leu at position 328 (EU numbering) with Glu have been labeled. “Mutation A” refers to an alteration produced by substituting Pro at position 238 (EU numbering) with Asp, and “mutation B” refers to an alteration produced by substituting Leu at position 328 (EU numbering) with Glu.
  • FIG. 4 shows comparison of Fc ⁇ RIIIa binding and Fc ⁇ RIIb binding. Binding of the antibody with substitution of Pro at position 238 (EU numbering) with Asp, and binding of the antibody with substitution of Leu at position 328 (EU numbering) with Glu have been labeled. “Mutation A” refers to an alteration produced by substituting Pro at position 238 (EU numbering) with Asp, and “mutation B” refers to an alteration produced by substituting Leu at position 328 (EU numbering) with Glu.
  • FIG. 5 shows the relationship between the amino acid residues constituting the Fc regions of IgG1, IgG2, IgG3, and IgG4, and EU numbering (herein, also referred to as EU INDEX).
  • FIG. 6 shows a graph in which the horizontal axis shows the relative value of Fc ⁇ RIIb-binding activity of each PD variant, and the vertical axis shows the relative value of Fc ⁇ RIIa type R-binding activity of each PD variant.
  • the value for the amount of binding of each PD variant to each Fc ⁇ R was divided by the value for the amount of binding of IL6R-F652, which is a control antibody prior to introduction of the alteration (altered Fc with substitution of Pro at position 238 (EU numbering) with Asp), to each Fc ⁇ R; and then the obtained value was multiplied by 100, and used as the relative binding activity value for each PD variant to each Fc ⁇ R.
  • the F652 plot in the figure shows the value for IL6R-F652.
  • FIG. 7 shows a graph in which the vertical axis shows the relative value of Fc ⁇ RIIb-binding activity of variants produced by introducing each alteration into GpH7-B3 which does not have the P238D alteration, and the horizontal axis shows the relative value of Fc ⁇ RIIb-binding activity of variants produced by introducing each alteration into IL6R-F652 which has the P238D alteration.
  • the value for the amount of Fc ⁇ RIIb binding of each variant was divided by the value for the amount of Fc ⁇ RIIb binding of the pre-altered antibody; and then the obtained value was multiplied by 100, and used as the value of relative binding activity.
  • region A contains alterations that exhibit the effect of enhancing Fc ⁇ RIIb binding in both cases where an alteration is introduced into GpH7-B3 which does not have P238D and where an alteration is introduced into IL6R-F652 which has P238D.
  • Region B contains alterations that exhibit the effect of enhancing Fc ⁇ RIIb binding when introduced into GpH7-B3 which does not have P238D, but do not exhibit the effect of enhancing Fc ⁇ RIIb binding when introduced into IL6R-F652 which has P238D.
  • FIG. 8 shows a crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • FIG. 9 shows an image of superimposing the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex and the model structure of the Fc(WT)/Fc ⁇ RIIb extracellular region complex, with respect to the Fc ⁇ RIIb extracellular region and the Fc CH2 domain A by the least squares fitting based on the C ⁇ atom pair distances.
  • FIG. 10 shows comparison of the detailed structure around P238D after superimposing the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex and the model structure of the Fc(WT)/Fc ⁇ RIIb extracellular region complex with respect to the only Fc CH2 domain A or the only Fc CH2 domain B by the least squares fitting based on the C ⁇ atom pair distances.
  • FIG. 11 shows that a hydrogen bond can be found between the main chain of Gly at position 237 (EU numbering) in Fc CH2 domain A, and Tyr at position 160 in Fc ⁇ RIIb in the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • FIG. 12 shows that an electrostatic interaction can be found between Asp at position 270 (EU numbering) in Fc CH2 domain B, and Arg at position 131 in Fc ⁇ RIIb in the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • FIG. 13 shows a graph in which the horizontal axis shows the relative value of Fc ⁇ RIIb-binding activity of each 2B variant, and the vertical axis shows the relative value of Fc ⁇ RIIa type R-binding activity of each 2B variant.
  • the value for the amount of binding of each 2B variant to each Fc ⁇ R was divided by the value for the amount of binding of a control antibody prior to alteration (altered Fc with substitution of Pro at position 238 (EU numbering) with Asp) to each Fc ⁇ R; and then the obtained value was multiplied by 100, and used as the value of relative binding activity of each 2B variant towards each Fc ⁇ R.
  • FIG. 14 shows Glu at position 233 (EU numbering) in Fc Chain A and the surrounding residues in the extracellular region of Fc ⁇ RIIb in the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • FIG. 15 shows Ala at position 330 (EU numbering) in Fc Chain A and the surrounding residues in the extracellular region of Fc ⁇ RIIb in the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • FIG. 16 shows the structures of Pro at position 271 (EU numbering) of Fc Chain B after superimposing the crystal structures of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex and the Fc(WT)/Fc ⁇ RIIIa extracellular region complex by the least squares fitting based on the C ⁇ atom pair distances with respect to Fc Chain B.
  • the present invention provides polypeptides comprising an IgG Fc region that have maintained or decreased Fc ⁇ RIIa-binding, and having enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide by introducing amino acid substitution(s) into the IgG Fc region.
  • the present invention provides a polypeptide comprising an antibody Fc region that comprises a substitution of Pro at position 238 (EU numbering) with Asp or substitution of Leu at position 328 (EU numbering) with Glu, and a polypeptide comprising an antibody Fc region that comprises combination of a substitution of Pro at position 238 (EU numbering) with Asp and several specific amino acid substitutions.
  • the present invention provides a method for maintaining or decreasing binding activity towards both allotypes of Fc ⁇ RIIa and enhancing the Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide.
  • the present invention also provides a method for suppressing the antibody production in comparison with a parent polypeptide when the polypeptide is administered in vivo.
  • Polypeptides of the present invention generally refers to peptides or proteins approximately ten amino acids or more in length. Furthermore, they are generally polypeptides derived from organisms, but are not particularly limited, and for example, they may be polypeptides comprising an artificially designed sequence. Furthermore, they may be any of naturally-occurring polypeptides, synthetic polypeptides, recombinant polypeptides, or such.
  • Fc ⁇ receptors refers to receptors that may bind to the Fc region of IgG1, IgG2, IgG3, and IgG4 monoclonal antibodies, and practically means any member of the family of proteins encoded by the Fc ⁇ receptor genes.
  • this family includes Fc ⁇ RI (CD64) including isoforms Fc ⁇ RIa, Fc ⁇ RIb, and Fc ⁇ RIc; Fc ⁇ RII (CD32) including isoforms Fc ⁇ RIIa (including allotypes H131 (type H) and R131 (type R)), Fc ⁇ RIIb (including Fc ⁇ RIIb-1 and Fc ⁇ RIIb-2), and Fc ⁇ RIIc; and Fc ⁇ RIII (CD16) including isoforms Fc ⁇ RIIIa (including allotypes V158 and F158), and Fc ⁇ RIIIb (including allotypes Fc ⁇ RIIIb-NA1 and Fc ⁇ RIIIb-NA2), and any human Fc ⁇ Rs, Fc ⁇ R isoforms or allotypes yet to be discovered, but is not limited thereto.
  • the Fc ⁇ R includes human, mouse, rat, rabbit, and monkey-derived Fc ⁇ Rs but is not limited thereto, and may be derived from any organism.
  • Mouse Fc ⁇ Rs include Fc ⁇ RI (CD64), Fc ⁇ RII (CD32), Fc ⁇ RIII (CD16), and Fc ⁇ RIII-2 (CD16-2), and any mouse Fc ⁇ Rs, or Fc ⁇ R isoforms or allotypes yet to be discovered, but are not limited thereto.
  • Favorable examples of such Fc ⁇ receptors include human Fc ⁇ RI (CD64), Fc ⁇ RIIA (CD32), Fc ⁇ RIIB (CD32), Fc ⁇ RIIIA (CD16), and/or Fc ⁇ RIIIB (CD16).
  • polynucleotide sequence and amino acid sequence of Fc ⁇ RI are set forth in SEQ ID NOs: 1 (NM — 000566.3) and 2 (NP — 000557.1), respectively;
  • the polynucleotide sequence and amino acid sequence of Fc ⁇ RIIA are set forth in SEQ ID NOs: 3 (BCO20823.1) and 4 (AAH20823.1), respectively; the polynucleotide sequence and amino acid sequence of Fc ⁇ RIIB are set forth in SEQ ID NOs: 5 (BC146678.1) and 6 (AAI46679.1), respectively; the polynucleotide sequence and amino acid sequence of Fc ⁇ RIIIA are set forth in SEQ ID NOs: 7 (BCO33678.1) and 8 (AAH33678.1), respectively; and the polynucleotide sequence and amino acid sequence of Fc ⁇ RIIIB are set forth in SEQ ID NOs 9 (BC128562.1) and 10 (AAI28563.1), respectively (the RefSeq Registration number is indicated inside the parentheses).
  • Fc ⁇ RIIa there are two allotypes, one where the amino acid at position 131 of Fc ⁇ RIIa is histidine (type H) and the other where this amino acid is substituted with arginine (type R) (J. Exp. Med, 172: 19-25, 1990).
  • parent polypeptide refers to a polypeptide that will serve as the basis for the production of polypeptides comprising an antibody Fc region of the present invention. More specifically, it is a polypeptide comprising an antibody Fc region and is the polypeptide prior to alteration of at least one amino acid in the Fc region.
  • the parent polypeptide in the present invention may be, for example, a polypeptide comprising the Fc region of a naturally-occurring IgG, or it may be a polypeptide comprising an Fc region of an IgG to which an alteration other than the amino acid alterations of the present invention has been made to a naturally-occurring IgG.
  • Naturally-occurring IgGs refers to polypeptides belonging to a class of antibodies practically encoded by immunoglobulin gamma genes and comprising an amino acid sequence identical to those of IgGs found in nature.
  • a naturally-occurring human IgG means a naturally-occurring human IgG1, naturally-occurring human IgG2, naturally-occurring human IgG3, naturally-occurring human IgG4, or such.
  • Naturally-occurring IgGs also include mutants spontaneously produced from them.
  • the Fc region of a naturally-occurring IgG means an Fc region comprising an amino acid sequence identical to that of the Fc region derived from an IgG found in nature.
  • the Fc region of a naturally-occurring IgG is shown in FIG. 5 (SEQ ID NOs: 11-14), and for example, it refers to Fc regions derived from naturally-occurring human IgG1, Fc regions derived from naturally-occurring human IgG2, Fc regions derived from naturally-occurring human IgG3, and Fc regions derived from naturally-occurring human IgG4.
  • the Fc regions of naturally-occurring IgGs also include mutants spontaneously produced from them.
  • whether or not the binding activity towards each type of Fc ⁇ R is enhanced, or maintained or decreased in a polypeptide or an Fc region of the present invention can be determined, for example, by observing whether there is a decrease or an increase in the dissociation constant (KD) value obtained from the results of sensorgram analysis, where various Fc ⁇ Rs are subjected to interaction as an analyte with antibodies immobilized onto the sensor chips or captured onto the sensor chips using Protein A, Protein L, Protein A/G, Protein G, anti-lamda chain antibodies, anti-kappa chain antibodies, antigenic peptides, antigenic proteins, or such using BIACORE which is an interaction analyzer that utilizes the surface plasmon resonance (SPR) phenomena, as shown in the Examples.
  • SPR surface plasmon resonance
  • RU resonance unit
  • KD dissociation constant
  • the binding activity of an Fc region towards an Fc ⁇ receptor can be measured by the Amplified Luminescent Proximity Homogeneous Assay (ALPHA) screening, the BIACORE method which utilizes the surface plasmon resonance (SPR) phenomena, or such, in addition to ELISA or fluorescence activated cell sorting (FACS) (Proc. Natl. Acad. Sci. USA (2006) 103 (11): 4005-4010).
  • APHA Amplified Luminescent Proximity Homogeneous Assay
  • SPR surface plasmon resonance
  • FACS fluorescence activated cell sorting
  • ALPHA screening is performed by ALPHA technology which uses two beads, a donor and an acceptor, based on the following principles.
  • Luminescent signals are detected only when molecules bound to donor beads physically interact with molecules bound to the acceptor beads, and the two beads are in close proximity to each other.
  • Laser-excited photosensitizer in the donor beads converts ambient oxygen to excited-state singlet oxygen. Singlet oxygen is dispersed around the donor beads, and when it reaches the adjacent acceptor beads, chemiluminescent reaction is induced in the beads, and light is ultimately emitted.
  • the molecules bound to the donor beads do not interact with the molecules bound to the acceptor beads, the chemiluminescent reaction does not take place because singlet oxygen produced by the donor beads does not reach the acceptor beads.
  • a biotinylated polypeptide complex is bound to the donor beads, and Fc ⁇ receptor tagged with glutathione S transferase (GST) is linked to the acceptor beads.
  • GST glutathione S transferase
  • the polypeptide complex comprising a wild-type Fc region interacts with the Fc ⁇ receptor and produces 520-620 nm signals.
  • the polypeptide complex comprising an untagged mutant Fc region competes with the polypeptide complex comprising a wild-type Fc region for interaction with the Fc ⁇ receptor. Relative binding activities can be determined by quantifying the decrease in fluorescence observed as a result of the competition.
  • Biotinylation of polypeptide complexes such as antibodies using Sulfo-NHS-biotin and such is well known.
  • the method of expressing the Fc ⁇ receptor and GST in a cell carrying a fusion gene produced by fusing a polynucleotide encoding the Fc ⁇ receptor in frame with a polynucleotide encoding GST in an expressible vector, and performing purification using a glutathione column is appropriately adopted as a method for tagging an Fc ⁇ receptor with GST.
  • the obtained signals are preferably analyzed, for example, by fitting them to a one-site competition model which uses a non-linear regression analysis using software such as GRAPHPAD PRISM (GraphPad, San Diego).
  • One of the substances (the ligand) in observation of an interaction is immobilized onto a gold thin film on a sensor chip, and by shining light from the reverse side of the sensor chip so that total reflection takes place at the interface between the gold thin film and glass, a portion of reduced reflection intensity is formed in part of the reflected light (SPR signal).
  • the other one of the substances (the analyte) in observation of an interaction is made to flow on the sensor chip surface and the ligand binds to the analyte, the mass of the immobilized ligand molecule increases and the refractive index of the solvent on the sensor chip surface changes.
  • the Biacore system indicates the amount of shift mentioned above, or more specifically the time variable of mass by plotting the change in mass on the sensor chip surface on the ordinate as the measurement data (sensorgram).
  • the amount of analyte bound to the ligand trapped on the sensor chip surface is determined from the sensorgram.
  • Kinetic parameters such as association rate constants (ka) and dissociation rate constants (kd) are determined from the curves of the sensorgram, and the dissociation constants (KD) are determined from the ratio of these constants.
  • a method for measuring inhibition is preferably used. An example of the method for measuring inhibition is described in Proc. Natl. Acad. Sci. USA (2006) 103 (11): 4005-4010.
  • a polypeptide with decreased Fc ⁇ R-binding activity refers to a polypeptide that binds to Fc ⁇ R with a substantially lower binding activity than the parent polypeptide when assay is performed by keeping the amount of the parent polypeptide and the amount of the polypeptide comprising at least one amino acid alteration in the Fc region of the parent polypeptide (also called a polypeptide variant) practically the same.
  • the KD value ratio (KD value of a polypeptide variant/KD value of a parent polypeptide) is preferably 1.25 or more, 2 or more, or 3 or more, and more preferably, 5 or more, 10 or more, 100 or more, 1,000 or more, or 10,000 or more.
  • the KD value is preferably increased by 1 ⁇ M or more, and more preferably increased by 2 ⁇ M or more, 3 ⁇ M or more, 5 ⁇ M or more, 10 ⁇ M or more, 20 ⁇ M or more, 50 ⁇ M or more, and 100 ⁇ M or more.
  • the KD value is preferably 0.0001 ⁇ M or more, and more preferably 0.001 ⁇ M or more, 0.01 ⁇ M or more, 0.1 ⁇ M or more, 0.5 ⁇ M or more, 1 ⁇ M or more, 2 ⁇ M or more, 3 ⁇ M or more, 5 ⁇ M or more, 10 ⁇ M or more, 100 ⁇ M or more, or 1,000 ⁇ M or more.
  • a polypeptide with enhanced Fc ⁇ R-binding activity refers to a polypeptide that binds to Fc ⁇ R with a substantially higher binding activity than the parent polypeptide when assay is performed by keeping the amount of the parent polypeptide and the amount of the polypeptide variant practically the same.
  • the KD value ratio (KD value of a parent polypeptide/KD value of a polypeptide variant) is preferably 1.25 or more, 2 or more, or 3 or more, and more preferably, 5 or more, 10 or more, 100 or more, 1,000 or more, or 10,000 or more.
  • the KD value is preferably decreased by 0.001 ⁇ M or more, and more preferably decreased by 0.01 ⁇ M, 0.1 ⁇ M, 1 ⁇ M or more, 2 ⁇ M or more, 3 ⁇ M or more, 5 ⁇ M or more, 10 ⁇ M or more, 20 ⁇ M or more, 50 ⁇ M or more, and 100 ⁇ M or more.
  • the KD value is preferably 5 ⁇ M or less, and more preferably 3 ⁇ M or less, 1 ⁇ M or less, 0.5 ⁇ M or less, 0.1 ⁇ M or less, 0.01 ⁇ M or less, 0.001 ⁇ M or less, or 0.0001 ⁇ M or less.
  • a polypeptide with unchanged (maintained) Fc ⁇ R-binding activity refers to a polypeptide that binds to Fc ⁇ R with a binding activity practically unchanged from or equivalent to the parent polypeptide when assay is performed by keeping the amount of the parent polypeptide and the amount of the polypeptide comprising at least one amino acid alteration in the Fc region of the parent polypeptide (also called a polypeptide variant) practically the same.
  • Whether or not a polypeptide is a polypeptide having maintained or decreased Fc ⁇ RIIa-binding activity and having enhanced Fc ⁇ RIIb-binding activity can be determined using the KD value of this polypeptide for Fc ⁇ RIIa and the KD value of this polypeptide for Fc ⁇ RIIb determined according to the above-mentioned examples.
  • An example is the case where the KD value of the polypeptide of the present invention for Fc ⁇ RIIb is decreased compared with the KD value of the parent polypeptide for Fc ⁇ RIIb; and the KD value of the polypeptide of the present invention for Fc ⁇ RIIa (type R and type H) is increased or maintained compared with the KD value of the parent polypeptide for Fc ⁇ RIIa (type R and type H). Furthermore, it is possible to determine by appropriately combining the KD value of the polypeptide for Fc ⁇ RIa and the KD value of the polypeptide for Fc ⁇ RIIIa, which were determined according to the above-mentioned example.
  • an increased Fc ⁇ RIIb-binding activity means that, for example, in the KD values measured by the measurement method described above, the KD ratio of [KD value of the parent polypeptide]/[KD value of the polypeptide variant] is preferably 1.6 or more, 2 or more, or 3 or more, and more preferably 5 or more, 10 or more, 20 or more, 30 or more, and 50 or more.
  • Maintained or decreased binding activities towards Fc ⁇ RIIa (type R) and Fc ⁇ RIIa (type H) means that, for example, in the KD values measured by the measurement method described above, the KD ratio of [KD value for the stronger of the binding activities of a polypeptide variant towards Fc ⁇ RIIa (type R) and Fc ⁇ RIIa (type H)]/[KD value for the stronger of the binding activities of a parent polypeptide towards Fc ⁇ RIIa (type R) and Fc ⁇ RIIa (type H)] is preferably 0.7 or more, 1 or more, 2 or more, or 3 or more, and more preferably 5 or more, 10 or more, 20 or more, 30 or more, and 50 or more.
  • Polypeptides of the present invention preferably have maintained or decreased binding activities towards Fc ⁇ RIIa type R and Fc ⁇ RIIa type H. Furthermore, they preferably have maintained or decreased binding activities towards Fc ⁇ RIIa type R and Fc ⁇ RIIa type H, as well as a maintained or decreased Fc ⁇ RIIIa-binding activity. In addition, they preferably have a maintained or decreased binding activity towards Fc ⁇ RIa.
  • a maintained or decreased binding activity towards Fc ⁇ RIIIa or Fc ⁇ RIa means that, for example, in the KD values measured by the measurement method described above, the KD ratio of [KD value of the polypeptide variant]/[KD value of the parent polypeptide] is preferably 1 or more, 2 or more, or 3 or more, and more preferably 5 or more, 10 or more, 20 or more, 30 or more, and 50 or more.
  • a polypeptide of the present invention is a polypeptide with improved binding selectivity for Fc ⁇ RIIb rather than for Fc ⁇ RIIa
  • KD value for Fc ⁇ RIIa/KD value for Fc ⁇ RIIb the ratio of the KD value for Fc ⁇ RIIa to the KD value for Fc ⁇ RIIb of the parent peptide
  • the polypeptide of the present invention when the value of the KD ratio for the polypeptide of the present invention is greater than that of the parent polypeptide, the polypeptide of the present invention can be determined to have an improved binding selectivity for Fc ⁇ RIIb rather than for Fc ⁇ RIIa in comparison with the parent polypeptide.
  • the binding selectivity between Fc ⁇ RIIa (type R) and Fc ⁇ RIIb is, for example, a KD value ratio [KD value of the polypeptide variant for Fc ⁇ RIIa (type R)]/[KD value of the polypeptide variant for Fc ⁇ RIIb] of preferably 1.2 or more, 2 or more, or 3 or more for the KD values measured by the measurement method described above, and more preferably 5 or more, 10 or more, 20 or more, or 30 or more.
  • the binding selectivity between Fc ⁇ RIIa (type H) and Fc ⁇ RIIb is, for example, a KD value ratio [KD value of the polypeptide variant for Fc ⁇ RIIa (type H)]/[KD value of the polypeptide variant for Fc ⁇ RIIb] of preferably 4.2 or more, 5 or more, or 10 or more for the KD values measured by the measurement method described above, and more preferably 20 or more, 30 or more, 50 or more, 100 or more, or 200 or more.
  • the amount of binding of the various Fc ⁇ Rs to the polypeptides refers to values obtained by determining the difference in the RU values of sensorgrams that changed before and after interaction of various Fc ⁇ Rs as the analyte with each polypeptide, and dividing them by differences in the RU values of sensorgrams that changed before and after capturing polypeptides to the sensor chips.
  • polypeptides of the present invention is a polypeptide having maintained or decreased binding activities towards Fc ⁇ RIIa (type R and type H), and having increased binding activity towards Fc ⁇ RIIb can be determined by using the amount of Fc ⁇ RIIa binding of the polypeptide and the amount of Fc ⁇ RIIb binding of the polypeptide, which were determined according to the examples described above.
  • an example is the case where the amount of Fc ⁇ RIIb binding of a polypeptide of the present invention is increased compared with the amount of Fc ⁇ RIIb binding of a parent polypeptide, and the amount of Fc ⁇ RIIa (type R and type H) binding of a polypeptide of the present invention is equivalent to (maintained at) or preferably decreased from the amount of binding of a parent polypeptide towards Fc ⁇ RIIa (type R and type H). Furthermore, it is possible to determine by appropriately combining the amount of Fc ⁇ RIa binding and the amount of Fc ⁇ RIIIa binding of the polypeptide determined according to the examples described above.
  • Fc region refers to the region comprising a fragment consisting of a hinge portion or a part thereof, CH2 domain, or CH3 domain in an antibody molecule.
  • EU numbering herein, also called the EU INDEX
  • an IgG-class Fc region refers to, for example, the region from cysteine at position 226 to the C terminus, or from proline at position 230 to the C terminus, but is not limited thereto.
  • the Fc region may be obtained preferably by re-eluting the fraction adsorbed onto protein A column after partially digesting IgG1, IgG2, IgG3, IgG4 monoclonal antibodies or such using a protease such as pepsin.
  • the protease is not particularly limited as long as it can digest a full-length antibody so that Fab and F(ab′)2 will be produced in a restrictive manner by appropriately setting the enzyme reaction conditions such as pH, and examples include pepsin and papain.
  • the present invention provides an antibody constant region comprising an Fc region which comprises an alteration produced by substituting Pro at position 238 (EU numbering) with Asp or substituting Leu at position 328 (EU numbering) with Glu in human IgG (IgG1, IgG2, IgG3, and IgG4).
  • Polypeptides with maintained or decreased binding activities towards Fc ⁇ RIa, Fc ⁇ RIIIa, and both allotypes of Fc ⁇ RIIa, types R and H, as well as enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide can be provided by introducing alteration of substituting Pro at position 238 (EU numbering) with Asp or substituting Leu at position 328 (EU numbering) with Glu in human IgG.
  • At least one alteration can be further added to the human IgG Fc region comprising the alteration produced by substituting Pro at position 238 (EU numbering) with Asp or substituting Leu at position 328 (EU numbering) with Glu.
  • alteration refers to any one of, or a combination of substitutions, deletions, additions, and insertions. Additional alterations can be further included with these alterations.
  • the additional alteration can be selected from any one of, or combinations of amino acid substitutions, deletions, or modifications. For example, alterations that enhance the binding activity to Fc ⁇ RIIb, as well as maintain or decrease binding activities towards Fc ⁇ RIIa (type H) and Fc ⁇ RIIa (type R) can be added. Adding such alterations improves the binding selectivity for Fc ⁇ RIIb rather than for Fc ⁇ RIIa.
  • alterations that improve the binding selectivity for Fc ⁇ RIIb rather than for Fc ⁇ RIIa are preferred, and alterations that improve the binding selectivity for Fc ⁇ RIIb rather than for Fc ⁇ RIIa (type H) are more preferred.
  • Preferred examples of alterations of substituting an amino acid include,
  • the alteration mentioned above may be an alteration introduced at one position, and alternatively, or alterations at two or more positions can be combined.
  • Preferred examples of such alterations include those mentioned in Tables 6-7 and Tables 9-12.
  • amino acid substitutions that improve FcRn-binding activity J. Immunol. 2006 Jan. 1; 176(1): 346-56; J Biol. Chem. 2006 Aug. 18; 281(33): 23514-24; Int. Immunol. 2006 December; 18(12): 1759-69; Nat. Biotechnol. 2010 February; 28(2): 157-9.; WO 2006/019447; WO 2006/053301; and WO 2009/086320), and amino acid substitutions for improving antibody heterogeneity or stability (WO 2009/041613) may be introduced into an antibody constant region portion.
  • polypeptides produced by conferring polypeptides of the present invention with the property of promoting disappearance of antigens which are described in WO 2011/122011 or PCT/JP2011/072550
  • polypeptides conferring the property for repeated binding to a plurality of antigen molecules which are described in WO 2009/125825 or PCT/JP2011/077619, are also included in the present invention.
  • polypeptides of the present invention include IgG antibodies.
  • IgG antibodies When an IgG antibody is used as the antibody, the type of constant region is not limited, and an IgG isotypes (subclasses) such as IgG1, IgG2, IgG3, and IgG4 can be used.
  • IgG antibodies of the present invention are preferably human IgG, and more preferably human IgG1 and human IgG4.
  • the amino acid sequences of the heavy-chain constant regions of human IgG1 and human IgG4 are known. A plurality of allotype sequences due to genetic polymorphisms have been described in Sequences of Proteins of Immunological Interest, NIH Publication No. 91-3242 for the human IgG1 constant region, and any of the sequences may be used in the present invention.
  • polypeptide backbone structure in the sheet-structure or helical-structure region (a) polypeptide backbone structure in the sheet-structure or helical-structure region; (b) electric charge or hydrophobicity at the target site; or (c) size of the side chain.
  • Amino acid residues are classified into the following groups based on their general side chain properties:
  • hydrophobic norleucine, met, ala, val, leu, and ile
  • neutral hydrophilic cys, ser, thr, asn, and gln
  • acidic asp and glu
  • basic his, lys, and arg
  • residues that affect the chain orientation gly and pro
  • aromatic tip, tyr, and phe.
  • substitutions in the present invention may be conservative substitutions or non-conservative substitutions, or a combination of conservative substitutions and non-conservative substitutions.
  • Amino acid sequence alterations are produced by various methods known to those skilled in the art. Such methods include the site-directed mutagenesis method (Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis. Gene 152: 271-275; Zoller, M J, and Smith, M. (1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors. Methods Enzymol.
  • Amino acid modification of the present invention includes post-translational modification.
  • a specific post-translational modification may be addition or deletion of a sugar chain.
  • the amino acid residue at position 297 (EU numbering) may be sugar chain-modified.
  • the sugar-chain structure for the modification is not limited.
  • antibodies expressed in eukaryotic cells comprise glycosylation in the constant region. Therefore, antibodies expressed in cells such as those below are normally modified by some type of sugar chain:
  • Eukaryotic cells shown here include yeast and animal cells.
  • CHO cells and HEK293H cells are representative animal cells used in transformation with an expression vector comprising an antibody-encoding DNA.
  • those without glycosylation at this site are also included in the constant region of the present invention.
  • Antibodies whose constant region is not glycosylated can be obtained by expressing an antibody-encoding gene in prokaryotic cells such as Escherichia coli.
  • sialic acid may be added to the sugar chain of an Fc region (MAbs. 2010 September-October; 2(5): 519-27).
  • the present invention provides antibodies comprising an Fc region in which any of the above-mentioned amino acid sequences is altered.
  • antibody/antibodies in the present invention is used in the broadest sense, and as long as the desired biological activity is shown, it comprises any antibody such as monoclonal antibodies (including full-length monoclonal antibodies), polyclonal antibodies, antibody variants, antibody fragments, polyspecific antibodies (multi-specific antibodies) (for example, bispecific antibodies (diabodies)), chimeric antibodies, and humanized antibodies.
  • the antigen type and antibody origin are not limited, and they may be any type of antibodies.
  • the origin of the antibodies is not particularly limited, but examples include human antibodies, mouse antibodies, rat antibodies, and rabbit antibodies.
  • monoclonal antibodies may be produced by the hybridoma method (Kohler and Milstein, Nature 256: 495 (1975)), or the recombination method (U.S. Pat. No. 4,816,567). Alternatively, they may be isolated from a phage antibody library (Clackson et al., Nature 352: 624-628 (1991); Marks et al., J. Mol. Biol. 222: 581-597 (1991)).
  • a humanized antibody is also called a reshaped human antibody.
  • humanized antibodies prepared by grafting the CDRs of a non-human animal antibody such as a mouse antibody to a human antibody and such are known.
  • Common genetic engineering techniques for obtaining humanized antibodies are also known.
  • overlap extension PCR is known as a method for grafting mouse antibody CDRs to human FRs.
  • a vector for expressing a humanized antibody can be produced by inserting a DNA encoding an antibody variable region in which three CDRs and four FRs are ligated and a DNA encoding a human antibody constant region into an expression vector so that these DNAs are fused in frame. After this integration vector is transfected into a host to establish recombinant cells, these cells are cultured, and the DNA encoding the humanized antibody is expressed to produce the humanized antibody in the culture of the cells (see, European Patent Publication No. EP 239,400, and International Patent Publication No. WO 1996/002576).
  • an amino acid residue in an FR may be substituted so that the CDRs of a reshaped human antibody form an appropriate antigen-binding site.
  • a mutation can be introduced into the amino acid sequence of an FR by applying the PCR method used for grafting mouse CDRs to human FRs.
  • a desired human antibody can be obtained by DNA immunization using a transgenic animal having the complete repertoire of human antibody genes (see International Publication Nos. WO 1993/012227, WO 1992/003918, WO 1994/002602, WO 1994/025585, WO 1996/034096, and WO 1996/033735) as an animal for immunization.
  • a human antibody V region is expressed on the surface of a phage as a single-chain antibody (scFv) by the phage display method.
  • the scFv-expressing phage that binds to the antigen can be selected.
  • the DNA sequence that encodes the V region of the antigen-bound human antibody can be determined by analyzing the genes of the selected phage.
  • an expression vector can be prepared by fusing the V-region sequence in-frame with the sequence of a desired human antibody C region, and then inserting this into a suitable expression vector.
  • the expression vector is introduced into suitable expression cells such as those described above, and the human antibody can be obtained by expressing the human antibody-encoding gene.
  • suitable expression cells such as those described above, and the human antibody can be obtained by expressing the human antibody-encoding gene.
  • These methods are already known (see, International Publication Nos. WO 1992/001047, WO 1992/020791, WO 1993/006213, WO 1993/011236, WO 1993/019172, WO 1995/001438, and WO 1995/15388).
  • Variable regions constituting the antibodies of the present invention can be variable regions that recognize any antigen.
  • antigen there is no particular limitation on the antigen, and it may be any antigens.
  • antigens preferably include ligands (cytokines, chemokines, and such), receptors, cancer antigens, MHC antigens, differentiation antigens, immunoglobulins, and immune complexes partly containing immunoglobulins.
  • cytokines examples include interleukins 1 to 18, colony stimulating factors (G-CSF, M-CSF, GM-CSF, etc.), interferons (IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , etc.), growth factors (EGF, FGF, IGF, NGF, PDGF, TGF, HGF, etc.), tumor necrosis factors (TNF- ⁇ and TNF- ⁇ ), lymphotoxin, erythropoietin, leptin, SCF, TPO, MCAF, and BMP.
  • G-CSF colony stimulating factors
  • M-CSF M-CSF, GM-CSF, etc.
  • interferons IFN- ⁇ , IFN- ⁇ , etc.
  • growth factors EGF, FGF, IGF, NGF, PDGF, TGF, HGF, etc.
  • tumor necrosis factors TNF- ⁇ and TNF- ⁇
  • lymphotoxin erythropoietin
  • leptin SCF
  • chemokines examples include CC chemokines such as CCL1 to CCL28, CXC chemokines such as CXCL1 to CXCL17, C chemokines such as XCL1 and XCL2, and CX3C chemokines such as CX3CL1.
  • receptors include receptors belonging to receptor families such as the hematopoietic growth factor receptor family, cytokine receptor family, tyrosine kinase-type receptor family, serine/threonine kinase-type receptor family, TNF receptor family, G protein-coupled receptor family, GPI anchor-type receptor family, tyrosine phosphatase-type receptor family, adhesion factor family, and hormone receptor family.
  • receptors belonging to these receptor families and their characteristics have been described in many documents such as Cooke B A., King R J B., van der Molen H J. ed. New Comprehesive Biochemistry Vol. 18B “Hormones and their Actions Part II” pp.
  • Examples of specific receptors belonging to the above-mentioned receptor families preferably include human or mouse erythropoietin (EPO) receptors (Blood (1990) 76 (1): 31-35; and Cell (1989) 57 (2): 277-285), human or mouse granulocyte-colony stimulating factor (G-CSF) receptors (Proc. Natl. Acad. Sci. USA. (1990) 87 (22): 8702-8706, mG-CSFR; Cell (1990) 61 (2): 341-350), human or mouse thrombopoietin (TPO) receptors (Proc Natl Acad Sci USA. (1992) 89 (12): 5640-5644; EMBO J.
  • EPO erythropoietin
  • human or mouse interferon (IFN)- ⁇ and ⁇ receptors Cell (1990) 60 (2): 225-234; and Cell (1994) 77 (3): 391-400
  • human or mouse leptin receptors human or mouse growth hormone (GH) receptors, human or mouse interleukin (IL)-10 receptors, human or mouse insulin-like growth factor (IGF)-I receptors, human or mouse leukemia inhibitory factor (LIF) receptors, and human or mouse ciliary neurotrophic factor (CNTF) receptors.
  • GH growth hormone
  • IL interleukin
  • IGF insulin-like growth factor
  • LIF leukemia inhibitory factor
  • CNTF human or mouse ciliary neurotrophic factor
  • Cancer antigens are antigens that are expressed as cells become malignant, and they are also called tumor-specific antigens. Abnormal sugar chains that appear on cell surfaces or protein molecules when cells become cancerous are also cancer antigens, and they are also called sugar-chain cancer antigens.
  • cancer antigens preferably include GPC3 which is a receptor belonging to the GPI anchor-type receptor family mentioned above, and is also expressed in several cancers including liver cancer (Int J. Cancer. (2003) 103 (4): 455-65), as well as EpCAM which is expressed in several cancers including lung cancer (Proc Natl Acad Sci USA. (1989) 86 (1): 27-31), CA19-9, CA15-3, and sialyl SSEA-1 (SLX).
  • MHC antigens are roughly classified into MHC class I antigens and MHC class II antigens.
  • MHC class I antigens include HLA-A, -B, -C, -E, -F, -G, and -H
  • MHC class II antigens include HLA-DR, -DQ, and -DP.
  • Differentiation antigens may include CD1, CD2, CD4, CD5, CD6, CD7, CD8, CD10, CD11a, CD11b, CD11c, CD13, CD14, CD15s, CD16, CD18, CD19, CD20, CD21, CD23, CD25, CD28, CD29, CD30, CD32, CD33, CD34, CD35, CD38, CD40, CD41a, CD41b, CD42a, CD42b, CD43, CD44, CD45, CD45RO, CD48, CD49a, CD49b, CD49c, CD49d, CD49e, CD49f, CD51, CD54, CD55, CD56, CD57, CD58, CD61, CD62E, CD62L, CD62P, CD64, CD69, CD71, CD73, CD95, CD102, CD106, CD122, CD126, and CDw130.
  • Immunoglobulins include IgA, IgM, IgD, IgG, and IgE Immunocomplexes include a component of at least any of the immunoglobulins.
  • antigens include, for example, the molecules below: 17-IA, 4-1BB, 4Dc, 6-keto-PGF1a, 8-iso-PGF2a, 8-oxo-dG, A1 adenosine receptor, A33, ACE, ACE-2, activin, activin A, activin AB, activin B, activin C, activin RIA, activin RIA ALK-2, activin RIB ALK-4, activin RITA, activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAMS, ADAMS, ADAMTS, ADAMTS4, ADAMTS5, addressin, aFGF, ALCAM, ALK, ALK-1, ALK-7, alpha-1-antitrypsin, alpha-V/beta-1 antagonist, ANG, Ang, APAF-1, APE, APJ, APP, APRIL, AR, ARC, ART, artemin, anti-Id,
  • One or more amino acid residue alterations are allowed in the amino acid sequences constituting the variable regions as long as their antigen-binding activities are maintained.
  • altering a variable region amino acid sequence there is no particularly limitation on the site of alteration and number of amino acids altered.
  • amino acids present in CDR and/or FR can be altered appropriately.
  • the binding activity is preferably maintained without particular limitation; and for example, as compared to before alteration, the binding activity is 50% or more, preferably 80% or more, and more preferably 100% or more.
  • the binding activity may be increased by amino acid alterations.
  • the binding activity may be 2-, 5-, 10-times higher or such than that before alteration.
  • alteration of amino acid sequence may be at least one of amino acid residue substitution, addition, deletion, and modification.
  • the modification of the N-terminal glutamine of a variable region into pyroglutamic acid by pyroglutamylation is a modification well known to those skilled in the art.
  • the heavy-chain N terminus is glutamine
  • the antibodies of the present invention comprise the variable regions in which the glutamine is modified to pyroglutamic acid.
  • Antibody variable regions of the present invention may have any sequences, and they may be antibody variable regions of any origin, such as mouse antibodies, rat antibodies, rabbit antibodies, goat antibodies, camel antibodies, humanized antibodies produced by humanizing these non-human antibodies, and human antibodies.
  • “Humanized antibodies”, also referred to as “reshaped human antibodies”, are antibodies in which the complementarity determining regions (CDRs) of an antibody derived from a non-human mammal, for example, a mouse antibody, are transplanted into the CDRs of a human antibody.
  • CDRs complementarity determining regions
  • light-chain constant regions of the present invention may be light-chain constant regions with amino acid alterations such as substitutions, deletions, additions, and/or insertions.
  • heavy chain constant regions of an antibody of the present invention heavy chain constant regions of human IgG antibodies may be used and heavy chain constant regions of human IgG1 antibodies and those of human IgG4 antibodies are preferred.
  • polypeptides of the present invention may be made into Fc fusion protein molecules by linking to other proteins, physiologically active peptides, and such.
  • proteins and biologically active peptides include receptors, adhesion molecules, ligands, and enzymes, but are not limited thereto.
  • Fc fusion protein molecules of the present invention include proteins with Fc domain fused to a receptor protein that binds to a target, and such examples include TNFR-Fc fusion protein, IL1R-Fc fusion protein, VEGFR-Fc fusion protein, and CTLA4-Fc fusion protein (Nat. Med. 2003 January; 9(1): 47-52; BioDrugs. 2006; 20(3): 151-60).
  • a protein to be fused to a polypeptide of the present invention may be any molecule as long as it binds to a target molecule, and examples include scFv molecules (WO 2005/037989), single-domain antibody molecules (WO 2004/058821; WO 2003/002609), antibody-like molecules (Current Opinion in Biotechnology 2006, 17: 653-658; Current Opinion in Biotechnology 2007, 18: 1-10; Current Opinion in Structural Biology 1997, 7: 463-469; and Protein Science 2006, 15: 14-27) such as DARPins (WO 2002/020565), Affibody (WO 1995/001937), Avimer (WO 2004/044011; WO 2005/040229), and Adnectin (WO 2002/032925).
  • antibodies and Fc fusion protein molecules may be multispecific antibodies that bind to multiple types of target molecules or epitopes.
  • the antibodies of the present invention include antibody modification products.
  • antibody modification products include, for example, antibodies linked with various molecules such as polyethylene glycol (PEG) and cytotoxic substances.
  • PEG polyethylene glycol
  • Such antibody modification products can be obtained by chemically modifying antibodies of the present invention. Methods for modifying antibodies are already established in this field.
  • the antibodies of the present invention may also be bispecific antibodies.
  • Bispecific antibody refers to an antibody that has in a single molecule variable regions that recognize different epitopes.
  • the epitopes may be present in a single molecule or in different molecules.
  • polypeptides of the present invention can be prepared by the methods known to those skilled in the art.
  • the antibodies can be prepared by the methods described below, but the methods are not limited thereto.
  • a DNA encoding a heavy chain in which one or more amino acid residues in the Fc region are substituted with other amino acids of interest can be prepared, for example, by obtaining a DNA encoding the Fc region of a natural heavy chain, and introducing an appropriate substitution so that a codon encoding a particular amino acid in the Fc region encodes another amino acid of interest.
  • a DNA encoding a heavy chain in which one or more amino acid residues in the Fc region are substituted with other amino acids of interest can also be prepared by designing and then chemically synthesizing a DNA encoding a protein in which one or more amino acid residues in the Fc region of the natural heavy chain are substituted with other amino acids of interest.
  • the position and type of amino acid substitution are not particularly limited.
  • alteration is not limited to substitution, and alteration may be any of deletion, addition, or insertion, or combination thereof.
  • a DNA encoding a heavy chain in which one or more amino acid residues in the Fc region are substituted with other amino acids of interest can be prepared as a combination of partial DNAs.
  • Such combinations of partial DNAs include, for example, the combination of a DNA encoding a variable region and a DNA encoding a constant region, and the combination of a DNA encoding an Fab region and a DNA encoding an Fc region, but are not limited thereto.
  • a DNA encoding a light chain can similarly be prepared as a combination of partial DNAs.
  • a heavy chain expression vector is constructed by inserting a DNA encoding a heavy chain variable region into an expression vector along with a DNA encoding a heavy chain constant region.
  • a light chain expression vector is constructed by inserting a DNA encoding a light chain variable region into an expression vector along with a DNA encoding a light chain constant region.
  • these heavy and light chain genes may be inserted into a single vector.
  • the DNA When inserting a DNA encoding the antibody of interest into an expression vector, the DNA is inserted so that the antibody is expressed under the control of an expression-regulating region such as an enhancer or promoter. Next, host cells are transformed with this expression vector to express the antibody. In such cases, an appropriate combination of host and expression vector may be used.
  • vectors examples include M13 vectors, pUC vectors, pBR322, pBluescript, and pCR-Script.
  • pGEM-T pDIRECT, pT7, and such can be used.
  • Expression vectors are particularly useful when using vectors for producing the polypeptides of the present invention.
  • a host cell is E. coli such as JM109, DH5 ⁇ , HB101, and XL1-Blue
  • the expression vectors must carry a promoter that allows efficient expression in E. coli , for example, lacZ promoter (Ward et al., Nature (1989) 341: 544-546; FASEB J. (1992) ⁇ : 2422-2427; its entirety are incorporated herein by reference), araB promoter (Better et al., Science (1988) 240: 1041-1043; its entirety are incorporated herein by reference), T7 promoter, or such.
  • lacZ promoter Ward et al., Nature (1989) 341: 544-546; FASEB J. (1992) ⁇ : 2422-2427; its entirety are incorporated herein by reference
  • araB promoter Better et al., Science (1988) 240: 1041-1043; its entirety
  • Such vectors include pGEX-5 ⁇ 1 (Pharmacia), “QIAexpress system” (Qiagen), pEGFP, or pET (in this case, the host is preferably BL21 that expresses T7 RNA polymerase) in addition to the vectors described above.
  • the vectors may contain signal sequences for polypeptide secretion.
  • a signal sequence for polypeptide secretion a pelB signal sequence (Lei, S. P. et al J. Bacteriol. (1987) 169: 4379; its entirety are incorporated herein by reference) may be used when a polypeptide is secreted into the E. coli periplasm.
  • the vector can be introduced into host cells by lipofectin method, calcium phosphate method, and DEAE-Dextran method, for example.
  • the vectors for producing the polypeptides of the present invention include mammalian expression vectors (for example, pcDNA3 (Invitrogen), pEGF-BOS (Nucleic Acids. Res.
  • insect cell-derived expression vectors for example, the “Bac-to-BAC baculovirus expression system” (Gibco-BRL) and pBacPAK8), plant-derived expression vectors (for example, pMH1 and pMH2), animal virus-derived expression vectors (for example, pHSV, pMV, and pAdexLcw), retroviral expression vectors (for example, pZIPneo), yeast expression vectors (for example, “ Pichia Expression Kit” (Invitrogen), pNV11, and SP-Q01), and Bacillus subtilis expression vectors (for example, pPL608 and pKTH50), for example.
  • Bacillus subtilis expression vectors for example, pPL608 and pKTH50
  • the vectors When aiming for expression in animal cells such as CHO, COS, and NIH3T3 cells, the vectors must have a promoter essential for expression in cells, for example, SV40 promoter (Mulligan et al., Nature (1979) 277: 108; its entirety are incorporated herein by reference), MMTV-LTR promoter, EF1 ⁇ promoter (Mizushima et al., Nucleic Acids Res. (1990) 18: 5322; its entirety are incorporated herein by reference), CAG promoter (Gene.
  • SV40 promoter Mulligan et al., Nature (1979) 277: 108; its entirety are incorporated herein by reference
  • MMTV-LTR promoter EF1 ⁇ promoter
  • CAG promoter Gene.
  • CMV promoter and CMV promoter, and more preferably they have a gene for selecting transformed cells (for example, a drug resistance gene that allows evaluation using an agent (neomycin, G418, or such)).
  • Vectors with such characteristics include pMAM, pDR2, pBK-RSV, pBK-CMV, pOPRSV, and pOP13, for example.
  • the following method can be used for stable gene expression and gene copy number amplification in cells: CHO cells deficient in a nucleic acid synthesis pathway are introduced with a vector that carries a DHFR gene which compensates for the deficiency (for example, pCHOI), and the vector is amplified using methotrexate (MTX).
  • MTX methotrexate
  • the following method can be used for transient gene expression: COS cells with a gene expressing SV40 T antigen on their chromosome are transformed with a vector with an SV40 replication origin (pcD and such). Replication origins derived from polyoma virus, adenovirus, bovine papilloma virus (BPV), and such can also be used.
  • the expression vectors may further carry selection markers such as aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, E. coli xanthine-guanine phosphoribosyltransferase (Ecogpt) gene, and dihydrofolate reductase (dhfr) gene.
  • APH aminoglycoside transferase
  • TK thymidine kinase
  • Ecogpt E. coli xanthine-guanine phosphoribosyltransferase
  • dhfr dihydrofolate reductase
  • Antibodies can be collected, for example, by culturing transformed cells, and then separating the antibodies from the inside of the transformed cells or from the culture media. Antibodies can be separated and purified using an appropriate combination of methods such as centrifugation, ammonium sulfate fractionation, salting out, ultrafiltration, 1q, FcRn, protein A, protein G column, affinity chromatography, ion exchange chromatography, and gel filtration chromatography.
  • the present invention provides methods for producing a polypeptide comprising an antibody Fc region having maintained or decreased Fc ⁇ RIIa-binding activity, and enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide, which comprises adding at least one amino acid alteration to the Fc region of the polypeptide.
  • Examples include production methods comprising the following steps:
  • step (a) adding at least one amino acid alteration to the Fc region of polypeptides comprising an antibody Fc region; (b) measuring the Fc ⁇ RIIa-binding activity and Fc ⁇ RIIb-binding activity of the polypeptides altered in step (a); and (c) selecting polypeptides having maintained or decreased Fc ⁇ RIIa-binding activity, and enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide.
  • a preferred embodiment is a method for producing a polypeptide comprising an antibody Fc region, which comprises the steps of:
  • antibodies and Fc fusion protein molecules produced by this production method are also included in the present invention.
  • the present invention also provides methods for producing a polypeptide in which antibody production against the polypeptide is suppressed compared with its parent polypeptide when administered in vivo, which comprise adding at least one amino acid alteration in the Fc region of a polypeptide comprising an antibody Fc region.
  • Examples include a production method comprising the following steps:
  • step (a) adding at least one amino acid alteration in the Fc region of a polypeptide comprising an antibody Fc region; and (b) confirming that antibody production is suppressed when the polypeptide altered in step (a) is administered in vivo in comparison with a parent polypeptide.
  • Whether or not production of antibodies against the polypeptide has been suppressed can be confirmed by methods of administering the polypeptide to an animal and such.
  • suppression of antibody production can be determined by measuring the binding activities towards Fc ⁇ RIIa and Fc ⁇ RIIb, and observing an increase in the value obtained by dividing the KD value for Fc ⁇ RIIa by the KD value for Fc ⁇ RIIb.
  • Such polypeptides are considered to be useful as pharmaceuticals since they can suppress antibody production without activating Fc ⁇ R.
  • a polypeptide comprising a human IgG Fc region is altered so that Pro at position 238 (EU numbering) is substituted with Asp or Leu at position 328 (EU numbering) is substituted with Glu.
  • Other preferred embodiments include altering the polypeptide so that at least one substitution selected from the group consisting of:
  • the present invention provides methods for altering a polypeptide for the production of a polypeptide having maintained or decreased Fc ⁇ RIIa-binding activity, and having enhanced Fc ⁇ RIIb-binding activity in comparison with its parent polypeptide.
  • the present invention also provides methods for altering a polypeptide for the production of a polypeptide whose antibody production is suppressed compared with that of a parent polypeptide when it is administered in vivo.
  • a polypeptide comprising a human IgG Fc region is altered so that Pro at position 238 (EU numbering) is substituted with Asp or Leu at position 328 (EU numbering) is substituted with Glu.
  • Other preferred embodiments include altering the polypeptide so that at least one substitution selected from the group consisting of:
  • the present invention provides a nucleic acid encoding a polypeptide comprising an antibody Fc region with at least one amino acid alteration, which has maintained or decreased Fc ⁇ RIIa-binding activity, and enhanced Fc ⁇ RIIb-binding activity in comparison with a parent polypeptide.
  • the nucleic acid of the present invention may be in any form such as DNA or RNA.
  • the present invention also provides vectors carrying the above-described nucleic acids of the present invention.
  • the type of vector can be appropriately selected by those skilled in the art depending on the host cells to be introduced with the vector.
  • the vectors include, for example, those described above.
  • the present invention relates to host cells transformed with the above-described vectors of the present invention.
  • Appropriate host cells can be selected by those skilled in the art.
  • the host cells include, for example, those described above.
  • the present invention provides methods for maintaining or decreasing Fc ⁇ RIIa-binding activity and enhancing Fc ⁇ RIIb-binding activity of a polypeptide comprising an antibody Fc region in comparison with a parent polypeptide, wherein the method comprises adding at least one amino acid alteration to the Fc region.
  • the present invention also provides methods for suppressing production of antibodies against a polypeptide compared with a parent polypeptide when the polypeptide is administered in vivo, wherein the method comprises adding at least one amino acid alteration in the Fc region of the polypeptide comprising an antibody Fc region.
  • a polypeptide comprising a human IgG Fc region is altered so that Pro at position 238 (EU numbering) is substituted with Asp or Leu at position 328 (EU numbering) is substituted with Glu.
  • Other preferred embodiments include altering the polypeptide so that at least one substitution selected from the group consisting of:
  • Fc ⁇ RIIb-binding activity it is preferable to enhance the Fc ⁇ RIIb-binding activity, and maintain or decrease binding activities towards Fc ⁇ RIIa (type R) and Fc ⁇ RIIa (type H); and it is preferable to additionally maintain or decrease binding activities towards Fc ⁇ RIa and/or Fc ⁇ RIIIa.
  • Polypeptides produced by any of the above-mentioned methods are also included in the present invention.
  • the present invention provides pharmaceutical compositions comprising the polypeptide of the present invention.
  • compositions of the present invention can be formulated, in addition to the antibody or Fc-fusion protein molecules of the present invention described above, with pharmaceutically acceptable carriers by known methods.
  • the compositions can be used parenterally, when the antibodies are formulated in a sterile solution or suspension for injection using water or any other pharmaceutically acceptable liquid.
  • the compositions can be formulated by appropriately combining the antibodies or Fc-fusion protein molecules with pharmaceutically acceptable carriers or media, specifically, sterile water or physiological saline, vegetable oils, emulsifiers, suspending agents, surfactants, stabilizers, flavoring agents, excipients, vehicles, preservatives, binding agents, and such, by mixing them at a unit dose and form required by generally accepted pharmaceutical implementations.
  • the carriers include light anhydrous silicic acid, lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyvinylacetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium-chain triglyceride, polyoxyethylene hardened castor oil 60, saccharose, carboxymethyl cellulose, corn starch, inorganic salt, and such.
  • the content of the active ingredient in such a formulation is adjusted so that an appropriate dose within the required range can be obtained.
  • Sterile compositions for injection can be formulated using vehicles such as distilled water for injection, according to standard protocols.
  • Aqueous solutions used for injection include, for example, physiological saline and isotonic solutions containing glucose or other adjuvants such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride. These can be used in conjunction with suitable solubilizers such as alcohol, specifically ethanol, polyalcohols such as propylene glycol and polyethylene glycol, and non-ionic surfactants such as Polysorbate 80TM and HCO-50.
  • suitable solubilizers such as alcohol, specifically ethanol, polyalcohols such as propylene glycol and polyethylene glycol, and non-ionic surfactants such as Polysorbate 80TM and HCO-50.
  • Oils include sesame oils and soybean oils, and can be combined with solubilizers such as benzyl benzoate or benzyl alcohol. These may also be formulated with buffers, for example, phosphate buffers or sodium acetate buffers; analgesics, for example, procaine hydrochloride; stabilizers, for example, benzyl alcohol or phenol; or antioxidants.
  • buffers for example, phosphate buffers or sodium acetate buffers
  • analgesics for example, procaine hydrochloride
  • stabilizers for example, benzyl alcohol or phenol
  • antioxidants antioxidants.
  • the administration is preferably carried out parenterally, and specifically includes injection, intranasal administration, intrapulmonary administration, and percutaneous administration.
  • injections can be administered systemically or locally by intravenous injection, intramuscular injection, intraperitoneal injection, or subcutaneous injection.
  • the method of administration can be appropriately selected according to the age and symptoms of the patient.
  • a single dosage of the pharmaceutical composition containing an antibody or a polynucleotide encoding an antibody can be selected, for example, from the range of 0.0001 to 1,000 mg per kg of body weight.
  • the dosage may be, for example, in the range of 0.001 to 100,000 mg/patient.
  • the dosage is not limited to these values.
  • the dosage and method of administration vary depending on the patient's body weight, age, and symptoms, and can be appropriately selected by those skilled in the art.
  • polypeptides of the present invention are useful as active ingredients of pharmaceutical agents that suppress the activation of B cells, mast cells, dendritic cells, and/or basophils.
  • Polypeptides of the present invention can suppress the activation of B cells, mast cells, dendritic cells, and/or basophils, by selectively working on Fc ⁇ RIIb without activating Fc ⁇ R.
  • B cell activation includes proliferation, IgE production, IgM production, and IgA production.
  • the above-mentioned polypeptides of the present invention cross-link Fc ⁇ RIIb with IgE to suppress IgE production of B cells, with IgM to suppress IgM production of B cells, and with IgA to suppress IgA production.
  • suppressive effects similar to those mentioned above are exhibited by directly or indirectly cross-linking Fc ⁇ RIIb with molecules that are expressed on B cells and comprise the ITAM domain inside the cell or interact with the ITAM domain such as BCR, CD19, and CD79b.
  • activation of mast cells includes proliferation, activation by IgE and such, and degranulation.
  • the above-mentioned polypeptides of the present invention can suppress proliferation, activation by IgE and such, and degranulation by directly or indirectly cross-linking Fc ⁇ RIIb with IgE receptor molecules that are expressed on mast cells and comprise the ITAM domain or interact with the ITAM domain such as Fc ⁇ RI, DAP12, and CD200R3.
  • Activation of basophils includes proliferation and degranulation of basophils.
  • the above-mentioned polypeptides of the present invention can suppress proliferation, activation, and degranulation by directly or indirectly cross-linking Fc ⁇ RIIb with molecules on the cell membrane, which comprise the ITAM domain inside the cell or interact with the ITAM domain.
  • Activation of dendritic cells includes proliferation and degranulation of dendritic cells.
  • the above-mentioned polypeptides of the present invention can suppress activation, degranulation, and proliferation by directly or indirectly cross-linking Fc ⁇ RIIb with molecules on the cell membrane, which comprise the ITAM domain inside the cell or interact with the ITAM domain.
  • polypeptides of the present invention are useful as an active ingredient of therapeutic agents or preventive agents for immunological inflammatory diseases.
  • polypeptides of the present invention can suppress activation of B cells, mast cells, dendritic cells and/or basophils, administration of the polypeptides of the present invention as a result can treat or prevent immunological inflammatory diseases.
  • the term “immunological inflammatory diseases” comprises, rheumatoid arthritis, autoimmune hepatitis, autoimmune thyroiditis, autoimmune blistering diseases, autoimmune adrenocortical disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, megalocytic anemia, autoimmune atrophic gastritis, autoimmune neutropenia, autoimmune orchitis, autoimmune encephalomyelitis, autoimmune receptor disease, autoimmune infertility, chronic active hepatitis, glomerulonephritis, interstitial pulmonary fibrosis, multiple sclerosis, Paget's disease, osteoporosis, multiple myeloma, uveitis, acute and chronic spondylitis, gouty arthritis, inflammatory bowel disease, adult respiratory distress syndrome (ARDS), psoriasis, Crohn's disease, Basedow's disease, juvenile diabetes, Addison's disease, myasth
  • the polypeptides of the present invention are useful as an active ingredient of pharmaceutical agents for treating or preventing the autoimmune diseases by suppressing production of those autoantibodies.
  • Use of a molecule produced by fusing an antibody Fc portion with AchR has been reported to suppress proliferation of B cells which express AchR-recognizing BCR, and induce apoptosis (J. Neuroimmunol, 227: 35-43, 2010).
  • a fusion protein formed between an antigen recognized by an autoantibody and an antibody Fc region of the present invention enables crosslinking of Fc ⁇ RIIb with BCR of a B cell expressing BCR for that autoantigen, suppression of proliferation of B cells expressing BCR for the autoantigen, and induction of apoptosis.
  • Such autoimmune diseases include Guillain-Barre syndrome, myasthenia gravis, chronic atrophic gastritis, autoimmune hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, autoimmune pancreatitis, aortitis syndrome, Goodpasture's syndrome, rapidly progressive glomerulonephritis, megaloblastic anemia, autoimmune hemolytic anemia, autoimmune neutropenia, idiopathic thrombocytopenic purpura, Basedow's disease, Hashimoto's thyroiditis, primary hypothyroidism, idiopathic Addison's disease, insulin-dependent diabetes mellitus, chronic discoid lupus erythematosus, localized scleroderma, pemphigus, pemphigoid, herpes gestationis, linear IgA bullous dermatosis, epidermolysis bullosa acquisita, alopecia greata,
  • polypeptides of the present invention are useful as an active ingredient in therapeutic agents for diseases with deficiency of a biologically essential protein.
  • therapeutic methods that administer and supplement the protein as a pharmaceutical agent are used.
  • the patient lacks the protein from the beginning, the externally supplemented protein is recognized as a foreign substance and antibodies against that protein are produced.
  • the protein becomes easily removed, and the effect as a pharmaceutical is reduced.
  • Use of a fusion protein comprising such a protein and an antibody Fc region of the present invention enables crosslinking between Fc ⁇ RIIb and BCR on B cells that recognize the protein, and enables suppression of antibody production against the protein.
  • the proteins to be supplemented include Factor VIII, Factor IX, TPO, EPO, ⁇ -iduronidase, iduronate sulfatase, A-type heparan N-sulfatase, B type ⁇ -N-acetylglucosaminidase, C type acetyl CoA: ⁇ -glucosaminidase acetyltransferase, D type N-acetylglucosamine 6-sulfatase, galactose 6-sulfatase, N-acetylgalactosamine 4-sulfatase, ⁇ -glucuronidase, ⁇ -galactosidase, acidic ⁇ -galactosidase, and glucocerebrosidase.
  • proteins may be supplemented for diseases such as hemophilia, idiopathic thrombocytopenic purpura, renal anemia, and lysosomal disease (mucopolysaccharidosis, Fabry's disease, Pompe disease, and Gaucher's disease), without being limited thereto.
  • diseases such as hemophilia, idiopathic thrombocytopenic purpura, renal anemia, and lysosomal disease (mucopolysaccharidosis, Fabry's disease, Pompe disease, and Gaucher's disease), without being limited thereto.
  • Antibodies that comprise an Fc region of the present invention and are anti-virus antibodies can suppress antibody-dependent enhancement observed with anti-virus antibodies.
  • Antibody-dependent enhancement is a phenomenon where a virus uses neutralizing antibodies against the virus to become phagocytosed via activating Fc ⁇ Rs, and infects Fc ⁇ R-expressing cells so that the infection spreads. Binding of anti-dengue-virus neutralizing antibodies to Fc ⁇ RIIb has been reported to play an important role in suppressing antibody-dependent enhancement (Proc. Natl. Acad. Sci. USA, 108: 12479-12484, 2011).
  • Crosslinking Fc ⁇ RIIb with an immunocomplex with dengue virus which is formed by the anti-dengue-virus neutralizing antibodies, inhibits Fc ⁇ R-mediated phagocytosis, resulting in the suppression of antibody-dependent enhancement.
  • dengue virus DENV1, DENV2, and DENV4
  • HIV but are not limited thereto.
  • polypeptides of the present invention described above are useful as an active ingredient in preventive agents or therapeutic agents for arteriosclerosis.
  • Antibodies against oxidized LDL i.e., a cause for arteriosclerosis, which are antibodies comprising an Fc region of the present invention, can prevent Fc ⁇ RIIa-dependent adhesion of inflammatory cells. It has been reported that while anti-oxidized LDL antibodies inhibit the interaction between oxidized LDL and CD36, anti-oxidized LDL antibodies bind to endothelial cells, and monocytes recognize their Fc portion in an Fc ⁇ RIIa-dependent or Fc ⁇ RI-dependent manner; and this leads to adhesion (Immunol. Lett., 108: 52-61, 2007). Using antibodies comprising an Fc region of the present invention for such antibodies may inhibit Fc ⁇ RIIa-dependent binding and suppress monocyte adhesion by Fc ⁇ RIIb-mediated inhibitory signals.
  • polypeptides of the present invention described above are useful as an active ingredient in therapeutic agents or preventive agents for cancer.
  • enhancing the Fc ⁇ RIIb binding enhances the agonistic activity of an agonist antibody, and enhances the antitumor effect of the antibody. Therefore, agonist antibodies using the Fc region of the present invention are useful for treatment or prevention of cancer.
  • the Fc region of the present invention enhances the agonistic activity of agonist antibodies against receptors of the TNF receptor family such as Aliases, CD120a, CD120b, Lymphotoxin 13 receptor, CD134, CD40, FAS, TNFRSF6B, CD27, CD30, CD137, TNFRSF10A, TNFRSF10B, TNFRSF10C, TNFRSF10D, RANK, Osteoprotegerin, TNFRSF12A, TNFRSF13B, TNFRSF13C, TNFRSF14, Nerve growth factor receptor, TNFRSF17, TNFRSF18, TNFRSF19, TNFRSF21, TNFRSF25, and Ectodysplasin A2 receptor.
  • TNF receptor family such as Aliases, CD120a, CD120b, Lymphotoxin 13 receptor, CD134, CD40, FAS, TNFRSF6B, CD27, CD30, CD137, TNFRSF10A, TNFRSF
  • cancer includes lung cancer (including small cell lung cancer, non-small cell lung cancer, pulmonary adenocarcinoma, and squamous cell carcinoma of the lung), large intestine cancer, rectal cancer, colon cancer, breast cancer, liver cancer, gastric cancer, pancreatic cancer, renal cancer, prostate cancer, ovarian cancer, thyroid cancer, cholangiocarcinoma, peritoneal cancer, mesothelioma, squamous cell carcinoma, cervical cancer, endometrial cancer, bladder cancer, esophageal cancer, head and neck cancer, nasopharyngeal cancer, salivary gland tumor, thymoma, skin cancer, basal cell tumor, malignant melanoma, anal cancer, penile cancer, testicular cancer, Wilms' tumor, acute myeloid leukemia (including acute myeloleukemia, acute myeloblastic leukemia, acute prom
  • the present invention relates to methods for treating or preventing immunological inflammatory diseases, which comprise the step of administering to a subject (patient) a polypeptide of the present invention or a polypeptide produced by production methods of the present invention.
  • kits for use in the therapeutic methods or preventive methods of the present invention which comprises at least a polypeptide of the present invention or a polypeptide produced by production methods of the present invention, or a pharmaceutical composition of the present invention.
  • pharmaceutically acceptable carriers, media, instructions on the method of use, and such may be included in the kit.
  • the present invention relates to use of a polypeptide of the present invention or a polypeptide produced by production methods of the present invention in the production of agents for treating or preventing immunological inflammatory diseases.
  • the present invention also relates to polypeptides of the present invention or polypeptides produced by production methods of the present invention for use in the therapeutic methods or preventive methods of the present invention.
  • Glycine Gly (G)
  • Lysine Lys (K)
  • Threonine Thr (T)
  • variable region (SEQ ID NO: 15) of a glypican 3 antibody comprising the CDR of GpH7 which is an anti-glypican 3 antibody with improved plasma kinetics disclosed in WO 2009/041062 was used as the common antibody H chain.
  • GpL16-k0 (SEQ ID NO: 16) of the glypican 3 antibody with improved plasma kinetics disclosed in WO 2009/041062 was used for the common antibody L chain.
  • B3 (SEQ ID NO: 17) in which a K439E mutation has been introduced into G1d produced by removing the C terminal Gly and Lys of IgG1 was used as the antibody H chain constant region.
  • This H chain is referred to as GpH7-B3 (SEQ ID NO: 18), and the L chain is referred to as GpL16-k0 (SEQ ID NO: 16).
  • B3 variants were expressed and purified using the method of Reference Example 1, and the binding to each Fc ⁇ R (Fc ⁇ RIa, Fc ⁇ RIIa type H, Fc ⁇ RIIa type R, Fc ⁇ RIIb, and Fc ⁇ RIIIa) was comprehensively evaluated using the method of Reference Example 2.
  • the horizontal axis shows the value of the relative Fc ⁇ RIIb-binding activity of each variant
  • the vertical axis shows the value of the respective relative binding activity of each variant towards activating Fc ⁇ Rs: Fc ⁇ RIa, Fc ⁇ RIIa type H, Fc ⁇ RIIa type R, and Fc ⁇ RIIIa ( FIGS. 1 , 2 , 3 , and 4 ).
  • Pro at position 238 (EU numbering) in IL6R-G1d was substituted with Asp to produce IL6R-G1d-v1 (SEQ ID NO: 21).
  • Leu at position 328 (EU numbering) in IL6R-G1d was substituted with Glu to produce IL6R-G1d-v2 (SEQ ID NO: 23).
  • Ser at position 267 (EU numbering) was substituted with Glu
  • Leu at position 328 (EU numbering) was substituted with Phe in IL6R-G1d to produce IL6R-G1d-v3 (SEQ ID NO: 24) as described in Non-patent Document 27.
  • IL6R-L (SEQ ID NO: 22), which is the L chain of tocilizumab, was utilized as a mutual antibody L chain; and together with each H chain, the antibodies were expressed and purified according to the method of Reference Example 1.
  • the obtained antibodies which comprise an amino acid sequence derived from IL6R-G1d, IL6R-G1d-v1, IL6R-G1d-v2, or IL6R-G1d-v3 as the antibody H chain are referred to as IgG1, IgG1-v1, IgG1-v2, and IgG1-v3, respectively.
  • the sensorgrams obtained as measurement results were analyzed by the 1:1 Langmuir binding model using the Biacore Evaluation Software to calculate the binding rate constant ka (L/mol/s) and dissociation rate constant kd (1/s), and the dissociation constant KD (mol/L) was calculated from these values.
  • Equation 1 The behavior of interacting molecules according to the 1:1 binding model on Biacore can be described by Equation 1 shown below.
  • KD can be expressed as Equation 2 shown below.
  • the amount of binding (R eq ) of IgG1-v1 and IgG1-v2 to Fc ⁇ RIIa type H was approximately 2.5 RU and 10 RU, respectively, and the amount of binding (R eq ) of IgG1-v1 and IgG1-v2 to Fc ⁇ RIIIa was approximately 2.5 RU and 5 RU, respectively.
  • the amount of IgG1, IgG1-v1, and IgG1-v2 captured in the analysis of interactions with H-type Fc ⁇ RIIa was 452 RU, 469.2 RU, and 444.2 RU, respectively, and the amount of IgG1, IgG1-v1, and IgG1-v2 captured in the analysis of interactions with Fc ⁇ RIIIa was 454.5 RU, 470.8 RU, and 447.1 RU, respectively.
  • the R max values obtained from global fitting of sensorgrams obtained as a result of analyzing the interaction of IgG1 with H-type Fc ⁇ RIIa and Fc ⁇ RIIIa using the 1:1 Langmuir binding model were 69.8 RU and 63.8 RU, respectively.
  • the calculated R max values of IgG1-v1 and IgG1-v2 to Fc ⁇ RIIa type H were 72.5 RU and 68.6 RU, respectively, and the calculated R max values of IgG1-v1 and IgG1-v2 to Fc ⁇ RIIIa were 66.0 RU and 62.7 RU, respectively. These values were substituted into Equation 2 to calculate the KD of IgG1-v1 and IgG1-v2 for Fc ⁇ RIIa type H and Fc ⁇ RIIIa.
  • the KD values of IgG1, IgG1-v1, IgG1-v2, and IgG1-v3 for each Fc ⁇ R are shown in Table 1, and the relative KD values of IgG1-v1, IgG1-v2, and IgG1-v3 obtained by taking the KD values of IgG1 for each Fc ⁇ R and dividing them by the KD values of IgG1-v1, IgG1-v2, and IgG1-v3 for each Fc ⁇ R (the relative KD values of each antibody for each Fc ⁇ R) are shown in Table 2.
  • IgG1-v1 having an alteration of substituting Pro at position 238 (EU numbering) with Asp and IgG1-v2 having an alteration of substituting Leu at position 328 (EU numbering) with Glu have the properties of weakening the binding to all activating Fc ⁇ Rs including both allotypes of Fc ⁇ RIIa, while enhancing the binding to Fc ⁇ RIIb which is an inhibitory Fc ⁇ R.
  • selectivity of the obtained variant to Fc ⁇ RIIb was evaluated by using the ratio of Fc ⁇ RIIb-binding activity to the binding activity towards type R or type H of Fc ⁇ RIIa as the indicator.
  • I/A(R) or I/A(H) which is a value obtained by dividing the KD value for Fc ⁇ RIIa type R or type H by the KD value for Fc ⁇ RIIb, was used as an indicator for the selectivity of Fc ⁇ RIIb with respect to each Fc ⁇ RIIa.
  • This indicator has a greater value when the KD value for Fc ⁇ RIIb becomes smaller or when the KD value for Fc ⁇ RIIa becomes larger. That is, a variant that shows a larger value shows an increased binding activity for Fc ⁇ RIIb relative to Fc ⁇ RIIa.
  • IgG1-v3 which was produced by applying the existing technology showed a greater 1/A(H) value than that of IgG1 and a greater selectivity for Fc ⁇ RIIb, but a smaller 1/A(R) value than that of IgG1 and an improved selectivity for Fc ⁇ RIIb.
  • IgG1-v1 and IgG1-v2 found in the Examples have larger I/A(R) and I/A(H) values than those of IgG1, and improved selectivity for Fc ⁇ RIIb over both allotypes of Fc ⁇ RIIa.
  • IgG1-v3 described in Non-Patent Document 27 certainly shows a 408-fold enhanced binding to Fc ⁇ RIIb, while the binding to Fc ⁇ RIIa type H is decreased to 0.51 fold, and the binding to Fc ⁇ RIIa type R is enhanced to 522 fold.
  • IgG1-v1 and IgG1-v2 suppress their binding to both Fc ⁇ RIIa types R and H, and enhance their binding to Fc ⁇ RIIb, they are considered to be variants that bind with a greater Fc ⁇ RIIb selectivity compared with IgG1-v3.
  • alterations produced by substituting Pro at position 238 (EU numbering) with Asp or substituting Leu at position 328 (EU numbering) with Glu are very useful for the development of therapeutic agents for immunological inflammatory diseases and such.
  • IL6R-G1d_v1 (SEQ ID NO: 21) produced by introducing into IL6R-G1d the alteration produced by substituting Pro at position 238 (EU numbering) with Asp, the substitution of Leu at position 328 (EU numbering) with Glu as described in Example 2 which enhances selectivity for Fc ⁇ RIIb was introduced to produce the IL6R-G1d-v4 variant (SEQ ID NO: 25).
  • the obtained antibody having the amino acid sequence derived from IL6R-G1d-v4 as the antibody H chain has been named IgG1-v4.
  • the binding activities of IgG1, IgG1-v1, IgG1-v2, and IgG1-v4 to Fc ⁇ RIIb were evaluated according to the method of Reference Example 2, and those results are shown in Table 4.
  • IL6R-G1d-v1 (SEQ ID NO: 21) produced by introducing into IL6R-G1d the alteration produced by substituting Pro at position 238 (EU numbering) with Asp, the substitutions of Ser at position 267 (EU numbering) with Glu and of Leu at position 328 (EU numbering) with Phe as described in Example 2 which improve Fc ⁇ RIIb-binding activity were introduced, and the IL6R-G1d-v5 variant (SEQ ID NO: 26) was prepared according to the method of Reference Example 1.
  • the obtained antibody having the amino acid sequence derived from IL6R-G1d-v5 as the antibody H chain has been named IgG1-v5.
  • the Fc ⁇ RIIb-binding activities of IgG1, IgG1-v1, IgG1-v3, and IgG1-v5 were evaluated according to the method of Reference Example 2, and those results are shown in Table 5.
  • IL6R-F11 SEQ ID NO: 27
  • IL6R-F652 SEQ ID NO: 28
  • Expression plasmids containing an antibody H chain sequence were prepared for each of the antibody H chain sequences produced by substituting the region near the residue at position 238 (EU numbering) (positions 234 to 237, and 239 (EU numbering)) in IL6R-F652 each with 18 amino acids excluding the original amino acids and Cys.
  • IL6R-L SEQ ID NO: 22
  • PD variants Interactions of each PD variant with Fc ⁇ RIIa type R and Fc ⁇ RIIb were comprehensively evaluated by the method of Reference Example 2.
  • FIG. 7 shows relative values for the Fc ⁇ RIIb-binding activity obtained by additionally introducing these eleven alterations into a variant carrying the P238D alteration, and relative values for the Fc ⁇ RIIb-binding activity obtained by introducing these alterations into an Fc that does not contain the P238D alteration in Example 1.
  • These eleven alterations enhanced the amount of Fc ⁇ RIIb binding compared with before introduction when they were further introduced into the P238D variant, but on the contrary, the effect of lowering Fc ⁇ RIIb binding was observed for eight of those alterations except G237F, G237W, and S239D, when they were introduced into the variant that does not contain P238D (GpH7-B3/GpL16-k0) used in Example 1.
  • Example 3 and these results showed that from the effects of introducing alterations into a naturally-occurring IgG1, it is difficult to predict the effects of introducing the same alterations into the variant containing an Fc with the P238D alteration. In other words, it would not have been possible to discover these eight alterations identified this time without this investigation.
  • KD(IIaR)/KD(IIb) and KD(IIaH)/KD(IIb) in the table respectively show the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaR by the KD value of each variant for Fc ⁇ RIIb, and the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaH by the KD value of each variant for Fc ⁇ RIIb.
  • KD(IIb) of the parent polypeptide/KD(IIb) of the altered polypeptide refers to a value obtained by dividing the KD value of the parent polypeptide for Fc ⁇ RIIb by the KD value of each variant for Fc ⁇ RIIb.
  • Table 7 shows KD values for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of each variant/KD values for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide.
  • parent polypeptide refers to a variant which has IL6R-F11 (SEQ ID NO: 27) as the H chain. It was determined that due to weak binding of Fc ⁇ R to IgG, it was impossible to accurately analyze by kinetic analysis, and thus the gray-filled cells in Table 7 show values calculated by using Equation 2 of Reference Example 2.
  • Table 7 shows that all variants improved their affinity for Fc ⁇ RIIb in comparison with IL6R-F11, and the range of improvement was 1.9 fold to 5.0 fold.
  • the ratio of KD value of each variant for Fc ⁇ RIIaR/KD value of each variant for Fc ⁇ RIIb, and the ratio of KD value of each variant for Fc ⁇ RIIaH/KD value of each variant for Fc ⁇ RIIb represent an Fc ⁇ RIIb-binding activity relative to the Fc ⁇ RIIaR-binding activity and Fc ⁇ RIIaH-binding activity, respectively. That is, these values show the degree of binding selectivity of each variant for Fc ⁇ RIIb, and a larger value indicates a higher binding selectivity for Fc ⁇ RIIb.
  • the ratio of KD value for Fc ⁇ RIIaR/KD value for Fc ⁇ RIIb and the ratio of KD value for Fc ⁇ RIIaH/KD value for Fc ⁇ RIIb are both 0.7, and accordingly all variants in Table 7 showed improvement of binding selectivity for Fc ⁇ RIIb in comparison with the parent polypeptide.
  • the KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant/KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide is 1 or more, this means that the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant has equivalent or reduced binding compared with the binding by the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide.
  • the three-dimensional structure of the complex formed between an IgG1 Fc containing the P238D mutation (hereinafter, Fc(P238D)) and the extracellular region of Fc ⁇ RIIb was elucidated by X-ray crystallographic analysis, and the three-dimensional structure and binding mode were compared to those of the complex formed between the Fc of a naturally-occurring IgG1 (hereinafter, Fc(WT)) and the extracellular region of Fc ⁇ RIIb.
  • the three-dimensional structure of the Fc(WT)/Fc ⁇ RIIb extracellular region complex has not been analyzed, the three-dimensional structure of a complex formed with Fc(WT) is known for Fc ⁇ RIIa, and the extracellular regions of Fc ⁇ RIIa and Fc ⁇ RIIb match 93% in amino acid sequence and have very high homology.
  • the three-dimensional structure of the Fc(WT)/Fc ⁇ RIIb extracellular region complex was predicted by modeling using the crystal structure of the Fc(WT)/Fc ⁇ RIIa extracellular region complex.
  • the three-dimensional structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex was determined by X-ray crystallographic analysis at 2.6 ⁇ resolution. The structure obtained as a result of this analysis is shown in FIG. 8 .
  • the Fc ⁇ RIIb extracellular region is bound between two Fc CH2 domains, and this is similar to the three-dimensional structures of complexes formed between Fc(WT) and the respective extracellular region of Fc ⁇ RIIIa, Fc ⁇ RIIIb, or Fc ⁇ RIIa analyzed so far.
  • the amino acid at position 160 is one of the few differences between Fc ⁇ RIIa and Fc ⁇ RIIb at the interface of interaction with Fc, the presence of this hydrogen bond which is specific to Fc ⁇ RIIb is presumed to have led to improvement of Fc ⁇ RIIb-binding activity and decrease of Fc ⁇ RIIa-binding activity in Fc(P238D), and improvement of its selectivity. Furthermore, in Fc CH2 domain B, an electrostatic interaction is observed between Asp at position 270 (EU numbering) and Arg at position 131 in Fc ⁇ RIIb ( FIG. 12 ). In Fc ⁇ RIIa type H, which is one of the allotypes of Fc ⁇ RIIa, the corresponding residue is His, and therefore cannot form this electrostatic interaction.
  • An Fc containing the P238D alteration was prepared as follows. First, Cys at position 220 (EU numbering) of hIL6R-IgG1-v1 (SEQ ID NO: 21) was substituted with Ser. Then, genetic sequence of Fc(P238D) from Glu at position 236 (EU numbering) to its C terminal was cloned by PCR. Using this cloned genetic sequence, production of expression vectors, and expression and purification of Fc(P238D) were carried out according to the method of Reference Example 1. Cys at position 220 (EU numbering) forms a disulfide bond with Cys of the L chain in general IgG1. The L chain is not co-expressed when Fc alone is prepared, and therefore, this residue was substituted with Ser to avoid formation of unnecessary disulfide bonds.
  • Endo F1 Protein Science 1996, 5: 2617-2622 expressed and purified from Escherichia coli as a glutathione S-transferase fusion protein was added. This was allowed to remain at room temperature for three days in 0.1 M Bis-Tris buffer at pH 6.5, and the N-linked oligosaccharide was cleaved, leaving N-acetylglucosamine directly bound to Asn.
  • this Fc ⁇ RIIb extracellular domain sample subjected to carbohydrate cleavage treatment was concentrated by ultrafiltration with 5000 MWCO, and purified by gel filtration chromatography (Superdex200 10/300) using a column equilibrated in 20 mM HEPS at pH 7.5 containing 0.05 M NaCl.
  • Fc(P238D) was added so that the molar ratio of the Fc ⁇ RIIb extracellular region would be present in slight excess, and after concentration by ultrafiltration with 10,000 MWCO, a sample of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex was obtained through purification by gel filtration chromatography (Superdex200 10/300) using a column equilibrated in 20 mM HEPS at pH 7.5 containing 0.05 M NaCl.
  • MATRIX Hydra II Plus One
  • Crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex was determined by the molecular replacement method using the program Phaser (CCP4 Software Suite). From the size of the obtained crystal lattice and the molecular weight of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex, the number of complexes in the asymmetric unit was predicted to be one. From the structural coordinates of PDB code: 3 SGJ which is the crystal structure of the Fc(WT)/Fc ⁇ RIIIa extracellular region complex, the amino acid residue portions of the A chain positions 239-340 and the B chain positions 239-340 were taken out as separate coordinates, and they were used respectively as models for searching the Fc CH2 domains.
  • the amino acid residue portions of the A chain positions 341-444 and the B chain positions 341-443 were taken out as a single set of coordinates from the same structural coordinates of PDB code: 3SGJ; and this was used as a model for searching the Fc CH3 domains. Finally, from the structural coordinates of PDB code: 2FCB which is a crystal structure of the Fc ⁇ RIIb extracellular region, the amino acid residue portions of the A chain positions 6-178 was taken out and used as a model for searching the Fc ⁇ RIIb extracellular region.
  • each search model in the crystal lattice were determined in the order of Fc CH3 domain, Fc ⁇ RIIb extracellular region, and Fc CH2 domain, based on the rotation function and translation function to obtain the initial model for the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • the crystallographic reliability factor, R value became 40.4%
  • the Free R value became 41.9% to diffraction intensity data from 25 ⁇ to 3.0 ⁇ at this point.
  • IL6R-B3 (SEQ ID NO: 40) was produced by introducing into IL6R-G1d (SEQ ID NO: 20) produced in Example 2, the alteration produced by substituting Lys at position 439 (EU numbering) with Glu.
  • IL6R-BF648 (SEQ ID NO: 41) was produced by introducing into IL6R-B3, the alteration produced by substituting Pro at position 238 (EU numbering) with Asp.
  • IL6R-L (SEQ ID NO: 22) was utilized as the common antibody L chain for all of the antibodies.
  • a figure was produced according to the following method for the results of analyzing the interactions with the respective Fc ⁇ Rs.
  • the value for the amount of binding of each variant to each Fc ⁇ R was divided by the value for the amount of binding of the pre-altered control antibody (IL6R-BF648/IL6R-L with Pro at position 238 (EU numbering) substituted with Asp) to each Fc ⁇ R, and the obtained was then multiplied by 100 and used as the relative binding activity value of each variant to each Fc ⁇ R.
  • the horizontal axis shows the relative binding activity value of each variant to Fc ⁇ RIIb, and the vertical axis shows the relative binding activity value of each variant to Fc ⁇ RIIa type R ( FIG. 13 ).
  • KD(IIaR)/KD(IIb) and KD(IIaH)/KD(IIb) in the table respectively represent the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaR by the KD value of each variant for Fc ⁇ RIIb, and the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaH by the KD value of each variant for Fc ⁇ RIIb.
  • KD(IIb) of the parent polypeptide/KD(IIb) of the altered polypeptide refers to the value obtained by dividing the KD value of the parent polypeptide for Fc ⁇ RIIb by the KD value of each variant for Fc ⁇ RIIb.
  • the KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of each variant/KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide are shown in Table 10.
  • parent polypeptide refers to the variant which has IL6R-B3 (SEQ ID NO: 40) as the H chain. It was determined that due to weak binding of Fc ⁇ R to IgG, it was impossible to accurately analyze by kinetic analysis, and thus the gray-filled cells in Table 10 show values calculated by using Equation 2 of Reference Example 2.
  • Table 10 shows that in comparison with IL6R-B3, all variants showed improvement of affinity for Fc ⁇ RIIb, and the range of improvement was 2.1 fold to 9.7 fold.
  • the ratio of KD value of each variant for Fc ⁇ RIIaR/KD value of each variant for Fc ⁇ RIIb, and the ratio of KD value of each variant for Fc ⁇ RIIaH/KD value of each variant for Fc ⁇ RIIb represent an Fc ⁇ RIIb-binding activity relative to the Fc ⁇ RIIaR-binding activity and Fc ⁇ RIIaH-binding activity, respectively. That is, these values show the degree of binding selectivity of each variant for Fc ⁇ RIIb, and a greater value indicates a higher binding selectivity for Fc ⁇ RIIb.
  • the KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant/KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide is 1 or more, this means that the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant has equivalent or decreased binding compared with the binding by the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide.
  • FIG. 14 shows the crystal structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • the H chain positioned on the left side is Fc Chain A
  • the H chain positioned on the right side is Fc Chain B.
  • the site at position 233 (EU numbering) in Fc Chain A is located near Lys at position 113 (EU numbering) of Fc ⁇ RIIb.
  • the E233 side chain is in a condition of considerably high mobility, and its electron density is not well observed.
  • FIG. 15 shows the environment near the site at position 330 (EU numbering) in the structure of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex.
  • This figure shows that the environment around the site at position 330 (EU numbering) of Fc Chain A of Fc (P238D) is a hydrophilic environment composed of Ser at position 85, Glu at position 86, Lys at position 163, and such (EU numbering) of Fc ⁇ RIIb. Therefore, the alteration produced by substituting Ala at position 330 (EU numbering) with Lys or Arg is speculated to contribute to strengthening the interaction with Ser at position 85 (EU numbering) or Glu at position 86 (EU numbering) in Fc ⁇ RIIb.
  • FIG. 16 depicts the structures of Pro at position 271 (EU numbering) of Fc Chain B after superimposing the crystal structures of the Fc(P238D)/Fc ⁇ RIIb extracellular region complex and the Fc(WT)/Fc ⁇ RIIIa extracellular region complex by the least squares fitting based on the C ⁇ atom pair distances with respect to Fc Chain B. These two structures match well, but have different three-dimensional structures of Pro at position 271 (EU numbering).
  • IL6R-L was utilized as the common antibody L chain for all of the antibodies, the antibodies were expressed and purified according to the method of Reference Example 1, and binding to each of the Fc ⁇ Rs (Fc ⁇ RIa, Fc ⁇ RIIa H type, Fc ⁇ RIIa R type, Fc ⁇ RIIb, and Fc ⁇ RIIIa V type) was comprehensively evaluated by the method of Reference Example 2.
  • Relative binding activities were calculated for the results of analyzing interactions with the respective Fc ⁇ Rs according to the following method.
  • the value for the amount of binding of each variant to each Fc ⁇ R was divided by the value for the amount of binding of the pre-altered control antibody (IL6R-BF648/IL6R-L with substitution of Pro at position 238 (EU numbering) with Asp to each Fc ⁇ R, and multiplied by 100; and then the value was used as the relative binding activity value of each variant to each Fc ⁇ R.
  • the horizontal axis shows the relative binding activity value of each variant to Fc ⁇ RIIb, and the vertical axis shows the relative binding activity value of each variant to Fc ⁇ RIIa type R (Table 11).
  • SEQ ID NO refers to the SEQ ID NO of the H chain of the evaluated variant
  • alteration refers to the alteration introduced into IL6R-B3 (SEQ ID NO: 40).
  • the template used for producing IL6R-B3, IL6R-G1d/IL6R-L, is indicated with an asterisk (*).
  • KD(IIaR)/KD(IIb) and KD(IIaH)/KD(IIb) in the table respectively represent the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaR by the KD value of each variant for Fc ⁇ RIIb, and the value obtained by dividing the KD value of each variant for Fc ⁇ RIIaH by the KD value of each variant for Fc ⁇ RIIb.
  • KD(IIb) of the parent polypeptide/KD(IIb) of the altered polypeptide refers to the value obtained by dividing the KD value of the parent polypeptide for Fc ⁇ RIIb by the KD value of each variant for Fc ⁇ RIIb.
  • the KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of each variant/KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide are shown in Table 12.
  • parent polypeptide refers to the variant which has IL6R-B3 (SEQ ID NO: 40) as the H chain. It was determined that due to weak binding of Fc ⁇ R to IgG, it was impossible to accurately analyze by kinetic analysis, and thus the gray-filled cells in Table 12 show values calculated by using Equation 2 of Reference Example 2.
  • Table 12 shows that in comparison with IL6R-B3, all variants showed improvement of affinity for Fc ⁇ RIIb, and the range of improvement was 3.0 fold to 99.0 fold.
  • the ratio of KD value of each variant for Fc ⁇ RIIaR/KD value of each variant for Fc ⁇ RIIb, and the ratio of KD value of each variant for Fc ⁇ RIIaH/KD value of each variant for Fc ⁇ RIIb represent an Fc ⁇ RIIb-binding activity relative to the Fc ⁇ RIIaR-binding activity and Fc ⁇ RIIaH-binding activity, respectively. That is, those values show the degree of binding selectivity of each variant for Fc ⁇ RIIb, and a greater value indicates a higher binding selectivity for Fc ⁇ RIIb.
  • the KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant/KD value for the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide is 1 or more, this means that the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of a variant has equivalent or decreased binding compared with the binding by the stronger of the Fc ⁇ RIIaR- and Fc ⁇ RIIaH-binding activities of the parent polypeptide.
  • Synthesis of full-length genes encoding the nucleotide sequences of the H chain and L chain of the antibody variable regions was carried out by production methods known to those skilled in the art using Assemble PCR and such. Introduction of amino acid substitutions was carried out by methods known to those skilled in the art using PCR or such.
  • the obtained plasmid fragment was inserted into an animal cell expression vector, and the H-chain expression vector and L-chain expression vector were produced.
  • the nucleotide sequence of the obtained expression vector was determined by methods known to those skilled in the art.
  • the produced plasmids were introduced transiently into the HEK293H cell line derived from human embryonic kidney cancer cells (Invitrogen) or into FreeStyle293 cells (Invitrogen) for antibody expression.
  • the obtained culture supernatant was collected, and then passed through a 0.22 ⁇ m MILLEX(R)-GV filter (Millipore), or through a 0.45 ⁇ m MILLEX(R)-GV filter (Millipore) to obtain the culture supernatant.
  • Antibodies were purified from the obtained culture supernatant by methods known to those skilled in the art using rProtein A Sepharose Fast Flow (GE Healthcare) or Protein G Sepharose 4 Fast Flow (GE Healthcare). For the concentration of the purified antibodies, their absorbance at 280 nm was measured using a spectrophotometer. From the obtained value, the extinction coefficient calculated by the methods such as PACE was used to calculate the antibody concentration (Protein Science 1995; 4: 2411-2423).
  • Extracellular domains of Fc ⁇ Rs were prepared by the following method. First, a gene of the extracellular domain of Fc ⁇ R was synthesized by a method well known to those skilled in the art. At that time, the sequence of each Fc ⁇ R was produced based on the information registered at NCBI. Specifically, Fc ⁇ RI was produced based on the sequence of NCBI Accession No. NM — 000566.3, Fc ⁇ RIIa was produced based on the sequence of NCBI Accession No. NM — 001136219.1, Fc ⁇ RIIb was produced based on the sequence of NCBI Accession No. NM — 004001.3, Fc ⁇ RIIIa was produced based on the sequence of NCBI Accession No.
  • Fc ⁇ RIIIb was produced based on the sequence of NCBI Accession No. NM — 000570.3, and a His tag was attached to the C terminus Furthermore, polymorphism is known for Fc ⁇ RIIa, Fc ⁇ RIIIa, and Fc ⁇ RIIIb, and the polymorphic sites were produced by referring to J. Exp. Med., 1990, 172: 19-25 for Fc ⁇ RIIa; J. Clin. Invest., 1997, 100 (5): 1059-1070 for Fc ⁇ RIIIa; and J. Clin. Invest., 1989, 84, 1688-1691 for Fc ⁇ RIIIb.
  • the obtained gene fragments were inserted into an animal cell expression vector, and expression vectors were produced.
  • the produced expression vectors were introduced transiently into human embryonic kidney cancer cell line-derived FreeStyle293 cells (Invitrogen) to express the proteins of interest.
  • Fc ⁇ RIIb used for crystallographic analysis
  • the protein of interest was expressed in the presence of Kifunensine at a final concentration of 10 ⁇ g/mL, so that the sugar chain added to Fc ⁇ RIIb will be the high-mannose type.
  • Cells were cultured, and after collection of the obtained culture supernatant, this was passed through a 0.22 ⁇ m filter to obtain the culture supernatant.
  • the obtained culture supernatants were purified in the following four steps.
  • step 1 cation exchange column chromatography
  • step 2 affinity column chromatography
  • step 3 gel filtration column chromatography
  • step 4 anion exchange column chromatography using Q sepharose FF was performed as step 1.
  • the purified proteins were subjected to absorbance measurements at 280 nm using a spectrophotometer; and from the obtained values, the concentrations of the purified proteins were calculated using the absorption coefficient calculated using methods such as PACE (Protein Science 1995; 4: 2411-2423).
  • Series S Sensor Chip CMS GE Healthcare
  • Series S sensor Chip CM4 GE Healthcare
  • an Fc ⁇ receptor diluted with the running buffer was allowed to interact, the amount bound to an antibody was measured, and the antibodies were compared.
  • the amount of Fc ⁇ receptor bound depends on the amount of the captured antibodies, the amount of Fc ⁇ receptor bound was divided by the amount of each antibody captured to obtain corrected values, and these values were compared.
  • antibodies captured onto the chips were washed by reaction with 10 mM glycine-HCl, pH 1.5, and the chips were regenerated and used repeatedly.
  • Equation 1 The behavior of interacting molecules according to the 1:1 binding model on Biacore can be described by Equation 1 shown below.
  • KD can be expressed as Equation 2 shown below.
  • KD can be calculated by substituting the values of R max , RI, and C into this equation.
  • the values of RI and C can be determined from the sensorgram of the measurement results and measurement conditions.
  • R max was calculated according to the following method. As a target of comparison, for antibodies that had sufficiently strong interactions as evaluated simultaneously in the same round of measurement, the R max value was obtained through global fitting using the 1:1 Langmuir binding model, and then it was divided by the amount of the comparison antibody captured onto the sensor chip, and multiplied by the captured amount of an altered antibody to be evaluated.
  • Polypeptides comprising an Fc region that have maintained or decreased binding activities towards both allotypes of Fc ⁇ RIIa, types R and H, and having enhanced Fc ⁇ RIIb-binding activity in comparison with the parent polypeptide are provided by the present invention.
  • polypeptides with enhanced binding selectivity for Fc ⁇ RIIb rather than for both allotypes of Fc ⁇ RIIa (types R and H)
  • anti-antibody production may be suppressed through Fc ⁇ RIIb-mediated immunosuppressive actions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Neurology (AREA)
  • Dermatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Virology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Neurosurgery (AREA)
  • Pulmonology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Communicable Diseases (AREA)
  • Cardiology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Oncology (AREA)
US14/001,218 2011-02-25 2012-02-24 Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY Abandoned US20140093496A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2011040923 2011-02-25
JP2011-040923 2011-02-25
JP2011219835 2011-10-04
JP2011-219835 2011-10-04
PCT/JP2012/054624 WO2012115241A1 (ja) 2011-02-25 2012-02-24 FcγRIIb特異的Fc抗体

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2012/054624 A-371-Of-International WO2012115241A1 (ja) 2011-02-25 2012-02-24 FcγRIIb特異的Fc抗体

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/846,672 Division US20230174655A1 (en) 2011-02-25 2022-06-22 Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY

Publications (1)

Publication Number Publication Date
US20140093496A1 true US20140093496A1 (en) 2014-04-03

Family

ID=46721013

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/001,218 Abandoned US20140093496A1 (en) 2011-02-25 2012-02-24 Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
US14/007,947 Active 2033-02-09 US10618965B2 (en) 2011-02-25 2012-03-30 Method for altering plasma retention and immunogenicity of antigen-binding molecule
US16/806,027 Active US11718678B2 (en) 2011-02-25 2020-03-02 Method for altering plasma retention and immunogenicity of antigen-binding molecule
US17/846,672 Pending US20230174655A1 (en) 2011-02-25 2022-06-22 Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/007,947 Active 2033-02-09 US10618965B2 (en) 2011-02-25 2012-03-30 Method for altering plasma retention and immunogenicity of antigen-binding molecule
US16/806,027 Active US11718678B2 (en) 2011-02-25 2020-03-02 Method for altering plasma retention and immunogenicity of antigen-binding molecule
US17/846,672 Pending US20230174655A1 (en) 2011-02-25 2022-06-22 Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY

Country Status (12)

Country Link
US (4) US20140093496A1 (ja)
EP (2) EP3604330A1 (ja)
JP (7) JP6032818B2 (ja)
KR (3) KR102147548B1 (ja)
CN (2) CN103492565B (ja)
AU (2) AU2012222252B2 (ja)
BR (2) BR112013021526B1 (ja)
CA (1) CA2827923C (ja)
MX (1) MX352889B (ja)
RU (1) RU2608504C2 (ja)
SG (2) SG10201609665PA (ja)
WO (1) WO2012115241A1 (ja)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015187779A1 (en) 2014-06-03 2015-12-10 Xbiotech, Inc. Compositions and methods for treating and preventing staphylococcus aureus infections
US20160039912A1 (en) * 2013-04-02 2016-02-11 Chugai Seiyaku Kabushiki Kaisha Fc REGION VARIANT
US9890218B2 (en) 2011-06-30 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US10526408B2 (en) 2015-08-28 2020-01-07 Research Development Foundation Engineered antibody FC variants
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11091539B2 (en) 2016-07-08 2021-08-17 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11142563B2 (en) 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
WO2022004761A1 (en) * 2020-06-30 2022-01-06 Chugai Seiyaku Kabushiki Kaisha Site specific notch-activating molecule and uses thereof
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11248042B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Polynucleotides encoding anti-ApoC3 antibodies
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11851476B2 (en) 2011-10-31 2023-12-26 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
US20240025995A1 (en) * 2021-10-21 2024-01-25 Seismic Therapeutic, Inc. Dual targeted immune regulating compositions
US12030945B2 (en) 2023-10-25 2024-07-09 Seismic Therapeutic, Inc. Variant IgG Fc polypeptides and uses thereof

Families Citing this family (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5624276B2 (ja) 2006-03-31 2014-11-12 中外製薬株式会社 抗体の血中動態を制御する方法
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
TWI564021B (zh) 2008-04-11 2017-01-01 Chugai Pharmaceutical Co Ltd Repeated binding of antigen to antigen binding molecules
TWI440469B (zh) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
MX365235B (es) 2010-11-30 2019-05-28 Chugai Pharmaceutical Co Ltd Molécula de unión a antígeno capaz de unir repetidamente a la pluralidad de moléculas de antígeno.
CN103429737B (zh) 2010-11-30 2020-07-14 中外制药株式会社 细胞毒诱导治疗剂
EP3825325A3 (en) 2011-03-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
CA2850322C (en) 2011-09-30 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule inducing immune response to target antigen
BR112014009069A2 (pt) 2011-10-13 2020-10-27 Bristol-Myers Squibb Company polipeptídeos de anticorpo que antagonizam cd40l
TW202015731A (zh) * 2012-02-24 2020-05-01 日商中外製藥股份有限公司 經FcγRIIB促進抗原消失之抗原結合分子
CA2874721A1 (en) * 2012-05-30 2013-12-05 Tomoyuki Igawa Target tissue-specific antigen-binding molecule
EP3597747B1 (en) 2012-08-24 2023-03-15 Chugai Seiyaku Kabushiki Kaisha Mouse fcgammarii-specific fc antibody
DK2895513T3 (en) * 2012-09-12 2018-09-03 Genzyme Corp FC-CONTAINING POLYPEPTIDES WITH CHANGED GLYCOSYLATION AND REDUCED EFFECTOR FUNCTION
WO2014113510A1 (en) 2013-01-15 2014-07-24 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US20140377253A1 (en) * 2013-03-15 2014-12-25 Abbvie Biotherapeutics Inc. Fc variants
KR20210094669A (ko) 2013-04-29 2021-07-29 에프. 호프만-라 로슈 아게 인간 fcrn-결합 변형된 항체 및 사용 방법
RU2655439C2 (ru) * 2013-05-31 2018-05-28 Займворкс Инк. Гетеромультимеры с уменьшенной или подавленной эффекторной функцией
DK3050896T3 (da) 2013-09-27 2021-07-19 Chugai Pharmaceutical Co Ltd Fremgangsmåde til fremstilling af en polypeptid-heteromultimer
JP2016538283A (ja) 2013-11-13 2016-12-08 ザイムワークス,インコーポレイテッド Egfr及び/またはher2を標的にする一価抗原結合性構築物及びその使用
SG11201604495PA (en) 2013-12-04 2016-07-28 Chugai Pharmaceutical Co Ltd Antigen-binding molecules, the antigen-binding activity of which varies according to the concentration of compounds, and libraries of said molecules
KR20160104636A (ko) 2014-01-15 2016-09-05 에프. 호프만-라 로슈 아게 단백질 A-결합이 개선된 Fc-영역 변이체
IL247715B (en) 2014-04-07 2022-07-01 Chugai Pharmaceutical Co Ltd Antigen binding molecules that activate the immune system
EP3144388B1 (en) 2014-05-13 2020-07-01 Chugai Seiyaku Kabushiki Kaisha T cell-redirecting antigen-binding molecule for cells having immunosuppression function
US20160060360A1 (en) 2014-07-24 2016-03-03 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
FR3024453B1 (fr) * 2014-08-01 2018-06-29 Lab Francais Du Fractionnement Procede de production de variants ayant un fc presentant une sialylation amelioree
MA40764A (fr) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd Agent thérapeutique induisant une cytotoxicité
DK3221346T3 (da) 2014-11-21 2020-10-12 Bristol Myers Squibb Co Antistoffer omfattende modificerede konstante områder af tungkæden
HUE050596T2 (hu) 2014-11-21 2020-12-28 Bristol Myers Squibb Co Antitestek CD73 ellen és azok felhasználásai
CN113045650A (zh) 2014-12-19 2021-06-29 中外制药株式会社 抗-c5抗体及使用方法
AU2015369683C1 (en) 2014-12-23 2024-06-20 Bristol-Myers Squibb Company Antibodies to TIGIT
US10633433B2 (en) 2015-01-28 2020-04-28 Prothena Biosciences Limited Anti-transthyretin antibodies
US9879080B2 (en) 2015-01-28 2018-01-30 Prothena Biosciences Limited Anti-transthyretin antibodies
US10464999B2 (en) 2015-01-28 2019-11-05 Prothena Biosciences Limited Anti-transthyretin antibodies
JP6724023B2 (ja) 2015-02-09 2020-07-15 リサーチ ディベロップメント ファウンデーション 改善された補体活性化を示す操作された免疫グロブリンfcポリペプチド
AU2016219534B2 (en) 2015-02-09 2021-07-01 Massachusetts Institute Of Technology Multi-specific antibodies with affinity for human A33 antigen and dota metal complex and uses thereof
WO2016130986A1 (en) * 2015-02-13 2016-08-18 Sorrento Therapeutics, Inc. Antibody therapeutics that bind ctla4
KR101892883B1 (ko) 2015-02-27 2018-10-05 추가이 세이야쿠 가부시키가이샤 Il-6 관련 질환 치료용 조성물
HRP20230060T1 (hr) 2015-05-29 2023-03-17 Bristol-Myers Squibb Company Antitijela protiv ox40 i njihova primjena
EP3305322A4 (en) * 2015-06-05 2018-12-26 Chugai Seiyaku Kabushiki Kaisha Combined use of immune activators
CA2990012A1 (en) 2015-06-29 2017-01-05 Bristol-Myers Squibb Company Antibodies to cd40
KR102538745B1 (ko) 2015-09-18 2023-06-01 추가이 세이야쿠 가부시키가이샤 Il-8에 결합하는 항체 및 그의 사용
JP6931329B2 (ja) 2015-11-18 2021-09-01 中外製薬株式会社 免疫抑制機能を有する細胞に対するt細胞リダイレクト抗原結合分子を用いた併用療法
US11649293B2 (en) 2015-11-18 2023-05-16 Chugai Seiyaku Kabushiki Kaisha Method for enhancing humoral immune response
BR112018011781A2 (pt) 2015-12-14 2018-12-04 Macrogenics Inc molécula biespecífica possuindo um ou mais sítios de ligação a epítopo capazes de ligação imunoespecífica a (um) epítopo(s) de pd-1 e um ou mais sítios de ligação a epítopo capazes de ligação imunoespecífica a (um) epítopo(s) de ctla-4, e composição farmacêutica
US20190284293A1 (en) 2016-03-04 2019-09-19 Bristol-Myers Squibb Company Combination therapy with anti-cd73 antibodies
RU2018141874A (ru) * 2016-04-29 2020-05-29 Виела Байо, Инк. СВЯЗЫВАЮЩИЕ МОЛЕКУЛЫ, СПЕЦИФИЧНЫЕ ДЛЯ FCγRIIA, И ИХ ПРИМЕНЕНИЕ
CN115304669A (zh) * 2016-06-08 2022-11-08 上海交通大学医学院 增强激动型抗体活性的抗体重链恒定区序列
SG10201806693XA (en) 2016-06-17 2018-09-27 Chugai Pharmaceutical Co Ltd Anti-myostatin antibodies and methods of use
AU2017325654A1 (en) 2016-08-02 2019-02-14 Visterra, Inc. Engineered polypeptides and uses thereof
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
JP7191833B2 (ja) 2017-01-30 2022-12-19 中外製薬株式会社 抗スクレロスチン抗体およびその使用
SG11201907299XA (en) 2017-02-08 2019-09-27 Dragonfly Therapeutics Inc Multi-specific binding proteins for activation of natural killer cells and therapeutic uses thereof to treat cancer
EP3579866A4 (en) * 2017-02-08 2020-12-09 Dragonfly Therapeutics, Inc. HEAVY CHAIN VARIABLE ANTIBODY DOMAINS WITH NKG2D RECEPTOR AIMING
ES2955074T3 (es) 2017-02-20 2023-11-28 Dragonfly Therapeutics Inc Proteínas que se unen a HER2, NKG2D Y CD16
TWI788340B (zh) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 抗icos促效劑抗體及其用途
WO2018203545A1 (ja) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Il-6及び好中球の関連する疾患の治療効果の予測及び判定方法
CN116333129A (zh) 2017-05-25 2023-06-27 百时美施贵宝公司 包含经修饰的重链恒定区的抗体
CA3065171A1 (en) * 2017-06-05 2018-12-13 Janssen Biotech, Inc. Engineered multispecific antibodies and other multimeric proteins with asymmetrical ch2-ch3 region mutations
EP3691447A4 (en) 2017-10-06 2021-08-11 Prothena Biosciences Limited ANTI-TRANSTHYRETINE ANTIBODIES
EA202091130A1 (ru) 2017-11-29 2020-08-28 Протена Биосайенсис Лимитед Лиофилизованный препарат моноклонального антитела к транстиретину
JP7284759B2 (ja) 2017-12-27 2023-05-31 ブリストル-マイヤーズ スクイブ カンパニー 抗cd40抗体およびその使用
WO2019157366A1 (en) 2018-02-08 2019-08-15 Dragonfly Therapeutics, Inc. Antibody variable domains targeting the nkg2d receptor
EP3765494A4 (en) 2018-03-15 2022-03-23 Chugai Seiyaku Kabushiki Kaisha ANTI-DENGUE VIRUS ANTIBODIES WITH CROSS-REACTIVITY AGAINST ZIKA VIRUS AND METHODS OF USE
SG11202003531WA (en) 2018-08-10 2020-05-28 Chugai Pharmaceutical Co Ltd Anti-cd137 antigen-binding molecule and utilization thereof
CN113348177A (zh) 2018-11-28 2021-09-03 百时美施贵宝公司 包含经修饰的重链恒定区的抗体
US20220153875A1 (en) 2019-03-19 2022-05-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing antigen-binding domain of which binding activity to antigen is changed depending on mta, and library for obtaining said antigen-binding domain
US20230165967A1 (en) * 2019-10-04 2023-06-01 TAE Life Sciences Antibody Compositions Comprising Fc Mutations and Site-Specific Conjugation Properties for use in Treating Cancer, Immunological Disorders, and Methods Thereof
EP4061420A1 (en) * 2019-11-20 2022-09-28 Abvision, Inc. Monoclonal antibodies that target human cd47 protein
PE20221661A1 (es) 2019-12-18 2022-10-26 Hoffmann La Roche Anticuerpos anti-ccl2 biespecificos
TW202140553A (zh) 2020-01-13 2021-11-01 美商威特拉公司 C5ar1抗體分子及其用途
TW202144395A (zh) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 用於癌症之治療的抗cd137抗原結合分子
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
CN112649417B (zh) * 2020-12-01 2021-10-29 西北大学 一种检测mmp-14的电化学发光生物传感器及其制备方法
EP4277931A1 (en) 2021-01-13 2023-11-22 Visterra, Inc. Humanized complement 5a receptor 1 antibodies and methods of use thereof
CN117500829A (zh) 2021-06-18 2024-02-02 豪夫迈·罗氏有限公司 双特异性抗ccl2抗体
CN113834939A (zh) * 2021-09-18 2021-12-24 胜亚生物科技(厦门)有限公司 一种用于检测川崎病及疗效评估的试剂、试剂盒及应用
GB202204016D0 (en) * 2022-03-22 2022-05-04 Ucl Business Plc Affinity chromatography ligands for antibody glycovariant separation
US11780910B1 (en) 2022-05-02 2023-10-10 Novo Nordisk A/S Anti-ANGPTL3 antibodies suitable for high concentration compositions and subcutaneous administration
WO2023239940A1 (en) 2022-06-10 2023-12-14 Research Development Foundation Engineered fcriib selective igg1 fc variants and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US20110021755A1 (en) * 2003-03-03 2011-01-27 Xencor, Inc. Optimized Fc Variants

Family Cites Families (288)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5144499B1 (ja) 1970-08-29 1976-11-29
JPS5334319B2 (ja) 1971-12-28 1978-09-20
JPS5334319U (ja) 1976-08-28 1978-03-25
US4769320A (en) 1982-07-27 1988-09-06 New England Medical Center Hospitals, Inc. Immunoassay means and methods useful in human native prothrombin and human abnormal prothorombin determinations
EP0091539B2 (en) 1982-03-31 1996-11-27 Ajinomoto Co., Inc. Gene coding for interleukin-2 polypeptide, recombinant DNA carrying said gene, cell lines possessing the recombinant DNA,and method for producing interleukin-2 using said cells
CA1209907A (en) 1982-04-12 1986-08-19 Richard M. Bartholomew Method of affinity purification employing monoclonal antibodies
US4689299A (en) 1982-09-30 1987-08-25 University Of Rochester Human monoclonal antibodies against bacterial toxins
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
JPH06104071B2 (ja) 1986-08-24 1994-12-21 財団法人化学及血清療法研究所 第▲ix▼因子コンホメ−シヨン特異性モノクロ−ナル抗体
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
US4851341A (en) 1986-12-19 1989-07-25 Immunex Corporation Immunoaffinity purification system
US5004697A (en) 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
JPH01144991A (ja) * 1987-12-02 1989-06-07 Kagaku Oyobi Ketsusei Riyouhou Kenkyusho 血液凝固第8因子の精製方法
US5322678A (en) 1988-02-17 1994-06-21 Neorx Corporation Alteration of pharmacokinetics of proteins by charge modification
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
CA2006684C (en) 1988-12-30 1996-12-17 Charles T. Esmon Monoclonal antibody against protein c
US5202253A (en) 1988-12-30 1993-04-13 Oklahoma Medical Research Foundation Monoclonal antibody specific for protein C and antibody purification method
JPH0636741B2 (ja) * 1989-11-08 1994-05-18 帝人株式会社 ヒト・プロテインcの分離方法
WO1991012023A2 (en) 1990-02-16 1991-08-22 Boston Biomedical Research Institute Hybrid reagents capable of selectively releasing molecules into cells
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
DK0585287T3 (da) 1990-07-10 2000-04-17 Cambridge Antibody Tech Fremgangsmåde til fremstilling af specifikke bindingsparelementer
KR100272077B1 (ko) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 이종 항체를 생산할 수 있는 전이유전자를 가진 인간이외의 동물
AU668349B2 (en) 1991-04-25 1996-05-02 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
ES2341666T3 (es) 1991-12-02 2010-06-24 Medimmune Limited Produccion de autoanticuerpos de repertorios de segmentos de anticue rpos expresados en la superficie de fagos.
CA2124967C (en) 1991-12-17 2008-04-08 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
GB9203459D0 (en) 1992-02-19 1992-04-08 Scotgen Ltd Antibodies with germ-line variable regions
CA2131151A1 (en) 1992-03-24 1994-09-30 Kevin S. Johnson Methods for producing members of specific binding pairs
JPH07509137A (ja) 1992-07-24 1995-10-12 セル ジェネシス,インク. 異種抗体の生産
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
EP0754225A4 (en) 1993-04-26 2001-01-31 Genpharm Int HETEROLOGIC ANTIBODY-PRODUCING TRANSGENIC NON-HUMAN ANIMALS
EP0710288B1 (en) 1993-06-10 2006-04-05 Genetic Therapy, Inc. Adenoviral vectors for treatment of hemophilia
GB9313509D0 (en) 1993-06-30 1993-08-11 Medical Res Council Chemisynthetic libraries
FR2707189B1 (fr) 1993-07-09 1995-10-13 Gradient Ass Procédé de traitement de résidus de combustion et installation de mise en Óoeuvre dudit procédé.
GB9314271D0 (en) 1993-07-09 1993-08-18 Inst Of Cancer The Research Cell growth factor receptors
IL107742A0 (en) 1993-11-24 1994-02-27 Yeda Res & Dev Chemically-modified binding proteins
JPH09506508A (ja) 1993-12-03 1997-06-30 メディカル リサーチ カウンシル 組換え結合タンパク質およびペプチド
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
WO1996002576A1 (fr) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Anticorps humain reconstitue contre l'interleukine-8 humaine
US6309636B1 (en) 1995-09-14 2001-10-30 Cancer Research Institute Of Contra Costa Recombinant peptides derived from the Mc3 anti-BA46 antibody, methods of use thereof, and methods of humanizing antibody peptides
ES2384222T3 (es) 1994-10-07 2012-07-02 Chugai Seiyaku Kabushiki Kaisha Inhibición del crecimiento anómalo de células sinoviales utilizando un antagonista de IL-6 como principio activo
WO1996012503A1 (fr) 1994-10-21 1996-05-02 Chugai Seiyaku Kabushiki Kaisha Remede contre des maladies provoquees par la production d'il-6
US6485943B2 (en) 1995-01-17 2002-11-26 The University Of Chicago Method for altering antibody light chain interactions
KR20050085971A (ko) 1995-04-27 2005-08-29 아브게닉스, 인크. 면역화된 제노마우스 유래의 인간 항체
AU2466895A (en) 1995-04-28 1996-11-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5830478A (en) 1995-06-07 1998-11-03 Boston Biomedical Research Institute Method for delivering functional domains of diphtheria toxin to a cellular target
US5783186A (en) 1995-12-05 1998-07-21 Amgen Inc. Antibody-induced apoptosis
CA2249195A1 (en) 1996-03-18 1997-09-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
AU720232B2 (en) 1996-07-19 2000-05-25 Amgen, Inc. Analogs of cationic proteins
US5990286A (en) 1996-12-18 1999-11-23 Techniclone, Inc. Antibodies with reduced net positive charge
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US20070059302A1 (en) 1997-04-07 2007-03-15 Genentech, Inc. Anti-vegf antibodies
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
ATE223229T1 (de) 1997-04-17 2002-09-15 Amgen Inc Zusammensetzungen aus konjugaten des stabilen, aktiven, menschlichen ob proteins mit der fc kette von immunoglobulinen und damit zusammenhängende verfahren
US7052873B2 (en) 1997-10-03 2006-05-30 Chugai Seiyaku Kabushiki Kaisha Natural human antibody
IL138801A0 (en) 1998-04-03 2001-10-31 Chugai Pharmaceutical Co Ltd Humanized antibody against human tissue factor and process for the preparation thereof
GB9809951D0 (en) * 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
CA2341029A1 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
US6475718B2 (en) 1998-09-08 2002-11-05 Schering Aktiengesellschaft Methods and compositions for modulating the interaction between the APJ receptor and the HIV virus
RU2236222C2 (ru) * 1998-09-11 2004-09-20 Айлексус Пти Лимитед Модуляторы fc-рецептора и их применение
EP1135415B1 (en) 1998-12-01 2010-08-11 Facet Biotech Corporation Humanized antibodies to gamma-interferon
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
ES2694002T3 (es) * 1999-01-15 2018-12-17 Genentech, Inc. Polipéptido que comprende una región Fc de IgG1 humana variante
US6972125B2 (en) 1999-02-12 2005-12-06 Genetics Institute, Llc Humanized immunoglobulin reactive with B7-2 and methods of treatment therewith
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
SE9903895D0 (sv) 1999-10-28 1999-10-28 Active Biotech Ab Novel compounds
DE60115613T2 (de) 2000-03-22 2006-08-24 Octagene Gmbh Herstellung von rekombinanten muteine des blutgerinnungsfaktors viii in humanen zellinien
FR2807767B1 (fr) 2000-04-12 2005-01-14 Lab Francais Du Fractionnement Anticorps monoclonaux anti-d
US20020034537A1 (en) 2000-05-03 2002-03-21 Brita Schulze Cationic diagnostic, imaging and therapeutic agents associated with activated vascular sites
EP2281843B1 (en) 2000-06-16 2016-10-12 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to BLyS
WO2002020565A2 (en) 2000-09-08 2002-03-14 Universität Zürich Collections of repeat proteins comprising repeat modules
JP2004526419A (ja) 2000-10-16 2004-09-02 フィロス インク. 抗体模倣物および他の結合タンパク質のためのタンパク質骨格
DK1355919T3 (da) 2000-12-12 2011-03-14 Medimmune Llc Molekyler med længere halveringstider, sammensætninger og anvendelser deraf
US20040001822A1 (en) 2000-12-29 2004-01-01 Avigdor Levanon Y1-isolated molecules comprising epitopes containing sulfated moieties, antibodies to such epitopes, and uses thereof
US20040002450A1 (en) 2000-12-29 2004-01-01 Janette Lazarovits Y17 - isolated molecules comprising epitopes containing sulfated moieties, antibodies to such epitopes, and uses thereof
US20040001839A1 (en) 2000-12-29 2004-01-01 Avigdor Levanon Multimers - isolated molecules comprising epitopes containing sulfated moieties, antibodies to such epitopes, and uses thereof
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
CN1561345B (zh) 2001-04-13 2011-09-14 比奥根艾迪克Ma公司 抗vla-1的抗体
US7667004B2 (en) 2001-04-17 2010-02-23 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US20030157561A1 (en) 2001-11-19 2003-08-21 Kolkman Joost A. Combinatorial libraries of monomer domains
CA2451493C (en) 2001-06-22 2016-08-23 Chugai Seiyaku Kabushiki Kaisha Cell growth inhibitors containing anti-glypican 3 antibody
DK1399484T3 (da) 2001-06-28 2010-11-08 Domantis Ltd Dobbelt-specifik ligand og anvendelse af denne
US20040161741A1 (en) 2001-06-30 2004-08-19 Elazar Rabani Novel compositions and processes for analyte detection, quantification and amplification
US20030049203A1 (en) 2001-08-31 2003-03-13 Elmaleh David R. Targeted nucleic acid constructs and uses related thereto
ES2624547T3 (es) 2001-11-14 2017-07-14 Janssen Biotech, Inc. Anticuerpos anti il 6, composiciones, métodos y usos
EP3960855A1 (en) 2001-12-28 2022-03-02 Chugai Seiyaku Kabushiki Kaisha Method for stabilizing proteins
WO2003068801A2 (en) 2002-02-11 2003-08-21 Genentech, Inc. Antibody variants with faster antigen association rates
AR038568A1 (es) 2002-02-20 2005-01-19 Hoffmann La Roche Anticuerpos anti-a beta y su uso
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US8188231B2 (en) * 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US20080199471A1 (en) 2002-03-01 2008-08-21 Bernett Matthew J Optimized cd40 antibodies and methods of using the same
WO2003074679A2 (en) 2002-03-01 2003-09-12 Xencor Antibody optimization
US20090042291A1 (en) 2002-03-01 2009-02-12 Xencor, Inc. Optimized Fc variants
DK1512015T3 (da) 2002-06-12 2009-07-06 Genencor Int Fremgangsmåder til forbedring af bindingsegenskaber hos et molekyle
US20060141456A1 (en) 2002-06-12 2006-06-29 Cynthia Edwards Methods and compositions for milieu-dependent binding of a targeted agent to a target
ITMI20021527A1 (it) 2002-07-11 2004-01-12 Consiglio Nazionale Ricerche Anticorpi anti componente c5 del complemento e loro uso
US8193318B2 (en) * 2002-08-14 2012-06-05 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
WO2004016740A2 (en) 2002-08-15 2004-02-26 Epitomics, Inc. Humanized rabbit antibodies
DK2345671T3 (en) * 2002-09-27 2016-02-15 Xencor Inc Optimized Fc variants and methods for their formation
RU2390527C2 (ru) 2002-09-27 2010-05-27 Ксенкор, Инк. АНТИТЕЛО, СОДЕРЖАЩЕЕ Fc-ВАРИАНТНУЮ ЧАСТЬ (ВАРИАНТЫ), ФАРМАЦЕВТИЧЕСКАЯ КОМПОЗИЦИЯ, СОДЕРЖАЩАЯ АНТИТЕЛО, И СПОСОБ ЛЕЧЕНИЯ МЛЕКОПИТАЮЩЕГО
US7217797B2 (en) 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7361740B2 (en) 2002-10-15 2008-04-22 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
AU2003286467B2 (en) 2002-10-15 2009-10-01 Abbvie Biotherapeutics Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US7217798B2 (en) 2003-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of Fc-fusion protein serum half-lives by mutagenesis
WO2004034988A2 (en) 2002-10-16 2004-04-29 Amgen Inc. Human anti-ifn-ϝ neutralizing antibodies as selective ifn-ϝ pathway inhibitors
CA2511910A1 (en) 2002-12-27 2004-07-15 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
CA2515081A1 (en) 2003-02-07 2004-08-19 Protein Design Labs, Inc. Amphiregulin antibodies and their use to treat cancer and psoriasis
DK1601697T3 (da) 2003-02-28 2007-10-01 Lonza Biologics Plc Oprensning af antistof ved protein A- og ionbytningskromatografi
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
GB2401040A (en) 2003-04-28 2004-11-03 Chugai Pharmaceutical Co Ltd Method for treating interleukin-6 related diseases
CA2766627C (en) 2003-05-02 2019-12-03 Xencor, Inc. Optimized fc variants and methods for their generation
EP1629001A2 (en) 2003-06-05 2006-03-01 Genentech, Inc. Blys antagonists and uses thereof
US20050152894A1 (en) 2003-09-05 2005-07-14 Genentech, Inc. Antibodies with altered effector functions
JP2005101105A (ja) 2003-09-22 2005-04-14 Canon Inc 位置決め装置、露光装置、デバイス製造方法
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
AU2003271174A1 (en) 2003-10-10 2005-04-27 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
AU2004284090A1 (en) 2003-10-24 2005-05-06 Avidia, Inc. LDL receptor class A and EGF domain monomers and multimers
PT1689777E (pt) 2003-11-05 2007-05-31 Ares Trading Sa Processo para a purificação da proteína de ligação a il-18
EP1697415A1 (en) 2003-11-12 2006-09-06 Biogen Idec MA Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
US20050142133A1 (en) 2003-12-03 2005-06-30 Xencor, Inc. Optimized proteins that target the epidermal growth factor receptor
PT1711207E (pt) 2003-12-10 2013-02-13 Medarex Inc Anticorpos alfa interferão e seus usos
WO2005077981A2 (en) 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
AP2006003689A0 (en) 2004-01-09 2006-08-31 Pfizer Antibodies to MAdCAM
BRPI0506771A (pt) 2004-01-12 2007-05-22 Applied Molecular Evolution anticorpo, e, composição farmacêutica
BRPI0506679A (pt) 2004-02-11 2007-05-15 Warner Lambert Co métodos de tratar osteoartrite com antagonistas de il-6
WO2005092925A2 (en) 2004-03-24 2005-10-06 Xencor, Inc. Immunoglobulin variants outside the fc region
US20050260711A1 (en) 2004-03-30 2005-11-24 Deepshikha Datta Modulating pH-sensitive binding using non-natural amino acids
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
WO2005112564A2 (en) 2004-04-15 2005-12-01 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Germline and sequence variants of humanized antibodies and methods of making and using them
JP5367982B2 (ja) * 2004-04-16 2013-12-11 マクロジェニクス,インコーポレーテッド FcγRIIB特異的抗体とその利用法
KR100620554B1 (ko) 2004-06-05 2006-09-06 한국생명공학연구원 Tag-72에 대한 인간화 항체
AR049390A1 (es) 2004-06-09 2006-07-26 Wyeth Corp Anticuerpos contra la interleuquina-13 humana y usos de los mismos
MXPA06014684A (es) 2004-06-18 2007-02-12 Ambrx Inc Novedosos polipeptidos de enlace antigeno y sus usos.
CN101163498B (zh) 2004-06-18 2011-10-05 诺华疫苗和诊断公司 用于诊断汉他病毒感染的方法和试剂
US20060019342A1 (en) 2004-06-25 2006-01-26 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
CN101987870B (zh) 2004-07-15 2013-07-03 赞科股份有限公司 优化的Fc变体
CA2574062A1 (en) 2004-07-20 2006-01-26 Symphogen A/S Anti-rhesus d recombinant polyclonal antibody and methods of manufacture
CA2573192C (en) 2004-08-04 2013-09-10 Applied Molecular Evolution Inc. Variant fc regions
CA2576405A1 (en) 2004-08-11 2006-02-16 Mitsubishi Chemical Corporation Antibody and utilization of the same
CA2577370A1 (en) 2004-08-16 2006-03-02 Medimmune, Inc. Integrin antagonists with enhanced antibody dependent cell-mediated cytotoxicity activity
BRPI0515230A (pt) 2004-08-19 2008-07-15 Genentech Inc polipeptìdeos, anticorpos e ácidos nucléicos isolados, composições, vetor de expressão, células hospedeiras isoladas, método para a produção de um anticorpo, artigos manufaturados, métodos de tratamento e para o alìvio de disfunções, métodos de produção e de seleção de um polipeptìdeo, anticorpo de ligação cd20 humanizado, anticorpo anti-her2 isolado e usos de um anticorpo
MX2007002883A (es) 2004-09-13 2007-06-15 Macrogenics Inc Anticuerpos humanizados contra el virus de nilo occidental y usos terapeuticos y profilacticos del mismo.
EP1799718A1 (en) 2004-09-14 2007-06-27 National Institute for Biological Standards and Control (NIBSC) Vaccine
US7563443B2 (en) 2004-09-17 2009-07-21 Domantis Limited Monovalent anti-CD40L antibody polypeptides and compositions thereof
EP1812068A4 (en) 2004-10-29 2010-06-09 Medimmune Inc METHODS FOR PREVENTING AND TREATING RSV INFECTIONS AND ASSOCIATED DISEASES
US7462697B2 (en) 2004-11-08 2008-12-09 Epitomics, Inc. Methods for antibody engineering
AU2005335714B2 (en) 2004-11-10 2012-07-26 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
CN101098890B (zh) 2004-11-12 2012-07-18 赞科股份有限公司 对FcRn的结合被改变的Fc变体
EP2845865A1 (en) 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
WO2006067847A1 (ja) 2004-12-22 2006-06-29 Chugai Seiyaku Kabushiki Kaisha フコーストランスポーターの機能が阻害された細胞を用いた抗体の作製方法
EP1831243B1 (en) 2004-12-23 2010-03-17 Novo Nordisk A/S Antibody binding affinity ligands
EP1858925A2 (en) * 2005-01-12 2007-11-28 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
GB0502358D0 (en) 2005-02-04 2005-03-16 Novartis Ag Organic compounds
EP3623473A1 (en) 2005-03-31 2020-03-18 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
EP1871808A2 (en) 2005-03-31 2008-01-02 Xencor, Inc. Fc VARIANTS WITH OPTIMIZED PROPERTIES
WO2006109592A1 (ja) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha 血液凝固第viii因子の機能代替抗体
ES2545769T3 (es) 2005-04-18 2015-09-15 Amgen Research (Munich) Gmbh Anticuerpos neutralizantes del factor estimulante de colonias de granulocitos y macrófagos humano
JP5255435B2 (ja) 2005-04-26 2013-08-07 メディミューン,エルエルシー ヒンジドメイン操作による抗体エフェクター機能の調節
JP5047947B2 (ja) 2005-05-05 2012-10-10 デューク ユニバーシティ 自己免疫疾患のための抗cd19抗体治療
WO2006130834A2 (en) 2005-05-31 2006-12-07 Board Of Regents, The University Of Texas System IGGl ANTIBODIES WITH MUTATED FC PORTION FOR INCREASED BINDING TO FCRN RECEPTOR AND USES THEREOF
FR2888850B1 (fr) 2005-07-22 2013-01-11 Pf Medicament Nouveaux anticorps anti-igf-ir et leurs applications
ES2526811T3 (es) 2005-08-10 2015-01-15 Macrogenics, Inc. Identificación y modificación de anticuerpos con regiones Fc variantes y métodos de uso de estos
CN101379086B (zh) 2005-08-19 2013-07-17 惠氏公司 抗gdf-8的拮抗剂抗体以及在als和其他gdf-8-相关病症治疗中的用途
US20070190063A1 (en) 2005-08-19 2007-08-16 Bahjat Keith S Antibody-mediated enhancement of immune response
DK1931709T3 (en) 2005-10-03 2017-03-13 Xencor Inc FC VARIETIES WITH OPTIMIZED FC RECEPTOR BINDING PROPERTIES
JP2009511067A (ja) * 2005-10-14 2009-03-19 メディミューン,エルエルシー 抗体ライブラリーの細胞提示
EP2465870A1 (en) 2005-11-07 2012-06-20 Genentech, Inc. Binding polypeptides with diversified and consensus VH/VL hypervariable sequences
WO2007060411A1 (en) 2005-11-24 2007-05-31 Ucb Pharma S.A. Anti-tnf alpha antibodies which selectively inhibit tnf alpha signalling through the p55r
EP1959997A4 (en) 2005-11-28 2009-12-23 Medimmune Llc ANTAGONISTS OF HMGB1 AND / OR RAGE AND METHOD OF USE THEREOF
ES2517420T3 (es) 2005-12-29 2014-11-03 Janssen Biotech, Inc. Anticuerpos anti-IL-23 humanos, composiciones, procedimientos y usos
US9670269B2 (en) 2006-03-31 2017-06-06 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
JP5624276B2 (ja) 2006-03-31 2014-11-12 中外製薬株式会社 抗体の血中動態を制御する方法
CA2911000A1 (en) 2006-04-05 2007-10-18 Min W. Wan Antibody purification
TWI395754B (zh) 2006-04-24 2013-05-11 Amgen Inc 人類化之c-kit抗體
US7786270B2 (en) 2006-05-26 2010-08-31 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
KR101557603B1 (ko) 2006-06-08 2015-10-06 추가이 세이야쿠 가부시키가이샤 염증성 질환의 예방 또는 치료제
CA2656224C (en) 2006-06-26 2018-01-09 Macrogenics, Inc. Combination of fc.gamma.riib antibodies and cd20-specific antibodies and methods of use thereof
ES2457072T3 (es) 2006-08-14 2014-04-24 Xencor, Inc. Anticuerpos optimizados que seleccionan como diana CD19
EP3424950A1 (en) 2006-10-06 2019-01-09 The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services Prevention of tissue ischemia, related methods and compositions
US20100034194A1 (en) 2006-10-11 2010-02-11 Siemens Communications Inc. Eliminating unreachable subscribers in voice-over-ip networks
WO2008140603A2 (en) 2006-12-08 2008-11-20 Macrogenics, Inc. METHODS FOR THE TREATMENT OF DISEASE USING IMMUNOGLOBULINS HAVING FC REGIONS WITH ALTERED AFFINITIES FOR FCγR ACTIVATING AND FCγR INHIBITING
WO2008091954A2 (en) 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
WO2008092117A2 (en) 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
EP2087111A2 (en) 2007-03-19 2009-08-12 Medimmune Limited Polypeptide variants
BRPI0811857A2 (pt) 2007-05-14 2014-10-21 Biogen Idec Inc Regiões fc (scfc) de cadeia simples, polipeptídeos de aglutinação que as compreendem e métodos relacionados.
ES2659517T3 (es) * 2007-05-30 2018-03-16 Xencor, Inc. Métodos y composiciones para inhibir células que expresan CD32B
EP3543691A1 (en) 2007-06-29 2019-09-25 Quest Diagnostics Investments Incorporated Analysis of amino acids in body fluid by liquid chromatography-mass spectrometry
JP5439176B2 (ja) 2007-07-06 2014-03-12 公益財団法人東京都医学総合研究所 Tdp−43凝集物に特異的に結合する抗体
JOP20080381B1 (ar) 2007-08-23 2023-03-28 Amgen Inc بروتينات مرتبطة بمولدات مضادات تتفاعل مع بروبروتين كونفيرتاز سيتيليزين ككسين من النوع 9 (pcsk9)
ES2566957T3 (es) 2007-09-26 2016-04-18 Chugai Seiyaku Kabushiki Kaisha Región constante de anticuerpo modificado
EP2206775B1 (en) 2007-09-26 2016-06-29 Chugai Seiyaku Kabushiki Kaisha Anti-il-6 receptor antibody
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
RU2445366C2 (ru) 2007-09-28 2012-03-20 Чугаи Сейяку Кабусики Кайся Антитело против глипикана-3 с улучшенными кинетическими показателями в плазме
KR100888133B1 (ko) * 2007-10-02 2009-03-13 에스케이에너지 주식회사 4종의 금속성분으로 구성된 다성분계 비스무스몰리브데이트 촉매 제조방법 및 상기촉매를 이용하여1,3-부타디엔을 제조하는 방법
CA2702448A1 (en) 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
WO2009062080A1 (en) 2007-11-08 2009-05-14 Pikamab, Inc. Methods for doing business using biomarkers
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
EP3575317A1 (en) * 2007-12-26 2019-12-04 Xencor, Inc. Fc variants with altered binding to fcrn
AU2008347438B2 (en) 2008-01-18 2013-08-29 Stichting Sanquin Bloedvoorziening Methods for increasing the therapeutic efficacy of immunoglobulin G class 3 (IgG3) antibodies
US8551485B2 (en) 2008-01-23 2013-10-08 Xencor, Inc. Anti-CD40 antibodies and methods of inhibiting proliferation of CD40 expressing cells
US20110183861A1 (en) 2008-01-29 2011-07-28 Ablynn N.V. Methods to stabilize proteins and polypeptides
TWI564021B (zh) 2008-04-11 2017-01-01 Chugai Pharmaceutical Co Ltd Repeated binding of antigen to antigen binding molecules
WO2009131702A2 (en) 2008-04-25 2009-10-29 Dyax Corp. Antibodies against fcrn and use thereof
AU2009246946B2 (en) 2008-05-01 2013-09-26 Amgen Inc. Anti-hepcidin antibodies and methods of use
MX2010014079A (es) 2008-06-20 2011-02-22 Novartis Ag Inmunoglobulinas con agregacion reducida.
MX356218B (es) 2008-08-05 2018-05-18 Novartis Ag Composiciones y métodos para anticuerpos que se dirigen a la proteína de complemento c5.
JP5028372B2 (ja) 2008-09-26 2012-09-19 京セラドキュメントソリューションズ株式会社 画像処理装置、画像処理方法及び画像処理プログラム
TWI440469B (zh) * 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
CN110317272A (zh) 2008-10-14 2019-10-11 霍夫曼-拉罗奇有限公司 免疫球蛋白变体及其用途
WO2010058860A1 (ja) 2008-11-18 2010-05-27 株式会社シノテスト 試料中のc反応性蛋白質の測定方法及び測定試薬
JO3672B1 (ar) 2008-12-15 2020-08-27 Regeneron Pharma أجسام مضادة بشرية عالية التفاعل الكيماوي بالنسبة لإنزيم سبتيليسين كنفرتيز بروبروتين / كيكسين نوع 9 (pcsk9).
EP3543256A1 (en) 2009-01-12 2019-09-25 Cytomx Therapeutics Inc. Modified antibody compositions, methods of making and using thereof
JP2012515556A (ja) 2009-01-23 2012-07-12 バイオジェン・アイデック・エムエイ・インコーポレイテッド 低下したエフェクタ機能を有する安定化Fcポリペプチドおよび使用方法
WO2010098863A1 (en) 2009-02-26 2010-09-02 Lpath, Inc. Humanized platelet activating factor antibody design using anti-lipid antibody templates
JP4885308B2 (ja) * 2009-03-19 2012-02-29 中外製薬株式会社 改良された抗体分子を含有する製剤
TWI682995B (zh) 2009-03-19 2020-01-21 日商中外製藥股份有限公司 抗體恆定區域改變體
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
AU2010201495B2 (en) 2009-04-16 2012-04-12 Accenture Global Services Limited Touchpoint customization system
EP2435079A4 (en) 2009-05-26 2012-11-14 Univ Johns Hopkins NEW DESMINPHOSPHORYLATION ORTE FOR DIAGNOSIS AND INFLUENCING DISEASES
EP2445936A1 (en) * 2009-06-26 2012-05-02 Regeneron Pharmaceuticals, Inc. Readily isolated bispecific antibodies with native immunoglobulin format
CA2766065C (en) 2009-06-30 2020-07-21 Research Development Foundation Immunoglobulin fc polypeptides
JP5208882B2 (ja) 2009-08-10 2013-06-12 ルネサスエレクトロニクス株式会社 表示装置用電源回路
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
NZ599148A (en) 2009-10-06 2014-08-29 Medimmune Ltd Rsv-specific binding molecule
SI2486141T1 (en) 2009-10-07 2018-06-29 Macrogenics, Inc. FC REGION-CONTAINING POLYPETHYDE, AFFECTING A BETTERED EFFECTORAL FUNCTION, BY CHANGES IN THE SCOPE OF FUKOZILATION AND METHODS FOR THEIR USE
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
CN102971342B (zh) 2010-01-28 2016-08-03 Ab生物科学公司 亲和力降低的新抗体和制备所述抗体的方法
LT2536745T (lt) 2010-02-19 2016-09-26 Xencor, Inc. Nauji ctla4-ig imunoadhezinai
JP5889181B2 (ja) 2010-03-04 2016-03-22 中外製薬株式会社 抗体定常領域改変体
EP2545079A2 (en) 2010-03-11 2013-01-16 Rinat Neuroscience Corporation ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING
JP2011184418A (ja) 2010-03-11 2011-09-22 Tokyo Institute Of Technology 親和性可変抗体
KR20220157513A (ko) 2010-03-30 2022-11-29 추가이 세이야쿠 가부시키가이샤 항원 제거를 촉진하는 FcRn에 대해 변경된 친화성을 갖는 항체
TWI667257B (zh) 2010-03-30 2019-08-01 中外製藥股份有限公司 促進抗原消失之具有經修飾的FcRn親和力之抗體
JP5234045B2 (ja) 2010-04-13 2013-07-10 Tdk株式会社 スパッタリングターゲット、及び、光メディアの製造方法
MX349622B (es) 2010-09-08 2017-08-07 Halozyme Inc Metodos para evaluar e identificar o evolucionar proteinas terapeuticas condicionalmente activas.
EP2622074B1 (en) 2010-09-30 2014-11-12 Board Of Trustees Of Northern Illinois University Library-based methods and compositions for introducing molecular switch functionality into protein affinity reagents
MX365235B (es) 2010-11-30 2019-05-28 Chugai Pharmaceutical Co Ltd Molécula de unión a antígeno capaz de unir repetidamente a la pluralidad de moléculas de antígeno.
WO2012132067A1 (ja) 2011-03-30 2012-10-04 中外製薬株式会社 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
US20140093496A1 (en) 2011-02-25 2014-04-03 Chugai Seiyaku Kabushiki Kaisha Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
CA2830254C (en) 2011-03-16 2019-09-10 Amgen Inc. Fc variants
EP3825325A3 (en) 2011-03-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
AU2012258907A1 (en) 2011-05-25 2013-11-07 Merck Sharp & Dohme Corp. Method for preparing Fc-containing polypeptides having improved properties
DK2728002T3 (da) 2011-06-30 2022-04-04 Chugai Pharmaceutical Co Ltd Heterodimeriseret polypeptid
UA117901C2 (uk) 2011-07-06 2018-10-25 Ґенмаб Б.В. Спосіб посилення ефекторної функції вихідного поліпептиду, його варіанти та їх застосування
CA2850322C (en) 2011-09-30 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule inducing immune response to target antigen
WO2013046722A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 イオン濃度依存性結合分子ライブラリ
SG11201401101XA (en) 2011-09-30 2014-08-28 Chugai Pharmaceutical Co Ltd Antigen-binding molecule for promoting loss of antigens
TW201817745A (zh) 2011-09-30 2018-05-16 日商中外製藥股份有限公司 具有促進抗原清除之FcRn結合域的治療性抗原結合分子
US20150050269A1 (en) 2011-09-30 2015-02-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
EP2765192A4 (en) 2011-10-05 2015-04-15 Chugai Pharmaceutical Co Ltd ANTIGEN BINDING MOLECULE FOR PROMOTING THE PLASMA CLAIR OF AN ANTIGEN COMPRISING A SACCHARIDIC CHAIN TYPE RECEPTOR BINDING DOMAIN
US20150056182A1 (en) 2011-11-30 2015-02-26 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
SG11201404751UA (en) 2012-02-09 2014-09-26 Chugai Pharmaceutical Co Ltd Modified fc region of antibody
TW202015731A (zh) 2012-02-24 2020-05-01 日商中外製藥股份有限公司 經FcγRIIB促進抗原消失之抗原結合分子
WO2013138681A1 (en) 2012-03-16 2013-09-19 Regeneron Pharmaceuticals, Inc. Mice that produce antigen-binding proteins with ph-dependent binding characteristics
NZ730271A (en) 2012-03-16 2022-09-30 Regeneron Pharma Non-human animals expressing ph-sensitive immunoglobulin sequences
LT2883449T (lt) 2012-03-16 2018-05-10 Regeneron Pharmaceuticals, Inc. Histidinu modifikuoti lengvosios grandinės antikūnai ir juos gaminantys genetiškai modifikuoti graužikai
TWI619729B (zh) 2012-04-02 2018-04-01 再生元醫藥公司 抗-hla-b*27抗體及其用途
DK2857419T3 (da) 2012-05-30 2021-03-29 Chugai Pharmaceutical Co Ltd Antigen-bindende molekyle til eliminering af aggregerede antigener
CA2874721A1 (en) 2012-05-30 2013-12-05 Tomoyuki Igawa Target tissue-specific antigen-binding molecule
US11142563B2 (en) 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
CN104540852B (zh) 2012-08-13 2018-10-02 瑞泽恩制药公司 具有pH-依赖性结合特性的抗-PCSK9抗体
MX371442B (es) 2012-08-24 2020-01-30 Chugai Pharmaceutical Co Ltd VARIANTE DE LA REGION FC ESPECIFICA PARA FCyRIIB.
EP3597747B1 (en) 2012-08-24 2023-03-15 Chugai Seiyaku Kabushiki Kaisha Mouse fcgammarii-specific fc antibody
WO2014104165A1 (ja) 2012-12-27 2014-07-03 中外製薬株式会社 ヘテロ二量化ポリペプチド
JO3532B1 (ar) 2013-03-13 2020-07-05 Regeneron Pharma الأجسام المضادة لمضاد انترلوكين-33 واستعمالاتها
US20160039945A1 (en) 2013-03-15 2016-02-11 Amgen Inc. Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9
WO2014144080A2 (en) 2013-03-15 2014-09-18 Amgen Inc. Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9
US9321686B2 (en) 2013-03-15 2016-04-26 Forta Corporation Reinforcement fiber coating compositions, methods of making and treating, and uses for improved adhesion to asphalt and portland cement concrete
WO2014140366A1 (en) 2013-03-15 2014-09-18 Affibody Ab New polypeptides
WO2014144577A1 (en) 2013-03-15 2014-09-18 Bayer Healthcare Llc Anti-tfpi antibody variants with differential binding across ph range for improved pharmacokinetics
EP3783017A1 (en) 2013-04-02 2021-02-24 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US10111953B2 (en) 2013-05-30 2018-10-30 Regeneron Pharmaceuticals, Inc. Methods for reducing remnant cholesterol and other lipoprotein fractions by administering an inhibitor of proprotein convertase subtilisin kexin-9 (PCSK9)
RS59003B1 (sr) 2013-09-18 2019-08-30 Regeneron Pharma Antitela sa histidinom modifikovanim lakim lancem i genetski modifikovane nehumane životinje koje ih generišu
BR112016011401A2 (pt) 2013-11-20 2017-09-26 Regeneron Pharma moduladores de aplnr e utilizações dos mesmos
NZ711451A (en) 2014-03-07 2016-05-27 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
EP3160996B1 (en) 2014-06-30 2022-06-08 Merck Patent GmbH Anti-tnfa antibodies with ph-dependent antigen binding
CN113045650A (zh) 2014-12-19 2021-06-29 中外制药株式会社 抗-c5抗体及使用方法
CA2963760A1 (en) 2014-12-19 2016-06-23 Yoshinao Ruike Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
EA201791754A1 (ru) 2015-02-05 2019-01-31 Чугаи Сейяку Кабусики Кайся АНТИТЕЛА, СОДЕРЖАЩИЕ ЗАВИСЯЩИЙ ОТ КОНЦЕНТРАЦИИ ИОНОВ АНТИГЕНСВЯЗЫВАЮЩИЙ ДОМЕН, ВАРИАНТЫ Fc-ОБЛАСТИ, IL-8-СВЯЗЫВАЮЩИЕ АНТИТЕЛА И ИХ ПРИМЕНЕНИЯ
US10233252B2 (en) 2015-12-21 2019-03-19 Wisconsin Alumni Research Foundation pH-dependent antibodies targeting the transferrin receptor and methods of use thereof to deliver a therapeutic agent
JP7211961B2 (ja) 2017-03-14 2023-01-24 ファイヴ プライム セラピューティクス インク 酸性pHでVISTAに結合する抗体
TWI788332B (zh) 2017-03-16 2023-01-01 英商梅迪繆思有限公司 抗-par2抗體及其用途

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6737056B1 (en) * 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US8802823B2 (en) * 2002-09-27 2014-08-12 Xencor, Inc. Optimized Fc variants
US20110021755A1 (en) * 2003-03-03 2011-01-27 Xencor, Inc. Optimized Fc Variants

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US9890218B2 (en) 2011-06-30 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US10253100B2 (en) 2011-09-30 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11851476B2 (en) 2011-10-31 2023-12-26 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule having regulated conjugation between heavy-chain and light-chain
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US11142563B2 (en) 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
US11267868B2 (en) * 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US20160039912A1 (en) * 2013-04-02 2016-02-11 Chugai Seiyaku Kabushiki Kaisha Fc REGION VARIANT
EP3970747A2 (en) 2014-06-03 2022-03-23 XBiotech Inc. Compositions and methods for treating and preventing staphylococcus aureus infections
WO2015187779A1 (en) 2014-06-03 2015-12-10 Xbiotech, Inc. Compositions and methods for treating and preventing staphylococcus aureus infections
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11454633B2 (en) 2014-12-19 2022-09-27 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10519229B2 (en) 2015-02-05 2019-12-31 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding IL-8 antibodies
US10526408B2 (en) 2015-08-28 2020-01-07 Research Development Foundation Engineered antibody FC variants
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11091539B2 (en) 2016-07-08 2021-08-17 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
US11248041B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Anti-ApoC3 antibodies
US11248042B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Polynucleotides encoding anti-ApoC3 antibodies
WO2022004761A1 (en) * 2020-06-30 2022-01-06 Chugai Seiyaku Kabushiki Kaisha Site specific notch-activating molecule and uses thereof
US20240025995A1 (en) * 2021-10-21 2024-01-25 Seismic Therapeutic, Inc. Dual targeted immune regulating compositions
US12030945B2 (en) 2023-10-25 2024-07-09 Seismic Therapeutic, Inc. Variant IgG Fc polypeptides and uses thereof

Also Published As

Publication number Publication date
KR20200103845A (ko) 2020-09-02
KR20230005405A (ko) 2023-01-09
CN103492565B (zh) 2021-01-29
KR102147548B1 (ko) 2020-08-24
AU2012222252B2 (en) 2016-08-25
AU2012222252A1 (en) 2013-10-10
US20140105889A1 (en) 2014-04-17
AU2016262766B2 (en) 2018-10-04
BR112013021526A2 (pt) 2016-11-01
EP2679681B2 (en) 2023-11-15
JP2017029150A (ja) 2017-02-09
EP2679681A1 (en) 2014-01-01
WO2012115241A1 (ja) 2012-08-30
SG10201609665PA (en) 2017-01-27
RU2608504C2 (ru) 2017-01-18
EP3604330A1 (en) 2020-02-05
US10618965B2 (en) 2020-04-14
JP6074360B2 (ja) 2017-02-01
JP6826620B2 (ja) 2021-02-03
CA2827923C (en) 2021-11-23
JPWO2012133782A1 (ja) 2014-07-28
EP2679681B1 (en) 2019-08-07
EP2679681A4 (en) 2015-01-28
JP2022164755A (ja) 2022-10-27
BR112013021526B1 (pt) 2021-09-21
BR112013025221A8 (pt) 2020-10-27
US20200199241A1 (en) 2020-06-25
KR20140014167A (ko) 2014-02-05
JP2019123725A (ja) 2019-07-25
JPWO2012115241A1 (ja) 2014-07-07
US11718678B2 (en) 2023-08-08
JP6032818B2 (ja) 2016-11-30
CN103492565A (zh) 2014-01-01
CA2827923A1 (en) 2012-08-30
CN112812184A (zh) 2021-05-18
MX2013009774A (es) 2013-11-21
JP2018138616A (ja) 2018-09-06
JP6348936B2 (ja) 2018-06-27
BR112013025221A2 (pt) 2020-09-08
AU2016262766A1 (en) 2016-12-15
MX352889B (es) 2017-12-13
RU2013143303A (ru) 2015-03-27
JP2021027845A (ja) 2021-02-25
US20230174655A1 (en) 2023-06-08
SG192945A1 (en) 2013-09-30

Similar Documents

Publication Publication Date Title
US20230174655A1 (en) Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
US20210261648A1 (en) FCgammaRIIB-SPECIFIC FC REGION VARIANT
JP7360242B2 (ja) マウスFcγRII特異的Fc抗体
RU2749357C2 (ru) ВАРИАНТ ПОЛИПЕПТИДА, КОТОРЫЙ ИМЕЕТ СОХРАНЕННУЮ ИЛИ СНИЖЕННУЮ АКТИВНОСТЬ СВЯЗЫВАНИЯ С FcγRIIa, СОДЕРЖАЩАЯ ЕГО ФАРМАЦЕВТИЧЕСКАЯ КОМПОЗИЦИЯ И ИХ ПРИМЕНЕНИЕ

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MIMOTO, FUTA;KURAMOCHI, TAICHI;IGAWA, TOMOYUKI;AND OTHERS;SIGNING DATES FROM 20131021 TO 20131025;REEL/FRAME:031925/0784

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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