[go: up one dir, main page]

WO2021210667A1 - Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition - Google Patents

Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition Download PDF

Info

Publication number
WO2021210667A1
WO2021210667A1 PCT/JP2021/015683 JP2021015683W WO2021210667A1 WO 2021210667 A1 WO2021210667 A1 WO 2021210667A1 JP 2021015683 W JP2021015683 W JP 2021015683W WO 2021210667 A1 WO2021210667 A1 WO 2021210667A1
Authority
WO
WIPO (PCT)
Prior art keywords
blood coagulation
coagulation factor
bleeding
binding molecule
antigen
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.)
Ceased
Application number
PCT/JP2021/015683
Other languages
French (fr)
Japanese (ja)
Inventor
光 古賀
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
Application filed by Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Priority to CN202180041295.5A priority Critical patent/CN115916836A/en
Publication of WO2021210667A1 publication Critical patent/WO2021210667A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/56Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving blood clotting factors, e.g. involving thrombin, thromboplastin, fibrinogen

Definitions

  • the present disclosure relates to bispecific antigen-binding molecules that recognize both of two specific blood coagulation-related factors, as well as compositions containing them, their use in the manufacture of compositions, and kits. ..
  • the present disclosure also relates to a method for promoting blood coagulation using the bispecific antigen-binding molecule.
  • the present disclosure relates to methods of screening for substances that are effective in the prevention and / or treatment of bleeding, bleeding-related diseases, or diseases caused by bleeding.
  • Blood coagulation consists of a series of components, especially fibrinogen, blood coagulation factor II, factor V, factor VII, factor VIII, factor IX, factor X, factor XI, factor XII (F.II, respectively).
  • Hemophilia is a bleeding disease in which blood coagulation is caused by deficiency of these components or inhibition of their functionality. If it is caused by F.VIII, it is called hemophilia A, and if it is caused by F.IX, it is called hemophilia B. In patients with hemophilia, bleeding symptoms are seen in deep tissues such as joints and muscles, and in severe cases, intracranial hemorrhage also occurs.
  • the severity of hemophilia is classified based on F.VIII activity or F.IX activity in the blood. Specifically, assuming that the F.VIII activity or F.IX activity of a healthy person is 100%, the activity is severe in patients with an activity of less than 1%, and moderate in patients with an activity of 1% or more and less than 5%. Patients with 5% or more and less than 40% are classified as mild. Patients with severe hemophilia present significantly more frequent bleeding symptoms than patients with moderate and mild illness.
  • F.VIII or F.IX replacement therapy can dramatically reduce the frequency of bleeding by maintaining F.VIII or F.IX activity in the patient's blood at 1% or higher. ..
  • coagulation factor preparations purified from plasma or prepared by genetic recombination technology are mainly used.
  • F.VIII preparation For bleeding in hemophilia A patients, F.VIII preparation is usually administered (on-demand administration). In recent years, in order to prevent bleeding events, F.VIII preparations are prophylactically administered (Non-Patent Documents 1 and 2) (preventive administration). The half-life of the F.VIII product in blood is about 12 to 16 hours. Therefore, for continuous prophylaxis, the F.VIII preparation is administered to the patient three times a week (Non-Patent Documents 3 and 4). In addition, in on-demand administration, in order to prevent rebleeding, the F.VIII preparation is additionally administered at regular intervals as needed. In addition, administration of F.VIII preparation is performed intravenously.
  • Bispecific antibodies that substitute for the function of F.VIII and their use have been reported as means for solving these problems (Patent Documents 1, 2, 3 and 4).
  • Bispecific antibodies against F.IXa and FX exert F.VIII cofactor function substitution activity and can substitute F.VIII function by positioning both factors in the vicinity (non-F.VIII cofactor function substitution activity).
  • Patent Document 5 One of the antibodies, ACE910 (Emicizumab), which has high F.VIII cofactor function substitution activity, has been confirmed to have excellent pharmacokinetics (long half-life) and tolerability in clinical trials in healthy subjects.
  • Non-Patent Document 6 in clinical trials in patients with hemophilia A who do not have or carry inhibitors, the number of bleedings significantly increased by administration of ACE910 (Emicizumab) compared to before administration of ACE910 (Emicizumab). Suppression was observed (Non-Patent Document 7).
  • F.IX preparation For bleeding in hemophilia B patients, F.IX preparation is regularly administered.
  • the conventional F.IX preparation having a standard half-life requires intravenous administration twice a week, and frequent administration is a burden on patients and their families.
  • anti-F.IX antibodies inhibitors
  • hemostasis management is often difficult.
  • patients with F.IX inhibitors exhibit anaphylactic symptoms as a characteristic side reaction not seen in F.VIII inhibitors, which is a problem (Non-Patent Document 8). Therefore, there has been a strong demand for a drug that is less burdensome to administer than the F.IX preparation and is not affected by the presence of the inhibitor.
  • F.XI is a protease precursor of F.XIa and contributes to hemostasis through activation of F.IX.
  • the activation peptide of F.X can be cleaved by adding a high concentration of F.XIa to F.X, and that F.Va is produced by adding it to F.V (Non-Patent Document 9).
  • the duration of bleeding was reduced by intravenous administration of an excessive amount of F.XIa (estimated plasma concentration of 60 nM) in hemophilia B model mice genetically deficient in F.IX. (Non-Patent Document 10).
  • the invention in the present disclosure has been made in view of the above circumstances and is used in a non-limiting aspect for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • the purpose is to provide new molecules.
  • the present inventors recognize two blood coagulation-related factors in which cofactors that are close to each other in vivo have not been confirmed as a result of diligent research. The production was successful. We also found that the bispecific antigen-binding molecule contributes to the promotion of blood coagulation.
  • a bispecific antigen-binding molecule that recognizes both an enzyme and a substrate that can undergo a catalytic reaction of the enzyme but a bispecific antigen-binding molecule that substitutes for the function of a peptidic cofactor or heparin expressed in a living body. Excludes molecules.
  • a bispecific antigen-binding molecule according to [A] which is a bispecific antigen-binding molecule that recognizes both an enzyme and a substrate capable of undergoing a catalytic reaction of the enzyme, and promotes hydrolysis of the substrate by the enzyme. Sex antigen binding molecule.
  • the bispecific antigen-binding molecule according to any one of (a) to (h) below: (a) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X; (b) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor X and blood coagulation factor X; (c) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X; (d) Activated blood coagulation factor VII-A bispecific antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X; (e) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor X; (f) A bispecific antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X; (g) A bispecific antigen-binding molecule that recognizes both
  • [1a] A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
  • [3] The bispecific antigen-binding molecule according to [1] or [1a], [2] or [2a], which is a bispecific antibody.
  • a composition comprising the antigen-binding molecule according to any one of [1] to [3] and a pharmaceutically acceptable carrier.
  • the composition according to [4] which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX.
  • composition according to [6] wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
  • Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX.
  • the composition according to [6] which is a disease in which an inhibitor appears.
  • Bleeding a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease.
  • composition according to [12], wherein the disease that develops and / or progresses due to a decrease or deficiency of the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
  • the composition according to [12] which is a disease in which an inhibitor appears.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding which comprises at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4].
  • a kit for use in methods of preventing and / or treating [17] Bleeding or bleeding containing at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4] and containing blood coagulation factor IX.
  • a screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
  • a quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
  • [26] A nucleic acid encoding the bispecific antigen-binding molecule according to any one of [1] to [3].
  • [27] A vector into which the nucleic acid according to [26] has been inserted.
  • [28] A cell containing the nucleic acid according to [26] or the vector according to [27].
  • the bispecific antigen-binding molecule according to any one of (i) to (o) below: (i) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XI and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X; (j) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor X and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X; (k) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X; (l) Activated blood coagulation factor VII-A bispecific antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X, and is a bispecific antigen
  • Binding molecule (m) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X; (n) A bispecific antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X and activates blood coagulation factor X; or (o) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor IX, and is a bispecific antigen-binding molecule that activates blood coagulation factor IX.
  • [30a] A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X.
  • a composition comprising the antigen-binding molecule according to [30], [30a], or [31] and a pharmaceutically acceptable carrier.
  • the composition according to [32] which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX.
  • the composition according to [34], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
  • Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease.
  • Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • the composition according to [37] which is a disease in which an inhibitor appears.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI.
  • the composition according to [40], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
  • Bleeding which comprises at least the bispecific antigen-binding molecule according to [30], [30a], or [31], or the composition according to [32], and contains blood coagulation factor IX.
  • Bleeding, bleeding comprising at least the bispecific antigen binding molecule according to [30], [30a] or [31], or the composition according to [32], and containing blood coagulation factor XI.
  • a method for promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X [50] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding. A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X. [51] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [50], which further comprises the following steps.
  • a quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding includes (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
  • the quality test method according to [52] further comprising the following steps: (3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
  • the nucleic acid encoding the bispecific antigen-binding molecule according to [30], [30a] or [31].
  • [55] A vector into which the nucleic acid according to [54] has been inserted.
  • [56] A cell containing the nucleic acid according to [54] or the vector according to [55].
  • [57] The method for producing the bispecific antigen-binding molecule according to [30], [30a], or [31] by culturing the cells according to [56].
  • [58] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the step of administering the composition according to any one of [32] to [42].
  • the bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A prophylactic and / or therapeutic agent for bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the composition according to any one of [32] to [42].
  • the antigen-binding molecule according to any one of (a) to (h) below: (a) Antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X; (b) Antigen-binding molecule that recognizes both activated blood coagulation factor X and blood coagulation factor X; (c) Antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X; (d) Activated blood coagulation factor VII-An antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X; (e) Antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor X; (f) Antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X; (g) Antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor I
  • [2b] The antigen-binding molecule according to [1b], which promotes activation of the substrate by the enzyme.
  • [2c] The antigen-binding molecule according to [1b], which has an absorbance of 0.1 or more 30 minutes after the addition of the color-developing substrate solution in an in vitro enzyme reaction measurement system.
  • [3b] The antigen-binding molecule according to [1b], [2b] or [2c], which is an antibody.
  • [4b] A composition comprising the antigen-binding molecule according to any one of [1b] to [3b] and a pharmaceutically acceptable carrier.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX.
  • [7b] The composition according to [6b], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
  • [8b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX.
  • the composition according to [6b] which is a disease in which an inhibitor appears.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease.
  • Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • the composition according to [9b] which is a disease in which an inhibitor appears.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI.
  • the composition according to [12b] which is a disease in which an inhibitor appears.
  • [15b] Use of any of the antigen-binding molecules of [1b] to [3b] for producing the composition according to any one of [4b] to [14b].
  • [16b] Preventing and / or bleeding, bleeding-related diseases, or diseases caused by bleeding, which contain at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b]. Or a kit for use in therapeutic methods.
  • [17b] Bleeding, a disease associated with bleeding, or a disease containing blood coagulation factor IX and containing at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b].
  • [18b] Bleeding, a disease associated with bleeding, or a disease containing at least the antigen-binding molecule according to any one of [1b] to [3b], or the composition according to [4b], and containing blood coagulation factor VIII.
  • [21b] A method for promoting blood coagulation using an antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
  • [22b] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • a screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
  • [23b] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [22b], which further comprises the following steps.
  • a quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
  • the quality test method according to [24b] further comprising the following steps: (3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
  • a nucleic acid encoding the antigen-binding molecule according to any one of [1b] to [3b].
  • [27b] A vector into which the nucleic acid according to [26b] has been inserted.
  • [28b] A cell containing the nucleic acid according to [26b] or the vector according to [27b].
  • [29b] A method for producing the antigen-binding molecule according to any one of [1b] to [3b] by culturing the cells according to [28b].
  • the antigen-binding molecule according to any one of (i) to (o) below: (i) Activated antigen-binding molecule that recognizes both blood coagulation factor XI and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X; (j) Activated antigen-binding molecule that recognizes both blood coagulation factor X and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X; (k) Activated antigen-binding molecule that recognizes both blood coagulation factor VII and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X; (l) Activated blood coagulation factor VII-An antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X; (m) Activated
  • the composition according to [32b] which is a pharmaceutical composition used for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX.
  • Bleeding a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease.
  • [39b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII.
  • Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI.
  • composition according to [40b] wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
  • Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI and / or activation are associated with blood coagulation factor XI and / or activated blood coagulation factor XI.
  • the composition according to [40b] which is a disease in which an inhibitor appears.
  • [44b] Prevents and / or treats bleeding, bleeding-related diseases, or diseases caused by bleeding, which comprises at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b].
  • [45b] Due to bleeding, bleeding-related disease, or bleeding, comprising at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b], and containing blood coagulation factor IX.
  • [46b] Due to bleeding, bleeding-related disease, or bleeding, comprising at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b], and containing blood coagulation factor VIII.
  • [47b] Caused by bleeding, a disease associated with bleeding, or bleeding, which comprises at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b] and contains blood coagulation factor XI.
  • a bleeding, a disease associated with bleeding, or a disease caused by bleeding which comprises at least the antigen-binding molecule described in [30b] or [31b] or the composition described in [32b], is used in combination with a bypass preparation. And a kit for use in prophylactic and / or therapeutic methods.
  • a screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
  • a quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
  • [54b] A nucleic acid encoding the antigen-binding molecule according to [30b] or [31b].
  • [55b] A vector into which the nucleic acid according to [54b] has been inserted.
  • [56b] A cell containing the nucleic acid according to [54b] or the vector according to [55b].
  • [57b] A method for producing the antigen-binding molecule according to [30b] or [31b] by culturing the cells according to [56b].
  • [58b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b].
  • a method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding which comprises the step of administering the composition of.
  • the binding molecule or the composition according to any one of [4b] to [14b] and [32b] to [42b].
  • [62b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b].
  • a method of promoting blood coagulation which comprises the step of administering the composition of.
  • [63b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b] for use in promoting blood coagulation, or [4b] to [14b] and [32b].
  • [64b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b] in the production of a blood coagulation promoter, or [4b] to [14b] and [32b] to. Use of the composition according to any one of [42b].
  • [65b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b].
  • a blood coagulation promoter which comprises the composition of.
  • the bispecific antigen-binding molecules, compositions, and kits of the present disclosure contribute to the promotion of blood coagulation and thus prevent bleeding, bleeding-related diseases, or diseases caused by bleeding. And / or may be useful for treatment.
  • Enzyme A and substrate B which are related so that peptide cofactors do not exist in the living body, hardly proceed with the reaction as they are (upper figure).
  • a bispecific antigen-binding molecule that recognizes the enzyme A and the substrate B, it is possible to promote an enzymatic reaction such as a hydrolysis reaction of the substrate B by the enzyme A (see the figure below).
  • a schematic diagram (intrinsic system / extrinsic system) of the blood coagulation cascade is shown.
  • the measurement results of the F.X activation promoting activity of anti-F.XIa and F.X bispecific antibodies by F.XIa in the in vitro enzyme reaction system are shown.
  • the prepared anti-F.XIa and F.X bispecific antibodies showed F.X activation promoting activity by F.XIa.
  • the coagulation time (APTT) in F.IX-deficient plasma is shown.
  • the prepared anti-F.XIa and F.X bispecific antibodies showed the coagulation-promoting activity of the plasma.
  • the measurement results of the F.X activation promoting activity of anti-F.XIa and F.X bispecific antibodies by F.XIa in the in vitro enzyme reaction system are shown.
  • the prepared anti-F.XIa and F.X bispecific antibodies showed F.X activation promoting activity by F.XIa.
  • the coagulation time (APTT) in F.IX-deficient plasma is shown.
  • the prepared anti-F.XIa and F.X bispecific antibodies showed the coagulation-promoting activity of the plasma.
  • antigen-binding molecule refers to a molecule that specifically binds to an antigenic determinant (epitope) in its broadest sense.
  • the antigen binding molecule is an antibody, antibody fragment, or antibody derivative.
  • the antigen-binding molecule is a non-antibody protein, or a fragment thereof, or a derivative thereof.
  • the bispecific antigen-binding molecule in the present invention is an antigen-binding molecule containing two types of antigen-binding domains having specificity for different antigens or epitopes.
  • the bispecific antigen-binding molecule in the present invention is a bispecific antibody.
  • the bispecific antibody is not particularly limited, but is preferably a monoclonal antibody.
  • an antigen binding domain refers to a region that specifically binds to and is complementary to a part or all of an antigen.
  • an antigen-binding molecule comprises an antigen-binding domain. When the molecular weight of an antigen is large, the antigen-binding domain can bind only to a specific portion of the antigen. The specific portion is called an epitope.
  • the antigen binding domain comprises an antibody fragment that binds to a particular antigen.
  • Antigen binding domains can be provided by variable domains of one or more antibodies.
  • the antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH).
  • antigen binding domains examples include “scFv (single chain Fv)", “single chain antibody”, “Fv”, “scFv2 (single chain Fv 2)", “Fab” or “Fab' , Etc. can be mentioned.
  • the antigen binding domain comprises a non-antibody protein or fragment thereof that binds to a particular antigen.
  • the antigen binding domain comprises a hinge region.
  • the term "specifically bound” means a state in which one molecule of a specifically bound molecule does not show any significant binding to a molecule other than the other molecule to which one or more of the specifically bound molecules are bound. It means to combine with. It is also used when the antigen-binding domain is specific for a specific epitope among a plurality of epitopes contained in a certain antigen. When the epitope to which the antigen-binding domain binds is contained in a plurality of different antigens, the antigen-binding molecule having the antigen-binding domain can bind to various antigens including the epitope.
  • antibody is used in the broadest sense, and is not limited to, but is not limited to, a monoclonal antibody, a polyclonal antibody, and a multispecific antibody (for example, as long as it exhibits a desired antigen-binding activity). Includes a variety of antibody structures, including bispecific antibodies), antibody fragments and antibody modifications.
  • Binding activity is the sum of non-covalent interactions between one or more binding sites of a molecule (eg, an antibody) and a molecule's binding partner (eg, an antigen). It refers to the strength of.
  • the binding activity is not strictly limited to a 1: 1 interaction between members of a binding pair (eg, antibody and antigen).
  • the binding activity refers to a unique binding affinity (“affinity”). If the members of the binding pair are capable of both monovalent and multivalent binding, the binding activity is the sum of these binding forces.
  • binding activity of the molecule X to its partner Y can generally be expressed by the dissociation constant (KD) or the "analyte binding amount per unit ligand amount". Binding activity can be measured by conventional methods known in the art, including those described herein.
  • mutant antibodies eg, mutant antibodies that contain naturally occurring mutations, or mutant antibodies that occur during the production of monoclonal antibody preparations, such variants are usually few. Except for the amount present), it binds to the same and / or the same epitope.
  • polyclonal antibody preparations which typically contain different antibodies against different determinants (epitopes)
  • each monoclonal antibody in the monoclonal antibody preparation is for a single determinant on the antigen.
  • the modifier "monoclonal” should not be construed as requiring the production of an antibody by any particular method, indicating the characteristic of the antibody that it is obtained from a substantially homogeneous population of antibodies.
  • the monoclonal antibody used according to the present invention is not limited to these, but is a hybridoma method, a recombinant DNA method, a phage display method, and a transgenic animal containing all or a part of the human immunoglobulin locus. It may be made by a variety of methods, including methods that utilize, such methods and other exemplary methods for making monoclonal antibodies are described herein.
  • Natural antibody refers to an immunoglobulin molecule with various naturally occurring structures.
  • a native IgG antibody is a heterotetrameric glycoprotein of approximately 150,000 daltons composed of two identical disulfide-bonded light chains and two identical heavy chains.
  • VH variable region
  • CH2, and CH3 constant domains
  • each light chain has a variable region (VL), also called a variable light chain domain or a light chain variable domain, followed by a stationary light chain (CL) domain.
  • VH variable region
  • VL variable region
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • chimeric antibody is one in which a portion of the heavy chain and / or light chain is derived from a particular source or species, while the rest of the heavy chain and / or light chain is derived from a different source or species. It refers to an antibody.
  • the "class" of an antibody refers to the type of constant domain or constant region in the heavy chain of an antibody.
  • Heavy chain constant domains corresponding to different classes of immunoglobulins are referred to as ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • the constant region as one embodiment of the present invention is preferably an antibody constant region, more preferably an IgG1, IgG2, IgG3, IgG4 type antibody constant region, and even more preferably a human IgG1, IgG2, IgG3. , IgG4 type antibody constant region.
  • the constant region as another embodiment of the present invention is preferably a heavy chain constant region, more preferably an IgG1, IgG2, IgG3, IgG4 type heavy chain constant region, and even more preferably a human. It is a heavy chain constant region of IgG1, IgG2, IgG3, and IgG4 types.
  • the amino acid sequences of human IgG1 constant region, human IgG2 constant region, human IgG3 constant region and human IgG4 constant region are known.
  • constant regions of human IgG1, human IgG2, human IgG3, and human IgG4 antibodies a plurality of allotype sequences due to gene polymorphisms are described in Sequences of proteins of immunological interest, NIH Publication No. 91-3242. In any of these cases.
  • the constant region in which the amino acid of the present invention is modified may contain other amino acid mutations and modifications as long as it contains the amino acid mutation of the present invention.
  • the term "hinge region” refers to linking the CH1 and CH2 domains in a wild-type antibody heavy chain, eg, from around 216 to 230 according to the EU numbering system, or around 226 according to the Kabat numbering system. It means the antibody heavy chain polypeptide portion from to the 243rd position.
  • the cysteine residue at position 220 of the EU numbering in the hinge region is known to form a disulfide bond with the cysteine residue at position 214 in the antibody light chain.
  • the cysteine residue at EU numbering 226 and the cysteine residue at 229 in the hinge region form a disulfide bond.
  • the hinge region herein includes the wild-type as well as variants in which amino acid residues have been substituted, added, or deleted in the wild-type.
  • the term "Fc region” is used to define the C-terminal region of an immunoglobulin heavy chain that contains at least part of the constant region.
  • the term includes the Fc region of a native sequence and the mutant Fc region.
  • the human IgG heavy chain Fc region extends from Cys226 or from Pro230 to the carboxyl end of the heavy chain.
  • the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) in the Fc region may or may not be present.
  • the numbering of amino acid residues in the Fc region or constant region is Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, follow the EU numbering system (also known as the EU index) described in MD 1991.
  • variable region refers to the heavy or light chain domain of an antibody involved in binding the antibody to an antigen.
  • the heavy and light chain variable domains of native antibodies are similar, with each domain usually containing four conserved framework regions (FR) and three hypervariable regions (HVR).
  • FR conserved framework regions
  • HVR hypervariable regions
  • antibodies that bind to a particular antigen may be isolated by screening complementary libraries of VL or VH domains using the VH or VL domains from antibodies that bind to that antigen, respectively. See, for example, Portolano et al., J. Immunol. 150: 880-887 (1993); Clarkson et al., Nature 352: 624-628 (1991).
  • hypervariable region are hypervariable in sequence (“complementarity determining regions” or “CDRs” (complementarity determining regions)) and / or are structurally defined. Refers to each region of the variable domain of an antibody that forms a loop (“hypervariable loop”) and / or contains antigen contact residues (“antigen contact”). Usually, the antibody contains 6 HVRs: 3 for VH (H1, H2, H3) and 3 for VL (L1, L2, L3).
  • Illustrative HVRs herein include: (a) At amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3) The resulting hypervariable loop (Chothia and Lesk, J. Mol. Biol. 196: 901-917 (1987)); (b) At amino acid residues 24-34 (L1), 50-56 (L2), 89-97 (L3), 31-35b (H1), 50-65 (H2), and 95-102 (H3).
  • the resulting CDR (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., Supra.
  • variable domain FR refers to variable domain residues other than hypervariable region (HVR) residues.
  • a variable domain FR usually consists of four FR domains: FR1, FR2, FR3, and FR4.
  • sequences of HVR and FR usually appear in VH (or VL) in the following order: FR1-H1 (L1) -FR2-H2 (L2) -FR3-H3 (L3) -FR4.
  • full-length antibody “complete antibody,” and “whole antibody” are used interchangeably herein and have a structure substantially similar to that of a native antibody, or are defined herein.
  • host cell refers to cells into which foreign nucleic acids have been introduced, including progeny of such cells. ..
  • Host cells include “transformants” and “transformants”, which include primary transformants and progeny derived from those cells regardless of the number of passages.
  • the offspring do not have to be exactly the same in the content of the parent cell and nucleic acid and may contain mutations.
  • Variant progeny with the same function or biological activity as those used when the original transformed cells were screened or selected are also included herein.
  • vector refers to a nucleic acid molecule that can augment another nucleic acid to which it is linked.
  • the term includes a vector as a self-replicating nucleic acid structure and a vector incorporated into the genome of the host cell into which it has been introduced. Certain vectors can result in the expression of nucleic acids to which they are operably linked. Such vectors are also referred to herein as "expression vectors.”
  • human antibody is an antibody having an amino acid sequence corresponding to the amino acid sequence of an antibody produced by a human or human cell or an antibody derived from a non-human source using a human antibody repertoire or other human antibody coding sequence. This definition of human antibody explicitly excludes humanized antibodies that contain non-human antigen-binding residues.
  • a “humanized” antibody is a chimeric antibody that contains an amino acid residue from a non-human HVR and an amino acid residue from a human FR.
  • the humanized antibody comprises substantially all of at least one, typically two variable domains, in which all or substantially all HVRs (eg, CDRs) are non-existent.
  • all or substantially all FRs correspond to those of human antibodies.
  • the humanized antibody may optionally include at least a portion of the antibody constant region derived from the human antibody.
  • the "humanized form" of an antibody (eg, a non-human antibody) refers to an antibody that has undergone humanization.
  • the antigen-binding molecule is a recombinant antigen-binding molecule produced using a gene recombination technique.
  • a gene recombination technique See, for example, Borrebaeck CAK and Larrick JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990.
  • the recombinant antigen-binding molecule clones the DNA encoding it from an antigen-binding molecule-producing cell such as a hybridoma or a sensitized lymphocyte that produces an antigen-binding molecule, incorporates it into an appropriate vector, and introduces it into a host. It can be obtained by producing it.
  • the antigen-binding molecule is not limited to an antibody and may be an aptamer.
  • the antibody fragment includes a diabody (Db), a linear antibody, a VHH (variable domain of heavy chain of heavy chain antibody), and a single chain antibody (hereinafter, also referred to as scFv).
  • Db diabody
  • VHH variable domain of heavy chain of heavy chain antibody
  • scFv single chain antibody
  • the "Fv” fragment is the smallest antibody fragment and contains the complete antigen recognition and binding sites.
  • the "Fv” fragment is a dimer (V H- V L dimer) in which one heavy (H) chain variable region (V H ) and a light (L) chain variable region (V L ) are strongly linked by a non-covalent bond. ..
  • variable region The three complementarity determining regions (CDRs) of each variable region interact to form an antigen-binding site on the surface of the V H - VL dimer. Six CDRs confer an antigen binding site on the antibody. However, even one variable region (or half of the Fv containing only three antigen-specific CDRs) has the ability to recognize and bind antigens, albeit with lower affinity than the full binding site. ..
  • the Fab fragment (also referred to as F (ab)) further contains a constant region of the L chain and a constant region of the H chain (CH1).
  • the Fab'fragment differs from the Fab fragment in that it additionally has a few residues from the carboxy terminus of the H chain CH1 region containing one or more cysteines from the hinge region of the antibody.
  • Fab'-SH refers to Fab'in which one or more cysteine residues in the constant region have a free thiol group.
  • the F (ab') fragment is produced by cleavage of the disulfide bond in cysteine at the hinge of the F (ab') 2 pepsin digest. Other chemically bound antibody fragments are also known to those of skill in the art.
  • Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger P et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP404,097, WO 93/11161, etc.).
  • a diabody is a dimer composed of two polypeptide chains, in which the L chain variable region ( VL ) and the H chain variable region (V H ) cannot bind to each other in the same chain, respectively. It is bound by a linker that is short in position, for example, about 5 residues. Since V L and V H encoded on the same polypeptide chain cannot form a single chain variable region fragment due to the short linker between them and form a dimer, the diabody binds two antigens. Will have a site.
  • a single chain antibody or scFv antibody fragment contains the V H and VL regions of the antibody, which are located within a single polypeptide chain.
  • Fv polypeptides also contain a polypeptide linker between the V H and VL regions, which allows scFv to form the structure required for antigen binding (for a review of scFv, Pluckthun. See The Pharmacology of Monoclonal Antibodies, Vol.113 (Rosenburg and Moore ed (Springer Verlag, New York) pp.269-315, 1994).
  • the linker in the present invention is not particularly limited as long as it does not inhibit the expression of the antibody variable region linked to both ends thereof.
  • IgG-type bispecific antibodies can be secreted by hybridomas (quadromas) produced by fusing two hybridomas that produce IgG antibodies (Milstein C et al. Nature 1983, 305: 537-540). In addition, it can be secreted by co-expressing a total of four genes, L chain and H chain genes constituting the two target IgGs, into cells.
  • IgG in a heterogeneous combination for the H chain can be preferentially secreted (Ridgway JB et al. Protein Engineering 1996, 9: 617-621, Merchant). AM et al. Nature Biotechnology 1998, 16: 677-681).
  • the L chain since the diversity of the L chain variable region is lower than that of the H chain variable region, it is expected that a common L chain capable of imparting a binding ability to both H chains can be obtained. Efficient bispecific IgG can be expressed by expressing IgG by introducing this common L-chain and both H-chain genes into cells (Nature Biotechnology. 1998, 16, 677-681). However, if two types of antibodies are arbitrarily selected, it is unlikely that they will contain the same L chain, and it is difficult to carry out the above idea. A method of selection has also been proposed (WO2004 / 065611). The H chain having the above mutation (Nature Biotechnology.
  • Bispecific antigen-binding molecules can also be made by chemically cross-linking Fab'.
  • Fab'prepared from one antibody is maleimided with o-PDM (ortho-phenylenedi-maleimide), and Fab'prepared from the other antibody is reacted with this to crosslink Fab'derived from different antibodies.
  • Bispecific F (ab') 2 can be produced (Keler T et al. Cancer Research 1997, 57: 4008-4014). Also known is a method of chemically binding a Fab'-thionitrobenzoic acid (TNB) derivative to an antibody fragment such as Fab'-thiol (SH) (Brennan M et al. Science 1985, 229: 81-83). ..
  • TNB Fab'-thionitrobenzoic acid
  • SH Fab'-thiol
  • Leucine zippers derived from Fos, Jun, etc. can be used instead of chemical cross-linking. Fos and Jun also form homodimers, but take advantage of the preferential formation of heterodimers.
  • the expression of Fab'with the Fos leucine zipper and the other Fab'with the addition of that of Jun are prepared.
  • Bispecific F (ab') 2 can be formed by mixing and reacting the reduced monomers Fab'-Fos and Fab'-Jun under mild conditions (Kostelny SA et al. J of Immunology, 1992, 148: 1547-53). This method is not limited to Fab', but can also be applied to scFv, Fv, etc.
  • a bispecific antigen-binding molecule can also be prepared in the diabody.
  • the bispecific diabody is a heterodimer of two cross-over scFv fragments.
  • Sc (Fv) 2 which can be produced by linking two types of scFv with a flexible and relatively long linker of about 15 residues, can also be a bispecific antigen-binding molecule (Mallender WD et al. J of Biological Chemistry, 1994, 269: 199-206).
  • antibody modifications include antibodies bound to various molecules such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the substance to be bound is not limited.
  • Such antibody modifications can be obtained by chemically modifying the obtained antibody. These methods have already been established in this field.
  • the origin of the antigen-binding molecule of the present invention is not limited, such as human antibody, mouse antibody, and rat antibody. Further, a gene-modified antibody such as a chimeric antibody or a humanized antibody may be used.
  • a human antibody of interest can be obtained by immunizing a transgenic animal having the entire repertoire of human antibody genes with the antigen of interest (publication of international patent application). See numbers WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735).
  • a chimeric antibody is an antibody consisting of a variable region of H chain and L chain of an immune animal antibody and a constant region of H chain and L chain of a human antibody.
  • a chimeric antibody can be obtained by ligating a DNA encoding a variable region of an antibody derived from an immune animal with a DNA encoding a constant region of a human antibody, incorporating this into an expression vector, introducing it into a host, and producing it. ..
  • a humanized antibody is a modified antibody that is also called a reshaped human antibody.
  • Humanized antibodies are constructed by transplanting the CDRs of antibodies from immune animals into the complementarity determining regions of human antibodies. The general gene recombination method is also known.
  • oligos prepared by preparing a DNA sequence designed to link the CDR of a mouse antibody and the framework region (FR) of a human antibody so as to have an overlapping portion at the terminal portion. It is synthesized from nucleotides by the PCR method. It is obtained by ligating the obtained DNA with the DNA encoding the human antibody constant region, then incorporating it into an expression vector, introducing it into a host and producing it (European Patent Application Publication No. EP 239400, International Patent Application Publication). See number WO 96/02576).
  • the FR of the human antibody linked via CDR is selected so that the complementarity determining regions form a good antigen-binding site.
  • the amino acids in the framework regions of the variable region of the antibody may be replaced so that the complementarity determining regions of the reconstituted human antibody form the appropriate antigen binding site (Sato K et al, Cancer Research 1993, 53: 851-856). It may also be replaced with framework regions derived from various human antibodies (see International Patent Application Publication No. WO 99/51743).
  • the enzyme hydrolyzes the substrate by recognizing both the enzyme and the substrate capable of undergoing a catalytic reaction of the enzyme, but blood coagulation factor V, blood coagulation factor VIII, tissue factor (TF). , From Thrombomodulin (TM), Protein S (PS), Protein Z (PZ) Complement C4b, Complement Regulatory Factor H, membrane cofactor Protein (MCP), complement receptor1 (CR1)
  • TM Thrombomodulin
  • PS Protein S
  • PZ Protein Z
  • C4b Complement Regulatory Factor H
  • MCP membrane cofactor Protein
  • CR1 complement receptor1
  • the enzyme in these embodiments is not particularly limited as long as it is a molecule having catalytic activity for a chemical reaction occurring in a living body.
  • the enzyme include protease, amylase, cellulase, and lipase.
  • proteases include serine proteases of the blood coagulation system.
  • the enzyme can be a blood coagulation enzyme.
  • serine proteases that are blood coagulation enzymes include F.XII (a), F.XI (a), F.IX (a), FX (a), F.VII (a), and F.IIa. ..
  • F.XII (a) means F.XII and / or F.XIIa, and the same applies to others.
  • the substrate capable of undergoing the catalytic reaction of the enzyme referred to in the present application means a substrate in which the catalytic reaction of the enzyme occurs when the enzyme is forcibly brought close to each other.
  • the catalytic reaction of the enzyme means that the substrate is hydrolyzed. Hydrolysis of the substrate may be performed as long as the substrate is hydrolyzed as compared with the case where the bispecific antigen-binding molecule of the present application is absent. Substrate hydrolysis reactions can be compared by measuring the amount of protein over time after the addition of a bispecific antigen-binding molecule by the Bradford method or the like using, for example, Protein Assey Kit II (Bio-Red).
  • Substrate hydrolysis includes, for example, activation of blood coagulation factor X or blood coagulation factor IX.
  • the activation of the substrate can be performed by using the enzyme and the reaction system containing the substrate, and the increase in the enzyme activity (substrate resolution) by adding the antigen-binding molecule can be used as an index.
  • the enzyme for example, the enzyme, the substrate, and the activity. It is a measurement system (in vitro enzyme reaction measurement system) consisting of a synthetic substrate, phospholipids, and Ca2 + of the converted substrate, and can be evaluated by the substrate activation promoting activity by the enzyme. Based on the results, as a bispecific antigen-binding molecule having the activity, in principle, a molecule showing the substrate activation promoting activity by the enzyme of 0.1 or more only in the enzyme-added group can be selected in this measurement system.
  • the substrate activation promoting activity by the enzyme referred to here can be measured by the value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.
  • “Promoting the activation of the substrate by the enzyme” means that the substrate activation promoting activity in the in vitro enzyme reaction measurement system shows an absorbance of 0.1 or more 30 minutes after the addition of the chromogenic substrate solution.
  • the in vitro enzyme reaction measurement system can be measured with each enzyme and each substrate. For example, as shown in Example 2, as the FX activation promoting activity by F.XIa, as the absorbance value 30 minutes after the addition of the color-developing substrate solution. When it is 0.1 or more, it can be evaluated as promoting the activation of the substrate by the enzyme.
  • the enzyme also referred to as “blood coagulation enzyme”
  • the substrate also referred to as “blood coagulation substrate”
  • the enzyme and the substrate are blood coagulation / fibrinolysis-related factors
  • the method for obtaining the bispecific antigen-binding molecule is not particularly limited, and any method may be used for obtaining the bispecific antigen-binding molecule.
  • a bispecific antigen-binding molecule for enzyme A and substrate B is obtained, each of enzyme A and substrate B is immunized against an immunized animal to obtain an anti-enzyme A antibody and an anti-substrate B antibody. Then, a bispecific antigen-binding molecule containing the H-chain variable region and the L-chain variable region of the anti-enzyme A antibody and the H-chain variable region and the L-chain variable region of the anti-substrate B antibody is prepared.
  • the antigen-binding molecule that activates substrate B is selected.
  • the antigen-binding molecule to the enzyme or substrate can be obtained by a method known to those skilled in the art. For example, it can be prepared by immunizing an immune animal with an antigen. Examples of the antigen that immunizes an animal include a complete antigen having immunogenicity and an incomplete antigen (including a hapten) having no immunogenicity. In immunization, an enzyme or substrate to which an antigen-binding molecule binds, or a nucleic acid expressing them is used as the above-mentioned antigen (immunogen). As the immunized animal, for example, mice, rats, hamsters, guinea pigs, rabbits, chickens, rhesus monkeys and the like can be used.
  • Immunization of these animals with an antigen can be performed by a method well known to those skilled in the art.
  • the variable regions of the L and H chains of the antibody are recovered from the immunized animal or the cells of the animal. This operation can be performed using a technique generally known to those skilled in the art.
  • Animals immunized with an antigen express antibodies against that antigen, especially in spleen cells. Therefore, for example, mRNA can be prepared from the spleen cells of an immunized animal, and the variable regions of L and H chains can be recovered by RT-PCR using the primers corresponding to the variable regions of the animal.
  • the enzyme or substrate used as an immunogen may be the whole protein or a partial peptide of the protein. Further, as the immunogen used for immunizing an animal, it is possible to bind an antigen to another molecule to obtain a soluble antigen, and in some cases, fragments thereof may be used.
  • Spleen cells are isolated from the spleen of immunized mice and fused with mouse myeloma cells to produce hybridomas.
  • Hybridomas that bind to the antigen can be selected, and the variable regions of the L and H chains can be recovered by RT-PCR using primers and the like corresponding to the variable regions.
  • Primers that correspond to CDRs, frameworks that are less diverse than CDRs, or primers that correspond to the signal sequence and the CH1 or L chain constant region ( CL ) can also be used. Alternatively, they can be produced by B cell cloning techniques known to those of skill in the art (Proc Natl Acad Sci US A. 1996; 93 (15): 7843-7848 .; WO2008 / 045140; and WO2009 / 113742). Not limited.
  • mRNA is extracted from the splenocytes of an immunized animal, and cDNA in the L-chain and H-chain variable regions is recovered by RT-PCR using a primer corresponding to the vicinity of the variable region. It can also immunize lymphocytes in vitro. Use this to build a library that presents scFv or Fab. Antigen-binding antibody clones can be concentrated and cloned by panning to obtain variable regions. At this time, it is also possible to perform screening using a similar library using mRNA derived from peripheral blood mononuclear cells, spleen, tonsils, etc. of humans and non-immune animals.
  • An antigen-binding molecule expression vector is prepared using the variable region.
  • a bispecific antigen-binding molecule can be obtained by introducing an anti-enzyme antigen-binding molecule expression vector and an anti-substrate antigen-binding molecule expression vector into the same cell and expressing the antigen-binding molecule.
  • the antigen-binding molecule in the present invention can be selected by, for example, the following method.
  • a reaction system containing the enzyme / substrate is used, and an increase in the enzyme activity (substrate resolution) due to the addition of the antigen-binding molecule is used as an index for selection.
  • the function recovery activity by adding the antigen-binding molecule under the conditions is used as an index for selection.
  • the obtained antigen-binding molecule can be purified to a uniform level.
  • the separation and purification methods used for ordinary proteins may be used.
  • an antibody can be separated and purified by appropriately selecting and combining a chromatography column such as affinity chromatography, a filter, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, isoelectric focusing, and the like. It can be done (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988), but it is not limited to these.
  • Examples of the column used for affinity chromatography include a protein A column and a protein G column.
  • the bispecific antigen-binding molecule of the present invention preferably has a variable region in an anti-F.XIa antibody when, for example, the combination of enzyme and substrate is the blood coagulation / fibrinolysis-related factors F.XIa and FX. And a variable region in the anti-FX antibody.
  • the bispecific antigen-binding molecule can be prepared by the following method. For example, when the combination of enzyme and substrate was F.XIa and FX, commercially available F.XIa and FX were subcutaneously immunized in mice, respectively. Spleen cells were isolated from the spleen of immune mice with elevated antibody titers and fused with mouse myeloma cells to prepare hybridomas. Hybridomas that bind to antigens (F.XIa, FX) were selected, and the variable regions of L and H chains were recovered by RT-PCR using primers corresponding to the variable regions. L chain variable region to the L chain expression vector containing a C L, H chain variable region incorporating the respective H chain expression vector containing an H chain constant region.
  • mRNA was extracted from the spleen of this immune mouse, and cDNA in the L-chain and H-chain variable regions was recovered by RT-PCR using primers corresponding to the variable regions.
  • a phage library that presents scFv was constructed using these variable regions.
  • An antigen-binding antibody clone was concentrated and cloned by panning, and an antigen-binding molecule expression vector was prepared using the variable region.
  • it can be produced by a B cell cloning technique by a method known to those skilled in the art. Bispecific antigen binding by introducing an anti-F.XIa antibody (H chain, L chain) expression vector and an FX antibody (H chain, L chain) expression vector into the same cell and expressing an antigen-binding molecule. Obtained a molecule.
  • the FX activation promoting activity by F.XIa was evaluated by a measurement system consisting of F.XIa, FX, F.Xa synthetic substrate (S-2222), and phospholipid. bottom. Based on the results, as the bispecific antigen-binding molecule to have, in principle, a molecule showing F.X activation promoting activity by F.XIa of 0.1 or more in this measurement system was selected.
  • the F.X activation promoting activity by F.XIa referred to here is a value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.
  • the coagulation recovery ability was measured using a coagulation time measurement system using blood coagulation factor-deficient human plasma.
  • a bispecific antigen-binding molecule having a shorter coagulation time than when no antibody was added was obtained.
  • the coagulation time referred to here is a measurement of the activated partial thromboplastin time using human plasma deficient in blood coagulation factor IX.
  • the most preferable bispecific antigen-binding molecule had the ability to shorten the coagulation time by 60 seconds or more.
  • the antigen-binding molecule of the present invention is not particularly limited, and examples thereof include bispecific antigen-binding molecules that recognize both the enzyme and the substrate of any combination shown in Table 1.
  • the antigen-binding molecule of the present invention is a combination of activated blood coagulation factor X (F.Xa) and blood coagulation factor X (FX) shown in Table 1, and activated blood coagulation factor XI (F.XIa).
  • activated blood coagulation factor XI F.XIa
  • it may be a monospecific antigen-binding molecule having the same heavy chain and light chain, and more preferably an antibody.
  • the constant region is not particularly limited, and constant regions known to those skilled in the art can be used.
  • constant regions known to those skilled in the art can be used.
  • Sequences of proteins of immunological interest. (1991), US Department of Health and Human Services. Public Health Service National Institutes of Health, An efficiency route to human bispecific IgG, (1998). Nature Biotechnology vol. 16, 677-681, etc. Constant regions can be used.
  • the antigen-binding molecule does not at least replace the function of a peptide cofactor or heparin expressed in the body, but complements the relationship between the enzyme and a substrate capable of undergoing a catalytic reaction of the enzyme. Since it fulfills factor-like functions, it is expected to be an effective drug for diseases caused by decreased activity (function) or deficiency of certain cofactors, enzymes, and substrates.
  • the enzyme and substrate to which the antigen-binding molecule of the present invention binds are blood coagulation / fibrinolysis-related factors
  • examples of the above-mentioned diseases include bleeding, diseases associated with bleeding, and diseases caused by bleeding. ..
  • hemophilia bleeding dysfunction due to congenital F.VIII / F.VIIIa dysfunction or deficiency is called hemophilia A
  • bleeding dysfunction due to F.IX / F.IXa dysfunction or deficiency is blood.
  • Called friendship B If hemophilia A patients bleed, F.VIII replacement therapy is given, and if hemophilia B patients bleed, F.IX replacement therapy is given.
  • prophylactic administration of these preparations may be performed on the day of strenuous exercise or excursion.
  • F.VIII preparation in blood is short, about 12 to 16 hours. Therefore, for continuous prophylaxis, it is necessary to administer the F.VIII preparation about three times a week. This corresponds to maintaining approximately 1% or more of F.VIII activity. Similarly, the F.IX preparation with a standard half-life should be administered about twice a week. In addition, in replacement therapy for bleeding, it is necessary to administer additional F.VIII and F.IX products regularly for a certain period of time in order to prevent rebleeding and completely stop bleeding, except when the bleeding is mild. There is.
  • the F.VIII preparation and the F.IX preparation are administered intravenously.
  • hemophilia A patients especially severe hemophilia A patients, may develop antibodies against F.VIII called inhibitors.
  • patients with hemophilia B may develop antibodies to F.IX called inhibitors.
  • the inhibitor When the inhibitor is generated, the effect of the F.VIII or F.IX preparation is hindered by the inhibitor. As a result, hemostasis management for the patient becomes very difficult.
  • hemophilia A inhibitor patients often do not have a sufficient hemostatic effect as compared to non-inhibitor hemophilia A patients. The same is true for patients with hemophilia B inhibitors.
  • the dosing interval is wide, (ii) the dosing is easy, (iii) independent of the presence of the inhibitor, and (iv) F.VIII / F.VIIIa or F.IX / F.IXa independent.
  • a method using an antibody can be considered for the creation of a drug that replaces the function.
  • the half-life of an antibody in blood is generally relatively long, ranging from days to weeks.
  • the antibody is transferred to the blood after subcutaneous administration. That is, in general, antibody drugs satisfy the above (i) and (ii).
  • the present invention provides a pharmaceutical composition containing the antigen-binding molecule of the present invention as an active ingredient.
  • the antigen-binding molecule of the present invention is an antigen-binding molecule that recognizes both the enzymes and substrates of the combinations shown in Table 1 and promotes the blood coagulation reaction in the blood coagulation factor-deficient plasma.
  • the antigen-binding molecule is expected to be a drug (pharmaceutical composition) or drug for preventing or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • the pharmaceutical composition containing the antigen-binding molecule of the present invention used for therapeutic or prophylactic purposes as an active ingredient is optionally mixed with an appropriate pharmaceutically acceptable carrier, medium or the like which is inert to them.
  • an appropriate pharmaceutically acceptable carrier for example, sterile water, physiological saline, stabilizers, excipients, antioxidants (ascorbic acid, etc.), buffers (phosphate, citric acid, other organic acids, etc.), preservatives, surfactants (PEG, etc.) Tween etc.), chelating agent (EDTA etc.), binder and the like can be mentioned.
  • low molecular weight polypeptides serum albumin, proteins such as gelatin and immunoglobulin, amino acids such as glycine, glutamine, asparagine, arginine and lysine, sugars and carbohydrates such as polysaccharides and monosaccharides, sugars such as mannitol and sorbitol. It may contain alcohol.
  • an aqueous solution for injection include physiological saline, isotonic solutions containing glucose and other adjuvants, such as D-sorbitol, D-mannitol, D-mannitol, and sodium chloride, which are appropriately dissolved.
  • Auxiliary agents such as alcohol (ethanol, etc.), polyalcohol (propylene glycol, PEG, etc.), nonionic surfactant (polysorbate 80, HCO-50), etc. may be used in combination.
  • the antigen-binding molecule of the present invention can be encapsulated in microcapsules (microcapsules of hydroxymethyl cellulose, gelatin, poly [methyl methacrylate], etc.), or a colloidal drug delivery system (liposomes, albumin microspheres, microemulsions, etc.). Nanoparticles and nanocapsules, etc.) (see “Remington's Pharmaceutical Science 16th edition", Oslo Ed. (1980), etc.). Furthermore, a method of making a drug a sustained release drug is also known and can be applied to the antigen-binding molecule of the present invention (Langer et al., J. Biomed. Mater. Res. 15: 267-277 (1981); Langer, Chemtech.
  • the antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor VIII.
  • Blood coagulation factor VIII may be made from human blood or genetically modified.
  • the antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor VIII, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor VIII.
  • the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with blood coagulation factor VIII, it is possible to reduce the dose of each if desired, as compared with the case where either of them is used alone.
  • the antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor IX.
  • Blood coagulation factor IX may be made from human blood or genetically modified.
  • the antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor IX, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor IX.
  • the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with blood coagulation factor IX, it is possible to reduce the dose of each if desired, as compared with the case where either of them is used alone.
  • the antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor XI.
  • Blood coagulation Factor XI may be made from human blood or genetically modified.
  • the antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor XI, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor XI.
  • the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with factor XI for blood coagulation, it is possible to reduce the dose of each dose as desired, as compared with the case where either one is used alone.
  • the antigen-binding molecule or composition of the present invention can be used in combination with a bypass preparation.
  • the bypass preparation may be made from human blood or by gene recombination, for example, plasma-derived active prothrombin complex preparation (APCC preparation), gene recombination type activation first.
  • Factor VII preparation rF.VIIa preparation
  • dry concentrated human blood coagulation factor X-activated factor VII preparation F.VIIa / FX preparation.
  • the antigen-binding molecule or composition of the present invention may be administered at the same time as the bypass preparation, or may be administered at different times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with a bypass preparation.
  • the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with the bypass preparation, it is possible to reduce the dose of each, if desired, as compared with the case where either of them is used alone.
  • the dose of the pharmaceutical composition of the present invention is finally determined by a doctor in consideration of the type of dosage form, administration method, age and weight of the patient, symptoms of the patient, type of disease and degree of progression, etc. Although it is determined as appropriate, in general, for adults, 0.1 to 2000 mg can be administered in 1 to several divided doses per day. It is more preferably 1 to 1000 mg / day, even more preferably 50 to 500 mg / day, and most preferably 100 to 300 mg / day. These doses vary depending on the body weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose. It is preferable that the administration period is also appropriately determined according to the healing course of the patient and the like.
  • the gene encoding the antigen-binding molecule of the present invention into a gene therapy vector to perform gene therapy.
  • a gene therapy vector to perform gene therapy.
  • it is packaged in liposomes or the like, or formed as various virus vectors such as retrovirus vector, adenovirus vector, vaccinia virus vector, poxvirus vector, adenovirus-related vector, and HVJ vector. (See Adolph "Virus Genome Method", CRC Press, Florid (1996)), or it can be administered by coating it on a bead carrier such as colloidal gold particles (WO93 / 17706, etc.).
  • the nucleic acid encoding the antigen-binding molecule of the present invention may be directly administered to a living body, or may be directly administered to a living body by an electroporation method.
  • the mRNA encoding the antigen-binding molecule of the present invention is chemically modified to enhance the stability of the mRNA in vivo, and the mRNA is directly administered to humans to express the antigen-binding molecule of the present invention in vivo.
  • the antigen-binding molecule of the present invention can be administered by the method of causing (see EP2101823B, WO2013 / 120629). However, as long as the antigen-binding molecule is expressed in vivo and exerts its action, it may be administered by any method.
  • injection, infusion, or gas-induced particle impact is performed via a suitable parenteral route (intravenous, intraperitoneal, subcutaneous, intradermal, intra-adipose tissue, intramammary gland tissue, inhalation or intramuscular route).
  • a sufficient amount is administered by (with a gun, etc.), a method via a mucosal route such as a nasal spray, etc.).
  • the present invention is produced by administering to blood cells, bone marrow-derived cells, etc. using liposome transfection, particle impact method (US Patent No. 4,945,050), or virus infection in ex vivo, and reintroducing the cells into animals.
  • a gene encoding an antigen-binding molecule may be administered.
  • the present invention also provides a method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the step of administering the antigen-binding molecule or composition of the present invention.
  • Administration of the antigen-binding molecule or composition can be carried out, for example, by the method described above.
  • bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX (eg,). , Hemophilia B).
  • the disease is a disease in which an inhibitor of blood coagulation factor IX or activated blood coagulation factor IX has emerged.
  • the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor IX or activated blood coagulation factor IX.
  • the method of the invention further comprises the step of administering blood coagulation factor IX.
  • bleeding, a disease associated with bleeding, or a disease resulting from bleeding is a disease that develops and / or progresses due to decreased or deficient activity of hemocoagulation factor VIII and / or activated hemocoagulation factor VIII.
  • hemophilia A acquired hemophilia, or Fonville brand disease
  • the disease is a disease in which an inhibitor of blood coagulation factor VIII or activated blood coagulation factor VIII has emerged.
  • the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor VIII or activated blood coagulation factor VIII.
  • the method of the invention further comprises the step of administering blood coagulation factor VIII.
  • bleeding, a disease associated with bleeding, or a disease resulting from bleeding is a disease that develops and / or progresses due to decreased or deficient activity of blood coagulation factor XI and / or activated blood coagulation factor XI.
  • hemophilia C or acquired hemophilia
  • the disease is a disease in which an inhibitor of blood coagulation factor XI or activated blood coagulation factor XI appears.
  • the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor XI or activated blood coagulation factor XI.
  • the method of the invention further comprises the step of administering blood coagulation factor XI.
  • the present invention also relates to the use of the antigen-binding molecule of the present invention for the production of the (pharmaceutical) composition of the present invention.
  • the present invention provides a kit for use in the above method, which comprises at least the antigen-binding molecule or composition of the present invention.
  • the kit may also be packaged with a syringe, a needle, a pharmaceutically acceptable medium, an alcohol cotton cloth, an adhesive plaster, or instructions describing how to use it.
  • the kit of the present invention is for use in a method of preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • the kit of the invention comprises a blood coagulation factor IX or factor VIII in addition to the antigen binding molecule or composition of the invention.
  • the present invention relates to a method of promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both the enzyme and the substrate of any combination shown in Table 1.
  • the method for promoting blood coagulation of the present invention comprises the step of administering a bispecific antigen-binding molecule that recognizes both the enzyme and the substrate of any combination shown in Table 1.
  • the administration of the antigen-binding molecule can be carried out by, for example, the above-mentioned administration method.
  • the bispecific antigen binding molecule used in the method for promoting blood coagulation of the present invention is a bispecific antigen that recognizes both activated blood coagulation factor XI and blood coagulation factor X. It is a binding molecule.
  • the present invention relates to a method for screening a substance effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • the screening method of the present invention comprises (1) a step of evaluating the binding of the test substance to any combination of enzymes shown in Table 1, and (2) the test substance and Table 1. Includes a step of evaluating the binding of the combination to the substrate according to.
  • the screening methods of the invention assess (1) the step of assessing the binding of the test substance to activated blood coagulation factor XI, and (2) assessing the binding of the test substance to blood coagulation factor X.
  • the screening methods of the present invention use a test substance that binds to both the enzyme and substrate of any of the combinations listed in Table 1 to prevent bleeding, bleeding-related diseases, or diseases caused by bleeding. / Or further includes the step of selecting as a candidate for a therapeutically effective substance.
  • test substance is not particularly limited, and includes, for example, a single substance such as a natural compound, an organic compound, an inorganic compound, a nucleic acid, a protein, and a peptide, as well as a compound library, a nucleic acid library, a peptide library, and a gene live. Examples thereof include rally expression products, cell extracts, cell culture supernatants, fermented microbial products, marine organism extracts, plant extracts, prokucleic acid cell extracts, eukaryotic single cell extracts, animal cell extracts and the like.
  • the test substance can be appropriately labeled and used as needed. Examples of the label include a radial label, a fluorescent label and the like. In addition to the above test substances, a mixture of a plurality of these test substances is also included.
  • the present invention relates to a quality test method for a substance or composition effective in the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
  • the quality test method of the present invention comprises (1) a step of evaluating the binding of a test substance or test composition to an enzyme of any combination shown in Table 1, and (2) a test. The step of evaluating the binding between the substance or the test composition and the substrate of the combination shown in Table 1 is included.
  • the quality test method of the present invention comprises (1) a step of evaluating the binding of the test substance or test composition to activated blood coagulation factor XI, and (2) the test substance or test composition and blood.
  • the quality test method of the invention results in bleeding, a disease associated with bleeding, or bleeding that binds a test substance or composition that binds to both of the enzymes and substrates of any of the combinations listed in Table 1. It further comprises the step of assessing the quality of the substance or composition as effective for the prevention and / or treatment of the disease.
  • test composition is not particularly limited, and examples thereof include pharmaceuticals and reagents.
  • Examples of the steps for evaluating the binding between the test substance and the activated blood coagulation factor XI include an ELISA method (Enzyme-Linked Immuno Sorbent Assay) and a surface plasmon resonance (SPR) method, for example, Biacore. It can be evaluated by measurement using a series (GE Healthcare) and biosensor interaction analysis system using biosensor technology (BLI method), for example, Octet system (Fortebio).
  • Examples of the steps for evaluating the binding between the test substance and blood coagulation factor X include an ELISA method (Enzyme-Linked ImmunoSorbent Assay) and a surface plasmon resonance (SPR) method, for example, the Biacore series (Biacore series). It can be evaluated by measurement using GE Healthcare) and biosensor interaction analysis system using biosensor technology (BLI method), for example, Octet system (Fortebio).
  • Activation As a step of evaluating the activation reaction of blood coagulation factor X by factor XI using a test substance, for example, the enzyme, the substrate, the synthetic substrate of the activated substrate, phosphorus It is a measurement system consisting of lipid and Ca2 +, and can be evaluated by the substrate activation promoting activity by the enzyme. Based on the results, as a bispecific antigen-binding molecule having the activity, in principle, a molecule showing the substrate activation promoting activity by the enzyme of 0.1 or more only in the enzyme-added group can be selected in this measurement system.
  • the substrate activation promoting activity by the enzyme referred to here can be measured by the value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.
  • the following antibody variable region sequences were used for anti-FX antibody preparation: F10B1 from SB04 described in WO2005035756A1 (SEQ ID NO:: heavy chain variable region SEQ ID NO: 3, light chain variable region herein). SEQ ID NO: 4); F10B2 consisting of a sequence in which two amino acid substitutions are introduced into the heavy chain variable region sequence of J327 and the light chain variable region sequence of JNL095 described in WO2019065795A1 (SEQ ID NO:: heavy chain in the present specification).
  • Variable region SEQ ID NO: 5 Light chain variable region SEQ ID NO: 6
  • FX antibody BIIB-12-917 described herein as F10B3: heavy chain variable region
  • SEQ ID NOs: 427 and 615 of WO2018098363 SEQ ID NO: 17
  • FX antibody mAb 00916 described in SEQ ID NOs: 4 and 5 of WO2019096874 (in the present specification, the first residue Ala of the light chain is removed and described as F10B4.
  • the variable region sequence of each antibody was linked to the heavy chain constant region or light chain constant region sequence to construct an expression vector containing a gene encoding the full length of the antibody sequence.
  • the expression vector was transiently introduced into Expi293 cells (Thermo Fisher Scientific) to express the antibody.
  • the obtained culture supernatant was purified by a method known to those skilled in the art by affinity purification using Protein A or the like.
  • bispecific antibodies consisting of these anti-F.XIa antibodies and anti-FX antibodies were prepared by methods known to those skilled in the art.
  • the correspondence between the sequence numbers of the variable region and the constant region of each antibody is shown in Table 2, and the names of the respective monospecific antibodies and bispecific antibodies are also named as shown in Table 2.
  • Example 2 Measurement of FX activation promoting activity by F.XIa of F.XIa and FX bispecific antibody by in vitro enzyme reaction measurement system The presence or absence of FX activation promoting activity by F.XIa was evaluated using an in vitro enzyme reaction measurement system using a synthetic color-developing substrate. Specifically, the measurement was carried out according to the following procedure, and all the reactions were carried out at room temperature. A mixture of 4 ⁇ L of 48 ng / mL F.XIa (Enzyme Research Laboratories) and 5 ⁇ L of antibody solution at each concentration was incubated in a 384-well plate for 30 minutes.
  • FX Enzyme Research Laboratories
  • 5 ⁇ L of FX Enzyme Research Laboratories
  • 50 ⁇ M Aprotinin Sigma Aldrich
  • 5 ⁇ L of a synthetic color-developing substrate solution colored by F.Xa was added to each well, and the absorbance at a wavelength of 405 nm after 30 minutes was measured by SpectraMax340PC (Molecular Devices).
  • the FX activation promoting activity of F.XIa and FX bispecific antibodies by F.XIa was expressed as the absorbance value 30 minutes after the addition of the coloring substrate solution [Figs. 3 and 5].
  • the antibody concentration is shown as the concentration in the solution during the enzymatic reaction.
  • F.XIa and FX an antibody concentration-dependent increase in the amount of F.Xa indicated by absorbance was confirmed in the group to which the bispecific antibody was added.
  • no increase in absorbance was confirmed in the group to which the monospecific antibody was added instead of the bispecific antibody. From the above, it was suggested that F.XIa and FX bispecific antibodies promote FX activation in an in vitro enzyme reaction measurement system in a binding-dependent manner to both F.XIa and FX.
  • TBSB Tris-buffered physiological saline containing 0.1% bovine serum albumin
  • TBSB (hereinafter referred to as TBCP) containing 1.5 mM CaCl2 and 4.0 ⁇ M phospholipid (Sysmex) was used as the solvent for F.XIa and FX.
  • TBCP Tris-buffered physiological saline containing 0.1% bovine serum albumin
  • Example 3 Measurement of plasma coagulation activity by activated partial thromboplastin time (APTT)
  • the blood coagulation reaction is a sequential substrate activation reaction by a plurality of serine proteases.
  • FX bispecific antibody having FX activation promoting activity promotes the same reaction in hemophilia B plasma and corrects coagulation ability
  • F.IX deficient plasma is used.
  • the effect of the antibody on activated partial thromboplastin time (APTT) was investigated. APTT measurement was carried out by a method known to those skilled in the art, and specifically, the following procedure was carried out.
  • Plasma samples were prepared. A mixture of 50 ⁇ L of plasma sample and 50 ⁇ L of APTT reagent (Sysmex) was heated at 37 ° C. for 190 seconds. The coagulation reaction was initiated by adding 50 ⁇ L of 0.02 M calcium chloride solution (Sysmex) to the mixture.
  • the coagulation time was measured using CS-2000i (Sysmex) based on the time when the decrease in permeability was 50% of the maximum decrease, and was shown as APTT [Figs. 4 and 6].
  • the antibody concentration is shown as the concentration in the plasma sample containing the antibody.
  • the APTT shortening effect was shown in the bispecific antibody-added group as in the F.IX-added group. From the above results, it was shown that the anti-F.XIa, FX bispecific antibody has a coagulation-promoting effect in F.IX-deficient plasma, and the anti-F.XIa, FX bispecific antibody is associated with hemophilia B. It was suggested that it may correct the coagulation ability of plasma of hemophilia B patients as well as F.IX used clinically as a therapeutic drug.
  • F.XIIa activates F.XI
  • F.XIa activates F.IX
  • F.IXa activates FX
  • F.VIIa activates FX
  • F.Xa activates prothrombin to proceed with the coagulation reaction.
  • Hemophilia is a hemorrhagic disease caused by a congenital or acquired decrease in activity of F.VIIIa, F.IX, and F.XI in Fig. 2.
  • blood coagulation factors other than the blood coagulation factor causing the disease state have a normal function. Therefore, by applying this concept to these normally active blood coagulation factors, the endogenous coagulation cascade can be normalized in hemophiliac plasma, or the reaction of the extrinsic cascade can be promoted to promote bleeding tendency. It can be suppressed.
  • the bleeding tendency in hemophilia can be suppressed by preparing a bispecific antigen-binding molecule that binds to the combination of the enzymes a to h shown in Table 1 and the substrate.
  • the bispecific antigen-binding molecule of the present disclosure contributes to the promotion of blood coagulation and may be useful for the prevention and / or treatment of bleeding, diseases associated with bleeding, or diseases caused by bleeding.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Mycology (AREA)
  • Plant Pathology (AREA)
  • Hematology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Diabetes (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The present invention provides a bispecific antigen-binding molecule that recognizes both of two specific factors out of various coagulation-related factors, as well as a composition and a kit containing the bispecific antigen-binding molecule. In one non-limiting embodiment, the bispecific antigen-binding molecule, composition, and kit of the present invention are used to prevent and/or treat hemorrhaging, diseases accompanied by hemorrhaging, or diseases caused by hemorrhaging. Additionally, the present invention provides a method for using the bispecific antigen-binding molecule to promote coagulation. Furthermore, the present invention provides a screening method for substances that are effective for preventing and/or treating hemorrhaging, diseases accompanied by hemorrhaging, or diseases caused by hemorrhaging.

Description

二重特異性抗原結合分子ならびに、それに関連する組成物、組成物の製造のための使用、キット、および方法Bispecific antigen-binding molecules and their associated compositions, uses, kits, and methods for the manufacture of compositions.

 本開示は、種々の血液凝固関連因子のうち特定の2種の因子の両方を認識する二重特異性抗原結合分子、ならびにそれを含む組成物、組成物の製造のための使用、およびキットに関する。また、本開示は、当該二重特異性抗原結合分子を用いて血液凝固を促進する方法に関する。さらに、本開示は、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法に関する。 The present disclosure relates to bispecific antigen-binding molecules that recognize both of two specific blood coagulation-related factors, as well as compositions containing them, their use in the manufacture of compositions, and kits. .. The present disclosure also relates to a method for promoting blood coagulation using the bispecific antigen-binding molecule. Furthermore, the present disclosure relates to methods of screening for substances that are effective in the prevention and / or treatment of bleeding, bleeding-related diseases, or diseases caused by bleeding.

 血液凝固は、一連の成分、特にフィブリノーゲン、血液凝固第II因子、第V因子、第VII因子、第VIII因子、第IX因子、第X因子、第XI因子、第XII因子(それぞれF.II、F.V、F.VII、F.VIII、F.IX、F.X、F.XI、F.XII)およびそれらの活性型(それぞれF.IIa、F.Va、F.VIIa、F.VIIIa、F.IXa、F.Xa、F.XIa、F.XIIa)、ならびにフォン・ヴィレブランド因子の連続的な相互作用を含む複雑な過程である。 Blood coagulation consists of a series of components, especially fibrinogen, blood coagulation factor II, factor V, factor VII, factor VIII, factor IX, factor X, factor XI, factor XII (F.II, respectively). FV, F.VII, F.VIII, F.IX, FX, F.XI, F.XII) and their active forms (F.IIa, F.Va, F.VIIa, F.VIIIa, F.IXa, respectively) , F.Xa, F.XIa, F.XIIa), as well as a complex process involving the continuous interaction of von Villebrand factors.

 血友病は、これらの成分が欠損、またはその機能性が阻害されることで、血液凝固不能が引き起こされる出血性疾患である。F.VIIIが原因である場合は血友病Aと呼ばれ、F.IXが原因である場合は血友病Bと呼ばれる。血友病患者においては、関節内、筋肉内などの深部組織に出血症状が見られ、重篤な例では頭蓋内出血も生じる。 Hemophilia is a bleeding disease in which blood coagulation is caused by deficiency of these components or inhibition of their functionality. If it is caused by F.VIII, it is called hemophilia A, and if it is caused by F.IX, it is called hemophilia B. In patients with hemophilia, bleeding symptoms are seen in deep tissues such as joints and muscles, and in severe cases, intracranial hemorrhage also occurs.

 血友病の重症度は、血液中のF.VIII活性又はF.IX活性に基づいて分類される。具体的には、健常者のF.VIII活性又はF.IX活性を100%として、活性が1%未満の患者を重症に、活性が1%以上5%未満の患者を中等症に、活性が5%以上40%未満の患者を軽症に分類する。重症血友病患者は、中等症及び軽症の患者に比べて顕著に高い頻度で出血症状を呈する。しかし、F.VIII又はF.IXの補充療法により、患者の血液中のF.VIII活性又はF.IX活性を1%以上に維持することで、出血の頻度を劇的に減少させることができる。補充療法には、主に、血漿から精製されたか又は遺伝子組換え技術により作製された凝固因子製剤が用いられる。 The severity of hemophilia is classified based on F.VIII activity or F.IX activity in the blood. Specifically, assuming that the F.VIII activity or F.IX activity of a healthy person is 100%, the activity is severe in patients with an activity of less than 1%, and moderate in patients with an activity of 1% or more and less than 5%. Patients with 5% or more and less than 40% are classified as mild. Patients with severe hemophilia present significantly more frequent bleeding symptoms than patients with moderate and mild illness. However, F.VIII or F.IX replacement therapy can dramatically reduce the frequency of bleeding by maintaining F.VIII or F.IX activity in the patient's blood at 1% or higher. .. For replacement therapy, coagulation factor preparations purified from plasma or prepared by genetic recombination technology are mainly used.

 血友病A患者の出血に対しては、F.VIII製剤が通常投与される(on-demand投与)。また、近年は、出血イベントを防ぐために、予防的に、F.VIII製剤が投与される(非特許文献1、2)(予防投与)。F.VIII製剤の血中半減期は、約12~16時間程度である。それ故、継続的な予防のためには、週に3回、F.VIII製剤が、患者に投与される(非特許文献3、4)。また、on-demand投与においては、再出血を防ぐため、F.VIII製剤を、必要に応じ、一定間隔で追加投与する。また、F.VIII製剤の投与は静脈内に実施される。また、時折F.VIIIに対する抗体(インヒビター)が、血友病患者に発生する。インヒビターは、F.VIII製剤の効果を打ち消す。従って、F.VIII製剤と比べて投与の負担が少なく、インヒビターの存在に左右されない薬剤が強く求められていた。 For bleeding in hemophilia A patients, F.VIII preparation is usually administered (on-demand administration). In recent years, in order to prevent bleeding events, F.VIII preparations are prophylactically administered (Non-Patent Documents 1 and 2) (preventive administration). The half-life of the F.VIII product in blood is about 12 to 16 hours. Therefore, for continuous prophylaxis, the F.VIII preparation is administered to the patient three times a week (Non-Patent Documents 3 and 4). In addition, in on-demand administration, in order to prevent rebleeding, the F.VIII preparation is additionally administered at regular intervals as needed. In addition, administration of F.VIII preparation is performed intravenously. Occasionally, antibodies (inhibitors) to F.VIII occur in patients with hemophilia. The inhibitor counteracts the effect of the F.VIII preparation. Therefore, there has been a strong demand for a drug that is less burdensome to administer than the F.VIII preparation and is not affected by the presence of the inhibitor.

 これらの課題を解決する手段として、F.VIIIの機能を代替する二重特異性抗体及びその使用が報告されている(特許文献1、2、3および4)。F.IXaとF.Xに対する二重特異性抗体は、両因子を近傍に位置付けることによって、F.VIII補因子機能代替の活性を発揮し、F.VIIIの機能を代替することが可能である(非特許文献5)。該抗体の一つであり、高いF.VIII補因子機能代替活性を有するACE910(Emicizumab)は健常人を対象とした臨床試験において、優れた薬物動態(長い半減期)と忍容性が確認され(非特許文献6)、インヒビター非保有、保有の血友病Aの患者を対象とした臨床試験において、ACE910(Emicizumab)の投与前と比較して、ACE910(Emicizumab)投与により顕著な出血回数の抑制が認められた(非特許文献7)。 Bispecific antibodies that substitute for the function of F.VIII and their use have been reported as means for solving these problems (Patent Documents 1, 2, 3 and 4). Bispecific antibodies against F.IXa and FX exert F.VIII cofactor function substitution activity and can substitute F.VIII function by positioning both factors in the vicinity (non-F.VIII cofactor function substitution activity). Patent Document 5). One of the antibodies, ACE910 (Emicizumab), which has high F.VIII cofactor function substitution activity, has been confirmed to have excellent pharmacokinetics (long half-life) and tolerability in clinical trials in healthy subjects. (Non-Patent Document 6), in clinical trials in patients with hemophilia A who do not have or carry inhibitors, the number of bleedings significantly increased by administration of ACE910 (Emicizumab) compared to before administration of ACE910 (Emicizumab). Suppression was observed (Non-Patent Document 7).

 血友病B患者の出血に対しては、F.IX製剤が定期投与される。しかしながら、標準的な半減期を有する従来のF.IX製剤は、週2回の静脈内投与が必要であり、頻回投与は患者・家族にとっての負担となっている。さらに、止血管理中に抗F.IX抗体(インヒビター)が発生することがある。その結果、止血管理がしばしば困難となる。F.IXインヒビター患者においては、F.VIIIのインヒビターには見られない特徴的な副反応としてアナフィラキシー症状を示すことが良く知られており問題となっている(非特許文献8)。従って、F.IX製剤と比べて投与の負担が少なく、インヒビターの存在に左右されない薬剤が強く求められていた。 For bleeding in hemophilia B patients, F.IX preparation is regularly administered. However, the conventional F.IX preparation having a standard half-life requires intravenous administration twice a week, and frequent administration is a burden on patients and their families. In addition, anti-F.IX antibodies (inhibitors) may develop during hemostasis management. As a result, hemostasis management is often difficult. It is well known that patients with F.IX inhibitors exhibit anaphylactic symptoms as a characteristic side reaction not seen in F.VIII inhibitors, which is a problem (Non-Patent Document 8). Therefore, there has been a strong demand for a drug that is less burdensome to administer than the F.IX preparation and is not affected by the presence of the inhibitor.

F.XIはF.XIaのプロテアーゼ前駆体であり、F.IXの活性化を通じて止血に寄与している。近年、高濃度のF.XIaをF.Xに加えることで、F.Xのactivation peptideを切断できること、F.Vに加えることでF.Vaを生成することが報告された(非特許文献9)。また、F.IXを遺伝的に欠損させた血友病Bモデルマウスにおいて、過剰量のF.XIa(想定血漿中濃度60 nM)を静脈内投与することによって出血持続時間を減少させることが報告されている(非特許文献10)。従って、過剰量のF.XIあるいはF.XIaを血友病Bの患者に投与することでインヒビターの有無にかかわらず、出血回数を抑制する可能性がある。しかしながら、F.XI製剤はF.XI欠損患者に投与した際に血栓症を起こした報告があり、安全な治療方法として確立されていない(非特許文献11)。これはF.XI製剤がF.XIaを含んでしまうことが原因である可能性があり、F.XIa製剤の投与も安全な治療方法とはいえない。従って、F.XI製剤もしくはF.XIa製剤を過剰投与する以外の方法で安全に血友病患者の出血を防ぐ治療方法が求められていた。 F.XI is a protease precursor of F.XIa and contributes to hemostasis through activation of F.IX. In recent years, it has been reported that the activation peptide of F.X can be cleaved by adding a high concentration of F.XIa to F.X, and that F.Va is produced by adding it to F.V (Non-Patent Document 9). In addition, it was reported that the duration of bleeding was reduced by intravenous administration of an excessive amount of F.XIa (estimated plasma concentration of 60 nM) in hemophilia B model mice genetically deficient in F.IX. (Non-Patent Document 10). Therefore, administration of excess F.XI or F.XIa to patients with hemophilia B may reduce the number of bleedings with or without inhibitors. However, it has been reported that the F.XI preparation causes thrombosis when administered to F.XI-deficient patients, and has not been established as a safe treatment method (Non-Patent Document 11). This may be due to the fact that the F.XIa preparation contains F.XIa, and administration of the F.XIa preparation is not a safe treatment method. Therefore, there has been a demand for a therapeutic method for safely preventing bleeding in hemophiliacs by a method other than overdose of the F.XI preparation or the F.XIa preparation.

WO 2005/035754WO 2005/035754 WO 2005/035756WO 2005/035756 WO 2006/109592WO 2006/109592 WO 2012/067176WO 2012/067176

Blood 58, 1-13 (1981)Blood 58, 1-13 (1981) Nature 312, 330-337(1984)Nature 312, 330-337 (1984) Nature 312, 337-342(1984)Nature 312, 337-342 (1984) Biochim.Biophys.Acta 871, 268-278(1986)Biochim.Biophys.Acta 871, 268-278 (1986) Nat Med. 2012 Oct;18(10):1570-4.Nat Med. 2012 Oct; 18 (10): 1570-4. Blood. 2016, Vol.127, 13: 1633-1641.Blood. 2016, Vol.127, 13: 1633-1641. New Eng J Med 2016 , 374;21, 2044-2053New Eng J Med 2016, 374; 21, 2044-2053 Semin Thromb Hemost. 2018;44(6):578-589.Semin Thromb Hemost. 2018; 44 (6): 578-589. J Thromb Haemost. 2013;11(12):2118-2127.J Thromb Haemost. 2013; 11 (12): 2118-2127. J Thromb Haemost. 2018;16(10):2044-2049.J Thromb Haemost. 2018; 16 (10): 2044-2049. Haemophilia. 2015;21(4):477-480.Haemophilia. 2015; 21 (4): 477-480.

 本開示中の発明は上記のような状況に鑑みてなされたものであり、非限定的な一態様において、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる新規分子の提供を目的とする。 The invention in the present disclosure has been made in view of the above circumstances and is used in a non-limiting aspect for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding. The purpose is to provide new molecules.

 非限定的な一態様において、本発明者らは、鋭意研究の結果、生体内で互いに近接させる補因子が確認されていない2種の血液凝固関連因子を認識する二重特異性抗原結合分子の作製に成功した。また、当該二重特異性抗原結合分子が血液凝固の促進に寄与することを見出した。 In one non-limiting aspect, the present inventors recognize two blood coagulation-related factors in which cofactors that are close to each other in vivo have not been confirmed as a result of diligent research. The production was successful. We also found that the bispecific antigen-binding molecule contributes to the promotion of blood coagulation.

 本開示はこのような知見に基づくものであり、具体的には以下に例示的に記載する実施態様を包含するものである。
〔A〕酵素、及び前記酵素の触媒反応を受け得る基質の両方を認識する二重特異性抗原結合分子、ただし生体に発現するペプチド性補因子またはヘパリンの機能を代替する二重特異性抗原結合分子を除く。
〔B〕酵素、及び前記酵素の触媒反応を受け得る基質の両方を認識する二重特異性抗原結合分子であって、該酵素による該基質の加水分解を促進する〔A〕記載の二重特異性抗原結合分子。
〔C〕酵素、及び前記酵素の触媒反応を受け得る基質の両方を認識する二重特異性抗原結合分子であって、該酵素による該基質の活性化を促進する〔A〕記載の二重特異性抗原結合分子。
〔D〕酵素がプロテアーゼである〔A〕から〔C〕のいずれか一つに記載の二重特異性抗原結合分子。
〔E〕酵素がセリンプロテアーゼである〔A〕から〔C〕のいずれか一つに記載の二重特異性抗原結合分子。
〔F〕酵素が血液凝固系の酵素であり、前記酵素の触媒反応を受けうる基質が血液凝固系の基質である〔A〕から〔E〕のいずれか一つに記載の二重特異性抗原結合分子。
〔G〕生体に発現するペプチド性補因子である血液凝固第V因子、血液凝固第VIII因子、組織因子(TF), トロンボモデュリン(TM)、プロテインS(PS)、プロテインZ(PZ)補体C4b、補体制御タンパクH因子(complement regulatory Factor H)、membrane cofactor Protein(MCP)、complement receptor1(CR1)からなる群から選択される少なくとも一の補因子あるいはヘパリンの機能を代替する二重特異性抗原結合分子を除く、〔A〕から〔F〕のいずれか一つに記載の二重特異性抗原結合分子。
〔H〕二重特異性抗体である、〔A〕から〔G〕のいずれか一つに記載の二重特異性抗原結合分子。
〔1〕以下の(a)~(h)のいずれかに記載の二重特異性抗原結合分子:
(a)活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(b)活性化血液凝固第X因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(c)活性化血液凝固第VII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(d)活性化血液凝固第VII因子-Tissue Factor complexと血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(e)活性化血液凝固第XII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(f)Thrombinと血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(g)活性化血液凝固第XII因子と血液凝固第IX因子の両方を認識する二重特異性抗原結合分子;または
(h)活性化血液凝固第XI因子と血液凝固第XI因子の両方を認識する二重特異性抗原結合分子。
〔1a〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子。
〔2〕〔1〕または〔1a〕に記載の二重特異性抗原結合分子であって、該酵素による該基質の活性化を促進する二重特異性抗原結合分子。
〔2a〕〔1〕または〔1a〕に記載の二重特異性抗原結合分子であって、in vitro 酵素反応測定系において発色基質溶液添加30分後の吸光度が0.1以上を示す二重特異性抗原結合分子。
〔3〕二重特異性抗体である、〔1〕または〔1a〕、〔2〕または〔2a〕に記載の二重特異性抗原結合分子。
〔4〕〔1〕~〔3〕のいずれかに記載の抗原結合分子および薬学的に許容される担体を含む、組成物。
〔5〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる医薬組成物である、〔4〕に記載の組成物。
〔6〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5〕に記載の組成物。
〔7〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Bである、〔6〕に記載の組成物。
〔8〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子に対するインヒビターが出現している疾患である、〔6〕に記載の組成物。
〔9〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5〕に記載の組成物。
〔10〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病A、後天性血友病、またはフォンビルブランド病である、〔9〕に記載の組成物。
〔11〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子に対するインヒビターが出現している疾患である、〔9〕に記載の組成物。
〔12〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5〕に記載の組成物。
〔13〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Cである、〔12〕に記載の組成物。
〔14〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子に対するインヒビターが出現している疾患である、〔12〕に記載の組成物。
〔15〕〔1〕~〔3〕のいずれかの二重特異性抗原結合分子の、〔4〕~〔14〕のいずれかに記載した組成物の製造のための使用。
〔16〕少なくとも〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕に記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのキット。
〔17〕少なくとも〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕に記載の組成物を含み、かつ血液凝固第IX因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第IX因子と併用して、予防および/または治療する方法に用いるためのキット。
〔18〕少なくとも〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕に記載の組成物を含み、かつ血液凝固第VIII因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第VIII因子と併用して、予防および/または治療する方法に用いるためのキット。
〔19〕少なくとも〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕に記載の組成物を含み、かつ血液凝固第XI因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第XI因子と併用して、予防および/または治療する方法に用いるためのキット。
〔20〕少なくとも〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕に記載の組成物を含み、かつ出血、出血を伴う疾患、もしくは出血に起因する疾患を、バイパス製剤と併用して、予防および/または治療する方法に用いるためのキット。
〔21〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子を用いて血液凝固を促進する方法。
〔22〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、スクリーニング方法。
〔23〕さらに以下の工程を含む、〔22〕に記載のスクリーニング方法
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔24〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、品質試験方法。
〔25〕さらに以下の工程を含む、〔24〕に記載の品質試験方法:
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔26〕〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子をコードする核酸。
〔27〕〔26〕に記載の核酸が挿入されたベクター。
〔28〕〔26〕に記載の核酸または〔27〕に記載のベクターを含む細胞。
〔29〕〔28〕に記載の細胞を培養することにより、〔1〕~〔3〕のいずれかに記載の二重特異性抗原結合分子を製造する方法。
〔30〕以下の(i)~(o)のいずれかに記載の二重特異性抗原結合分子:
(i)活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;
(j)活性化血液凝固第X因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;
(k)活性化血液凝固第VII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;
(l)活性化血液凝固第VII因子-Tissue Factor complexと血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;
(m)活性化血液凝固第XII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;
(n)Thrombinと血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子;または
(o)活性化血液凝固第XII因子と血液凝固第IX因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第IX因子を活性化する二重特異性抗原結合分子。
〔30a〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子であって、血液凝固第X因子を活性化する二重特異性抗原結合分子。
〔31〕二重特異性抗体である、〔30〕または〔30a〕記載の二重特異性抗原結合分子。
〔32〕〔30〕、〔30a〕、または〔31〕に記載の抗原結合分子および薬学的に許容される担体を含む、組成物。
〔33〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる医薬組成物である、〔32〕に記載の組成物。
〔34〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33〕に記載の組成物。
〔35〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Bである、〔34〕に記載の組成物。
〔36〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子に対するインヒビターが出現している疾患である、〔34〕に記載の組成物。
〔37〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33〕に記載の組成物。
〔38〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病A、後天性血友病、またはフォンビルブランド病である、〔37〕に記載の組成物。
〔39〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子に対するインヒビターが出現している疾患である、〔37〕に記載の組成物。
〔40〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33〕に記載の組成物。
〔41〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Cである、〔40〕に記載の組成物。
〔42〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子に対するインヒビターが出現している疾患である、〔40〕に記載の組成物。
〔43〕〔30〕または〔31〕の二重特異性抗原結合分子の、〔32〕~〔42〕のいずれかに記載した組成物の製造のための使用。
〔44〕少なくとも〔30〕、〔30a〕、もしくは〔31〕に記載の二重特異性抗原結合分子、または〔32〕に記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのキット。
〔45〕少なくとも〔30〕、〔30a〕、もしくは〔31〕に記載の二重特異性抗原結合分子、または〔32〕に記載の組成物を含み、かつ血液凝固第IX因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第IX因子と併用して、予防および/または治療する方法に用いるためのキット。
〔46〕少なくとも〔30〕、〔30a〕もしくは〔31〕に記載の二重特異性抗原結合分子、または〔32〕に記載の組成物を含み、かつ血液凝固第VIII因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第VIII因子と併用して、予防および/または治療する方法に用いるためのキット。
〔47〕少なくとも〔30〕、〔30a〕もしくは〔31〕に記載の二重特異性抗原結合分子、または〔32〕に記載の組成物を含み、かつ血液凝固第XI因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第XI因子と併用して、予防および/または治療する方法に用いるためのキット。
〔48〕少なくとも〔30〕、〔30a〕もしくは〔31〕に記載の二重特異性抗原結合分子、または〔32〕に記載の組成物を含み、かつ出血、出血を伴う疾患、もしくは出血に起因する疾患を、バイパス製剤と併用して、予防および/または治療する方法に用いるためのキット。
〔49〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子を用いて血液凝固を促進する方法。
〔50〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、スクリーニング方法。
〔51〕さらに以下の工程を含む、〔50〕に記載のスクリーニング方法
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔52〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、品質試験方法。
〔53〕さらに以下の工程を含む、〔52〕に記載の品質試験方法:
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔54〕〔30〕、〔30a〕または〔31〕に記載の二重特異性抗原結合分子をコードする核酸。
〔55〕〔54〕に記載の核酸が挿入されたベクター。
〔56〕〔54〕に記載の核酸または〔55〕に記載のベクターを含む細胞。
〔57〕〔56〕に記載の細胞を培養することにより、〔30〕、〔30a〕、または〔31〕に記載の二重特異性抗原結合分子を製造する方法。
〔58〕〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物を投与する工程を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法。
〔59〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療において用いるための、〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物。
〔60〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防剤および/または治療剤の製造における、〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物の使用。
〔61〕〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防剤および/または治療剤。
〔62〕〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物を投与する工程を含む、血液凝固を促進する方法。
〔63〕血液凝固の促進において用いるための、〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物。
〔64〕血液凝固促進剤の製造における、〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物の使用。
〔65〕〔A〕~〔H〕、〔1〕~〔3〕、および〔30〕~〔31〕のいずれかに記載の二重特異性抗原結合分子、または〔4〕~〔14〕および〔32〕~〔42〕のいずれかに記載の組成物を含む、血液凝固促進剤。
The present disclosure is based on such findings and specifically includes embodiments exemplified below.
[A] A bispecific antigen-binding molecule that recognizes both an enzyme and a substrate that can undergo a catalytic reaction of the enzyme, but a bispecific antigen-binding molecule that substitutes for the function of a peptidic cofactor or heparin expressed in a living body. Excludes molecules.
[B] The bispecific antigen-binding molecule according to [A], which is a bispecific antigen-binding molecule that recognizes both an enzyme and a substrate capable of undergoing a catalytic reaction of the enzyme, and promotes hydrolysis of the substrate by the enzyme. Sex antigen binding molecule.
[C] The bispecific antigen-binding molecule according to [A], which is a bispecific antigen-binding molecule that recognizes both an enzyme and a substrate capable of undergoing a catalytic reaction of the enzyme, and promotes activation of the substrate by the enzyme. Sex antigen binding molecule.
[D] The bispecific antigen-binding molecule according to any one of [A] to [C], wherein the enzyme is a protease.
[E] The bispecific antigen-binding molecule according to any one of [A] to [C], wherein the enzyme is a serine protease.
[F] The bispecific antigen according to any one of [A] to [E], wherein the enzyme is a blood coagulation enzyme and the substrate capable of undergoing a catalytic reaction of the enzyme is a blood coagulation substrate. Binding molecule.
[G] Blood coagulation factor V, blood coagulation factor VIII, tissue factor (TF), thrombomodulin (TM), protein S (PS), protein Z (PZ), which are peptide complements expressed in the living body. A dual alternative to the function of at least one complement or heparin selected from the group consisting of complement C4b, complement regulatory factor H, membrane cofactor protein (MCP), and complement receptor 1 (CR1). The bispecific antigen-binding molecule according to any one of [A] to [F], excluding the specific antigen-binding molecule.
[H] The bispecific antigen-binding molecule according to any one of [A] to [G], which is a bispecific antibody.
[1] The bispecific antigen-binding molecule according to any one of (a) to (h) below:
(a) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X;
(b) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor X and blood coagulation factor X;
(c) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X;
(d) Activated blood coagulation factor VII-A bispecific antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X;
(e) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor X;
(f) A bispecific antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X;
(g) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor IX; or
(h) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor XI.
[1a] A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
[2] The bispecific antigen-binding molecule according to [1] or [1a], which promotes activation of the substrate by the enzyme.
[2a] The bispecific antigen-binding molecule according to [1] or [1a], which exhibits an absorbance of 0.1 or more 30 minutes after the addition of the color-developing substrate solution in an in vitro enzyme reaction measurement system. Sex antigen binding molecule.
[3] The bispecific antigen-binding molecule according to [1] or [1a], [2] or [2a], which is a bispecific antibody.
[4] A composition comprising the antigen-binding molecule according to any one of [1] to [3] and a pharmaceutically acceptable carrier.
[5] The composition according to [4], which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
[6] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [5].
[7] The composition according to [6], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
[8] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [6], which is a disease in which an inhibitor appears.
[9] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [5].
[10] Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease. The composition according to [9].
[11] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [9], which is a disease in which an inhibitor appears.
[12] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [5].
[13] The composition according to [12], wherein the disease that develops and / or progresses due to a decrease or deficiency of the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
[14] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI and / or activation with respect to blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [12], which is a disease in which an inhibitor appears.
[15] Use of the bispecific antigen-binding molecule according to any one of [1] to [3] for producing the composition according to any one of [4] to [14].
[16] Bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4]. A kit for use in methods of preventing and / or treating.
[17] Bleeding or bleeding containing at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4] and containing blood coagulation factor IX. A kit for use in methods of preventing and / or treating associated diseases or diseases caused by bleeding in combination with blood coagulation factor IX.
[18] Bleeding, bleeding containing at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4] and containing blood coagulation factor VIII. A kit for use in methods of preventing and / or treating associated diseases or diseases caused by bleeding in combination with blood coagulation factor VIII.
[19] Bleeding or bleeding containing at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4] and containing blood coagulation factor XI. A kit for use in a method of preventing and / or treating a concomitant disease or a disease caused by bleeding in combination with blood coagulation factor XI.
[20] Containing at least the bispecific antigen-binding molecule according to any one of [1] to [3] or the composition according to [4], and caused by bleeding, a disease accompanied by bleeding, or bleeding. A kit for use in methods of preventing and / or treating a disease in combination with a bypass formulation.
[21] A method for promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
[22] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
[23] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [22], which further comprises the following steps.
[24] A quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
[25] The quality test method according to [24], which further comprises the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
[26] A nucleic acid encoding the bispecific antigen-binding molecule according to any one of [1] to [3].
[27] A vector into which the nucleic acid according to [26] has been inserted.
[28] A cell containing the nucleic acid according to [26] or the vector according to [27].
[29] A method for producing the bispecific antigen-binding molecule according to any one of [1] to [3] by culturing the cells according to [28].
[30] The bispecific antigen-binding molecule according to any one of (i) to (o) below:
(i) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XI and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X;
(j) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor X and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X;
(k) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X;
(l) Activated blood coagulation factor VII-A bispecific antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X, and is a bispecific antigen that activates blood coagulation factor X. Binding molecule;
(m) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X;
(n) A bispecific antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X and activates blood coagulation factor X; or
(o) Activated A bispecific antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor IX, and is a bispecific antigen-binding molecule that activates blood coagulation factor IX.
[30a] A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X, and is a bispecific antigen-binding molecule that activates blood coagulation factor X.
[31] The bispecific antigen-binding molecule according to [30] or [30a], which is a bispecific antibody.
[32] A composition comprising the antigen-binding molecule according to [30], [30a], or [31] and a pharmaceutically acceptable carrier.
[33] The composition according to [32], which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
[34] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [33].
[35] The composition according to [34], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
[36] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [34], which is a disease in which an inhibitor appears.
[37] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [33].
[38] Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease. The composition according to [37].
[39] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [37], which is a disease in which an inhibitor appears.
[40] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [33].
[41] The composition according to [40], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
[42] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI and / or activation are associated with blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [40], which is a disease in which an inhibitor appears.
[43] Use of the bispecific antigen-binding molecule of [30] or [31] for producing the composition according to any one of [32] to [42].
[44] Due to bleeding, a disease associated with bleeding, or bleeding, which comprises at least the bispecific antigen binding molecule according to [30], [30a], or [31], or the composition according to [32]. A kit for use in methods of preventing and / or treating bleeding disorders.
[45] Bleeding, which comprises at least the bispecific antigen-binding molecule according to [30], [30a], or [31], or the composition according to [32], and contains blood coagulation factor IX. A kit for use in methods of preventing and / or treating bleeding-related or bleeding-related disorders in combination with blood coagulation factor IX.
[46] Bleeding, bleeding, comprising at least the bispecific antigen binding molecule according to [30], [30a] or [31], or the composition according to [32], and containing blood coagulation factor VIII. A kit for use in a method of preventing and / or treating a disease associated with blood coagulation or a disease caused by bleeding in combination with blood coagulation factor VIII.
[47] Bleeding, bleeding, comprising at least the bispecific antigen binding molecule according to [30], [30a] or [31], or the composition according to [32], and containing blood coagulation factor XI. A kit for use in a method of preventing and / or treating a disease associated with or caused by bleeding in combination with blood coagulation factor XI.
[48] Containing at least the bispecific antigen-binding molecule according to [30], [30a] or [31], or the composition according to [32], and caused by bleeding, a disease associated with bleeding, or bleeding. A kit for use in methods of preventing and / or treating bleeding disorders in combination with bypass formulations.
[49] A method for promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
[50] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
[51] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [50], which further comprises the following steps.
[52] A quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
[53] The quality test method according to [52], further comprising the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
[54] The nucleic acid encoding the bispecific antigen-binding molecule according to [30], [30a] or [31].
[55] A vector into which the nucleic acid according to [54] has been inserted.
[56] A cell containing the nucleic acid according to [54] or the vector according to [55].
[57] The method for producing the bispecific antigen-binding molecule according to [30], [30a], or [31] by culturing the cells according to [56].
[58] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the step of administering the composition according to any one of [32] to [42].
[59] [A]-[H], [1]-[3], and [30]-[31] for use in the prevention and / or treatment of bleeding, diseases associated with bleeding, or diseases caused by bleeding. ], Or the composition according to any one of [4] to [14] and [32] to [42].
[60] [A]-[H], [1]-[3], and [30]-[in the manufacture of prophylactic and / or therapeutic agents for bleeding, diseases associated with bleeding, or diseases caused by bleeding. Use of the bispecific antigen-binding molecule according to any one of 31] or the composition according to any one of [4] to [14] and [32] to [42].
[61] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A prophylactic and / or therapeutic agent for bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the composition according to any one of [32] to [42].
[62] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A method for promoting blood coagulation, which comprises the step of administering the composition according to any one of [32] to [42].
[63] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31] for use in promoting blood coagulation. Alternatively, the composition according to any one of [4] to [14] and [32] to [42].
[64] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31] in the production of a blood coagulation promoter, or Use of the composition according to any one of [4] to [14] and [32] to [42].
[65] The bispecific antigen-binding molecule according to any one of [A] to [H], [1] to [3], and [30] to [31], or [4] to [14] and A blood coagulation promoter comprising the composition according to any one of [32] to [42].

〔1b〕以下の(a)~(h)のいずれかに記載の抗原結合分子:
(a)活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する抗原結合分子;
(b)活性化血液凝固第X因子と血液凝固第X因子の両方を認識する抗原結合分子;
(c)活性化血液凝固第VII因子と血液凝固第X因子の両方を認識する抗原結合分子;
(d)活性化血液凝固第VII因子-Tissue Factor complexと血液凝固第X因子の両方を認識する抗原結合分子;
(e)活性化血液凝固第XII因子と血液凝固第X因子の両方を認識する抗原結合分子;
(f)Thrombinと血液凝固第X因子の両方を認識する抗原結合分子;
(g)活性化血液凝固第XII因子と血液凝固第IX因子の両方を認識する抗原結合分子;または
(h)活性化血液凝固第XI因子と血液凝固第XI因子の両方を認識する抗原結合分子。
〔2b〕〔1b〕に記載の抗原結合分子であって、該酵素による該基質の活性化を促進する抗原結合分子。
〔2c〕〔1b〕に記載の抗原結合分子であって、in vitro 酵素反応測定系において発色基質溶液添加30分後の吸光度が0.1以上を示す抗原結合分子。
〔3b〕抗体である、〔1b〕、〔2b〕または〔2c〕に記載の抗原結合分子。
〔4b〕〔1b〕~〔3b〕のいずれかに記載の抗原結合分子および薬学的に許容される担体を含む、組成物。
〔5b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる医薬組成物である、〔4b〕に記載の組成物。
〔6b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5b〕に記載の組成物。
〔7b〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Bである、〔6b〕に記載の組成物。
〔8b〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子に対するインヒビターが出現している疾患である、〔6b〕に記載の組成物。
〔9b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5b〕に記載の組成物。
〔10b〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病A、後天性血友病、またはフォンビルブランド病である、〔9b〕に記載の組成物。
〔11b〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子に対するインヒビターが出現している疾患である、〔9b〕に記載の組成物。
〔12b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔5b〕に記載の組成物。
〔13b〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Cである、〔12b〕に記載の組成物。
〔14b〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子に対するインヒビターが出現している疾患である、〔12b〕に記載の組成物。
〔15b〕〔1b〕~〔3b〕のいずれかの抗原結合分子の、〔4b〕~〔14b〕のいずれかに記載した組成物の製造のための使用。
〔16b〕少なくとも〔1b〕~〔3b〕のいずれかに記載の抗原結合分子、または〔4b〕に記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのキット。
〔17b〕少なくとも〔1b〕~〔3b〕のいずれかに記載の抗原結合分子、または〔4b〕に記載の組成物を含み、かつ血液凝固第IX因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第IX因子と併用して、予防および/または治療する方法に用いるためのキット。
〔18b〕少なくとも〔1b〕~〔3b〕のいずれかに記載の抗原結合分子、または〔4b〕に記載の組成物を含み、かつ血液凝固第VIII因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第VIII因子と併用して、予防および/または治療する方法に用いるためのキット。
〔19b〕少なくとも〔1b〕~〔3b〕のいずれかに記載の抗原結合分子、または〔4b〕に記載の組成物を含み、かつ血液凝固第XI因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第XI因子と併用して、予防および/または治療する方法に用いるためのキット。
〔20b〕少なくとも〔1b〕~〔3b〕のいずれかに記載の抗原結合分子、または〔4b〕に記載の組成物を含み、かつ出血、出血を伴う疾患、もしくは出血に起因する疾患を、バイパス製剤と併用して、予防および/または治療する方法に用いるためのキット。
〔21b〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する抗原結合分子を用いて血液凝固を促進する方法。
〔22b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、スクリーニング方法。
〔23b〕さらに以下の工程を含む、〔22b〕に記載のスクリーニング方法
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔24b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、品質試験方法。
〔25b〕さらに以下の工程を含む、〔24b〕に記載の品質試験方法:
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔26b〕〔1b〕~〔3b〕のいずれかに記載の抗原結合分子をコードする核酸。
〔27b〕〔26b〕に記載の核酸が挿入されたベクター。
〔28b〕〔26b〕に記載の核酸または〔27b〕に記載のベクターを含む細胞。
〔29b〕〔28b〕に記載の細胞を培養することにより、〔1b〕~〔3b〕のいずれかに記載の抗原結合分子を製造する方法。
〔30b〕以下の(i)~(o)のいずれかに記載の抗原結合分子:
(i)活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;
(j)活性化血液凝固第X因子と血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;
(k)活性化血液凝固第VII因子と血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;
(l)活性化血液凝固第VII因子-Tissue Factor complexと血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;
(m)活性化血液凝固第XII因子と血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;
(n)Thrombinと血液凝固第X因子の両方を認識する抗原結合分子であって、血液凝固第X因子を活性化する抗原結合分子;または
(o)活性化血液凝固第XII因子と血液凝固第IX因子の両方を認識する抗原結合分子であって、血液凝固第IX因子を活性化する抗原結合分子。
〔31b〕抗体である、〔30b〕記載の抗原結合分子。
〔32b〕〔30b〕または〔31b〕に記載の抗原結合分子および薬学的に許容される担体を含む、組成物。
〔33b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる医薬組成物である、〔32b〕に記載の組成物。
〔34b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33b〕に記載の組成物。
〔35b〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Bである、〔34b〕に記載の組成物。
〔36b〕血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子に対するインヒビターが出現している疾患である、〔34b〕に記載の組成物。
〔37b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33b〕に記載の組成物。
〔38b〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病A、後天性血友病、またはフォンビルブランド病である、〔37b〕に記載の組成物。
〔39b〕血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子に対するインヒビターが出現している疾患である、〔37b〕に記載の組成物。
〔40b〕出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患である、〔33b〕に記載の組成物。
〔41b〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Cである、〔40b〕に記載の組成物。
〔42b〕血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子に対するインヒビターが出現している疾患である、〔40b〕に記載の組成物。
〔43b〕〔30b〕または〔31b〕の抗原結合分子の、〔32b〕~〔42b〕のいずれかに記載した組成物の製造のための使用。
〔44b〕少なくとも〔30b〕もしくは〔31b〕に記載の抗原結合分子、または〔32b〕に記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのキット。
〔45b〕少なくとも〔30b〕もしくは〔31b〕に記載の抗原結合分子、または〔32b〕に記載の組成物を含み、かつ血液凝固第IX因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第IX因子と併用して、予防および/または治療する方法に用いるためのキット。
〔46b〕少なくとも〔30b〕もしくは〔31b〕に記載の抗原結合分子、または〔32b〕に記載の組成物を含み、かつ血液凝固第VIII因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第VIII因子と併用して、予防および/または治療する方法に用いるためのキット。
〔47b〕少なくとも〔30b〕もしくは〔31b〕に記載の抗原結合分子、または〔32b〕に記載の組成物を含み、かつ血液凝固第XI因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第XI因子と併用して、予防および/または治療する方法に用いるためのキット。
〔48b〕少なくとも〔30b〕もしくは〔31b〕に記載の抗原結合分子、または〔32b〕に記載の組成物を含み、かつ出血、出血を伴う疾患、もしくは出血に起因する疾患を、バイパス製剤と併用して、予防および/または治療する方法に用いるためのキット。
〔49b〕活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する抗原結合分子を用いて血液凝固を促進する方法。
〔50b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、スクリーニング方法。
〔51b〕さらに以下の工程を含む、〔50b〕に記載のスクリーニング方法
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔52b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、品質試験方法。
〔53b〕さらに以下の工程を含む、〔52b〕に記載の品質試験方法:
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
〔54b〕〔30b〕または〔31b〕に記載の抗原結合分子をコードする核酸。
〔55b〕〔54b〕に記載の核酸が挿入されたベクター。
〔56b〕〔54b〕に記載の核酸または〔55b〕に記載のベクターを含む細胞。
〔57b〕〔56b〕に記載の細胞を培養することにより、〔30b〕または〔31b〕に記載の抗原結合分子を製造する方法。
〔58b〕〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物を投与する工程を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法。
〔59b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療において用いるための、〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物。
〔60b〕出血、出血を伴う疾患、もしくは出血に起因する疾患の予防剤および/または治療剤の製造における、〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物の使用。
〔61b〕〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防剤および/または治療剤。
〔62b〕〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物を投与する工程を含む、血液凝固を促進する方法。
〔63b〕血液凝固の促進において用いるための、〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物。
〔64b〕血液凝固促進剤の製造における、〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物の使用。
〔65b〕〔1b〕~〔3b〕、および〔30b〕~〔31b〕のいずれかに記載の抗原結合分子、または〔4b〕~〔14b〕および〔32b〕~〔42b〕のいずれかに記載の組成物を含む、血液凝固促進剤。
[1b] The antigen-binding molecule according to any one of (a) to (h) below:
(a) Antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X;
(b) Antigen-binding molecule that recognizes both activated blood coagulation factor X and blood coagulation factor X;
(c) Antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X;
(d) Activated blood coagulation factor VII-An antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X;
(e) Antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor X;
(f) Antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X;
(g) Antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor IX; or
(h) An antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor XI.
[2b] The antigen-binding molecule according to [1b], which promotes activation of the substrate by the enzyme.
[2c] The antigen-binding molecule according to [1b], which has an absorbance of 0.1 or more 30 minutes after the addition of the color-developing substrate solution in an in vitro enzyme reaction measurement system.
[3b] The antigen-binding molecule according to [1b], [2b] or [2c], which is an antibody.
[4b] A composition comprising the antigen-binding molecule according to any one of [1b] to [3b] and a pharmaceutically acceptable carrier.
[5b] The composition according to [4b], which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
[6b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [5b].
[7b] The composition according to [6b], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
[8b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [6b], which is a disease in which an inhibitor appears.
[9b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [5b].
[10b] Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease. The composition according to [9b].
[11b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [9b], which is a disease in which an inhibitor appears.
[12b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [5b].
[13b] The composition according to [12b], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
[14b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI and / or activation are associated with blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [12b], which is a disease in which an inhibitor appears.
[15b] Use of any of the antigen-binding molecules of [1b] to [3b] for producing the composition according to any one of [4b] to [14b].
[16b] Preventing and / or bleeding, bleeding-related diseases, or diseases caused by bleeding, which contain at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b]. Or a kit for use in therapeutic methods.
[17b] Bleeding, a disease associated with bleeding, or a disease containing blood coagulation factor IX and containing at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b]. A kit for use in methods of preventing and / or treating diseases caused by bleeding in combination with blood coagulation factor IX.
[18b] Bleeding, a disease associated with bleeding, or a disease containing at least the antigen-binding molecule according to any one of [1b] to [3b], or the composition according to [4b], and containing blood coagulation factor VIII. A kit for use in methods of preventing and / or treating diseases caused by bleeding in combination with blood coagulation factor VIII.
[19b] Bleeding, a disease associated with bleeding, or a disease containing blood coagulation factor XI and containing at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b]. A kit for use in methods of preventing and / or treating diseases caused by bleeding in combination with blood coagulation factor XI.
[20b] Bypasses bleeding, diseases associated with bleeding, or diseases caused by bleeding, which contain at least the antigen-binding molecule according to any one of [1b] to [3b] or the composition according to [4b]. A kit for use in prophylactic and / or therapeutic methods in combination with formulations.
[21b] A method for promoting blood coagulation using an antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
[22b] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
[23b] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [22b], which further comprises the following steps.
[24b] A quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
[25b] The quality test method according to [24b], further comprising the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
[26b] A nucleic acid encoding the antigen-binding molecule according to any one of [1b] to [3b].
[27b] A vector into which the nucleic acid according to [26b] has been inserted.
[28b] A cell containing the nucleic acid according to [26b] or the vector according to [27b].
[29b] A method for producing the antigen-binding molecule according to any one of [1b] to [3b] by culturing the cells according to [28b].
[30b] The antigen-binding molecule according to any one of (i) to (o) below:
(i) Activated antigen-binding molecule that recognizes both blood coagulation factor XI and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X;
(j) Activated antigen-binding molecule that recognizes both blood coagulation factor X and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X;
(k) Activated antigen-binding molecule that recognizes both blood coagulation factor VII and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X;
(l) Activated blood coagulation factor VII-An antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X, and is an antigen-binding molecule that activates blood coagulation factor X;
(m) Activated antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor X, and activates blood coagulation factor X;
(n) An antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X and activates blood coagulation factor X; or
(o) Activated antigen-binding molecule that recognizes both blood coagulation factor XII and blood coagulation factor IX, and is an antigen-binding molecule that activates blood coagulation factor IX.
[31b] The antigen-binding molecule according to [30b], which is an antibody.
[32b] A composition comprising the antigen-binding molecule according to [30b] or [31b] and a pharmaceutically acceptable carrier.
[33b] The composition according to [32b], which is a pharmaceutical composition used for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
[34b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [33b].
[35b] The composition according to [34b], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation of blood coagulation factor IX is hemophilia B.
[36b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX are associated with blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to [34b], which is a disease in which an inhibitor appears.
[37b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [33b].
[38b] Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or Fonville brand disease. The composition according to [37b].
[39b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII are associated with blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to [37b], which is a disease in which an inhibitor appears.
[40b] Bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [33b].
[41b] The composition according to [40b], wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.
[42b] Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI and / or activation are associated with blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to [40b], which is a disease in which an inhibitor appears.
[43b] Use of the antigen-binding molecule of [30b] or [31b] for producing the composition according to any one of [32b] to [42b].
[44b] Prevents and / or treats bleeding, bleeding-related diseases, or diseases caused by bleeding, which comprises at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b]. Kit for use in the method.
[45b] Due to bleeding, bleeding-related disease, or bleeding, comprising at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b], and containing blood coagulation factor IX. A kit for use in methods of preventing and / or treating bleeding disorders in combination with blood coagulation factor IX.
[46b] Due to bleeding, bleeding-related disease, or bleeding, comprising at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b], and containing blood coagulation factor VIII. A kit for use in methods of preventing and / or treating bleeding disorders in combination with blood coagulation factor VIII.
[47b] Caused by bleeding, a disease associated with bleeding, or bleeding, which comprises at least the antigen-binding molecule according to [30b] or [31b], or the composition according to [32b] and contains blood coagulation factor XI. A kit for use in methods of preventing and / or treating bleeding disorders in combination with factor XI for blood coagulation.
[48b] A bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises at least the antigen-binding molecule described in [30b] or [31b] or the composition described in [32b], is used in combination with a bypass preparation. And a kit for use in prophylactic and / or therapeutic methods.
[49b] A method for promoting blood coagulation using an antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.
[50b] A method for screening a substance effective for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
[51b] A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using the screening method (3) test substance according to [50b], which further comprises the following steps.
[52b] A quality test method for a substance or composition effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.
A quality test method including (1) a step of evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) a step of evaluating the binding between the test substance and blood coagulation factor X.
[53b] The quality test method according to [52b], further comprising the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
[54b] A nucleic acid encoding the antigen-binding molecule according to [30b] or [31b].
[55b] A vector into which the nucleic acid according to [54b] has been inserted.
[56b] A cell containing the nucleic acid according to [54b] or the vector according to [55b].
[57b] A method for producing the antigen-binding molecule according to [30b] or [31b] by culturing the cells according to [56b].
[58b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b]. A method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the step of administering the composition of.
[59b] The antigen according to any one of [1b] to [3b] and [30b] to [31b] for use in the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding. The binding molecule or the composition according to any one of [4b] to [14b] and [32b] to [42b].
[60b] The description of any of [1b] to [3b] and [30b] to [31b] in the manufacture of a prophylactic and / or therapeutic agent for bleeding, a disease associated with bleeding, or a disease caused by bleeding. Use of an antigen-binding molecule or the composition according to any of [4b]-[14b] and [32b]-[42b].
[61b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b]. A prophylactic and / or therapeutic agent for bleeding, bleeding-related diseases, or diseases caused by bleeding, which comprises the composition of.
[62b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b]. A method of promoting blood coagulation, which comprises the step of administering the composition of.
[63b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b] for use in promoting blood coagulation, or [4b] to [14b] and [32b]. The composition according to any one of [42b].
[64b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b] in the production of a blood coagulation promoter, or [4b] to [14b] and [32b] to. Use of the composition according to any one of [42b].
[65b] The antigen-binding molecule according to any one of [1b] to [3b] and [30b] to [31b], or any of [4b] to [14b] and [32b] to [42b]. A blood coagulation promoter, which comprises the composition of.

 非限定的な一態様において、本開示の二重特異性抗原結合分子、組成物、およびキットは、血液凝固の促進に寄与し、ひいては出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有用であり得る。 In one non-limiting aspect, the bispecific antigen-binding molecules, compositions, and kits of the present disclosure contribute to the promotion of blood coagulation and thus prevent bleeding, bleeding-related diseases, or diseases caused by bleeding. And / or may be useful for treatment.

本発明の基本コンセプトを図示した。ペプチド性補因子が生体に存在しないような関係性である酵素Aと基質Bはそのままでは反応がほとんど進行しない(上図)。しかし、該酵素Aと基質Bを認識するような二重特異性抗原結合分子を添加することで、酵素Aによる基質Bの加水分解反応などの酵素反応を促進することが出来る(下図)。The basic concept of the present invention is illustrated. Enzyme A and substrate B, which are related so that peptide cofactors do not exist in the living body, hardly proceed with the reaction as they are (upper figure). However, by adding a bispecific antigen-binding molecule that recognizes the enzyme A and the substrate B, it is possible to promote an enzymatic reaction such as a hydrolysis reaction of the substrate B by the enzyme A (see the figure below). 血液凝固カスケードの模式図(内因系・外因系)を示した。A schematic diagram (intrinsic system / extrinsic system) of the blood coagulation cascade is shown. In vitro酵素反応系における、抗F.XIa、F.X二重特異性抗体の、F.XIaによるF.X活性化促進活性の測定結果を示した。作製した抗F.XIa、F.X二重特異性抗体は、F.XIaによるF.X活性化促進活性を示した。The measurement results of the F.X activation promoting activity of anti-F.XIa and F.X bispecific antibodies by F.XIa in the in vitro enzyme reaction system are shown. The prepared anti-F.XIa and F.X bispecific antibodies showed F.X activation promoting activity by F.XIa. F.IX欠乏血漿における凝固時間(APTT)を示した。作製した抗F.XIa、F.X二重特異性抗体は同血漿の凝固促進活性を示した。The coagulation time (APTT) in F.IX-deficient plasma is shown. The prepared anti-F.XIa and F.X bispecific antibodies showed the coagulation-promoting activity of the plasma. In vitro酵素反応系における、抗F.XIa、F.X二重特異性抗体の、F.XIaによるF.X活性化促進活性の測定結果を示した。作製した抗F.XIa、F.X二重特異性抗体は、F.XIaによるF.X活性化促進活性を示した。The measurement results of the F.X activation promoting activity of anti-F.XIa and F.X bispecific antibodies by F.XIa in the in vitro enzyme reaction system are shown. The prepared anti-F.XIa and F.X bispecific antibodies showed F.X activation promoting activity by F.XIa. F.IX欠乏血漿における凝固時間(APTT)を示した。作製した抗F.XIa、F.X二重特異性抗体は同血漿の凝固促進活性を示した。The coagulation time (APTT) in F.IX-deficient plasma is shown. The prepared anti-F.XIa and F.X bispecific antibodies showed the coagulation-promoting activity of the plasma.

I.定義
 本明細書で用語「抗原結合分子」は、その最も広い意味において、抗原決定基(エピトープ)に特異的に結合する分子を指す。一態様において、抗原結合分子は、抗体、抗体断片、または抗体誘導体である。一態様において、抗原結合分子は、非抗体タンパク質、またはその断片、もしくはその誘導体である。
I. Definitions The term "antigen-binding molecule" as used herein refers to a molecule that specifically binds to an antigenic determinant (epitope) in its broadest sense. In one embodiment, the antigen binding molecule is an antibody, antibody fragment, or antibody derivative. In one embodiment, the antigen-binding molecule is a non-antibody protein, or a fragment thereof, or a derivative thereof.

 本発明における二重特異性抗原結合分子は、異なる抗原またはエピトープに対して特異性を有する2種類の抗原結合ドメインを含む抗原結合分子である。一態様において、本発明における二重特異性抗原結合分子は二重特異性抗体である。二重特異性抗体は特に制限されないが、モノクローナル抗体であることが好ましい。 The bispecific antigen-binding molecule in the present invention is an antigen-binding molecule containing two types of antigen-binding domains having specificity for different antigens or epitopes. In one aspect, the bispecific antigen-binding molecule in the present invention is a bispecific antibody. The bispecific antibody is not particularly limited, but is preferably a monoclonal antibody.

 本明細書において「抗原結合ドメイン」とは、抗原の一部または全部に特異的に結合し且つ相補的である領域をいう。本明細書において、抗原結合分子は抗原結合ドメインを含んで成る。抗原の分子量が大きい場合、抗原結合ドメインは抗原の特定部分にのみ結合することができる。当該特定部分はエピトープと呼ばれる。一態様において、抗原結合ドメインは特定の抗原に結合する抗体断片を含む。抗原結合ドメインは一または複数の抗体の可変ドメインより提供され得る。非限定的な一態様において、抗原結合ドメインは抗体軽鎖可変領域(VL)と抗体重鎖可変領域(VH)とを含む。こうした抗原結合ドメインの例としては、「scFv(single chain Fv)」、「単鎖抗体(single chain antibody)」、「Fv」、「scFv2(single chain Fv 2)」、「Fab」または「Fab'」等が挙げられる。別の態様において、抗原結合ドメインは特定の抗原に結合する非抗体タンパク質またはその断片を含む。特定の態様において、抗原結合ドメインはヒンジ領域を含む。 As used herein, the term "antigen binding domain" refers to a region that specifically binds to and is complementary to a part or all of an antigen. As used herein, an antigen-binding molecule comprises an antigen-binding domain. When the molecular weight of an antigen is large, the antigen-binding domain can bind only to a specific portion of the antigen. The specific portion is called an epitope. In one aspect, the antigen binding domain comprises an antibody fragment that binds to a particular antigen. Antigen binding domains can be provided by variable domains of one or more antibodies. In one non-limiting aspect, the antigen binding domain comprises an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH). Examples of such antigen-binding domains are "scFv (single chain Fv)", "single chain antibody", "Fv", "scFv2 (single chain Fv 2)", "Fab" or "Fab' , Etc. can be mentioned. In another embodiment, the antigen binding domain comprises a non-antibody protein or fragment thereof that binds to a particular antigen. In certain embodiments, the antigen binding domain comprises a hinge region.

 本明細書において「特異的に結合する」とは、特異的に結合する分子の一方の分子がその一または複数の結合する相手方の分子以外の分子に対しては何ら有意な結合を示さない状態で結合することをいう。また、抗原結合ドメインが、ある抗原中に含まれる複数のエピトープのうち特定のエピトープに対して特異的である場合にも用いられる。また、抗原結合ドメインが結合するエピトープが複数の異なる抗原に含まれる場合には、当該抗原結合ドメインを有する抗原結合分子は当該エピトープを含む様々な抗原と結合することができる。 As used herein, the term "specifically bound" means a state in which one molecule of a specifically bound molecule does not show any significant binding to a molecule other than the other molecule to which one or more of the specifically bound molecules are bound. It means to combine with. It is also used when the antigen-binding domain is specific for a specific epitope among a plurality of epitopes contained in a certain antigen. When the epitope to which the antigen-binding domain binds is contained in a plurality of different antigens, the antigen-binding molecule having the antigen-binding domain can bind to various antigens including the epitope.

 本明細書で用語「抗体」は、最も広い意味で使用され、所望の抗原結合活性を示す限りは、これらに限定されるものではないが、モノクローナル抗体、ポリクローナル抗体、多重特異性抗体(例えば、二重特異性抗体)、抗体断片および抗体修飾物を含む、種々の抗体構造を包含する。 As used herein, the term "antibody" is used in the broadest sense, and is not limited to, but is not limited to, a monoclonal antibody, a polyclonal antibody, and a multispecific antibody (for example, as long as it exhibits a desired antigen-binding activity). Includes a variety of antibody structures, including bispecific antibodies), antibody fragments and antibody modifications.

 「結合活性(binding activity)」は、分子(例えば、抗体)の1個またはそれ以上の結合部位と、分子の結合パートナー(例えば、抗原)との間の、非共有結合的な相互作用の合計の強度のことをいう。ここで、結合活性は、ある結合対のメンバー(例えば、抗体と抗原)の間の1:1相互作用に厳密に限定されない。例えば、結合対のメンバーが1価での1:1相互作用を反映する場合、結合活性は固有の結合アフィニティ(「アフィニティ」)のことをいう。結合対のメンバーが、1価での結合および多価での結合の両方が可能である場合、結合活性は、これらの結合力の総和となる。分子XのそのパートナーYに対する結合活性は、一般的に、解離定数 (KD) または「単位リガンド量当たりのアナライト結合量」により表すことができる。結合活性は、本明細書に記載のものを含む、当該技術分野において知られた通常の方法によって測定され得る。 "Binding activity" is the sum of non-covalent interactions between one or more binding sites of a molecule (eg, an antibody) and a molecule's binding partner (eg, an antigen). It refers to the strength of. Here, the binding activity is not strictly limited to a 1: 1 interaction between members of a binding pair (eg, antibody and antigen). For example, when the members of a binding pair reflect a 1: 1 interaction in monovalent, the binding activity refers to a unique binding affinity (“affinity”). If the members of the binding pair are capable of both monovalent and multivalent binding, the binding activity is the sum of these binding forces. The binding activity of the molecule X to its partner Y can generally be expressed by the dissociation constant (KD) or the "analyte binding amount per unit ligand amount". Binding activity can be measured by conventional methods known in the art, including those described herein.

 本明細書でいう用語「モノクローナル抗体」は、実質的に均一な抗体の集団から得られる抗体のことをいう。すなわち、その集団を構成する個々の抗体は、生じ得る変異抗体(例えば、自然に生じる変異を含む変異抗体、またはモノクローナル抗体調製物の製造中に発生する変異抗体。そのような変異体は通常若干量存在している。)を除いて、同一でありおよび/または同じエピトープに結合する。異なる決定基(エピトープ)に対する異なる抗体を典型的に含むポリクローナル抗体調製物とは対照的に、モノクローナル抗体調製物の各モノクローナル抗体は、抗原上の単一の決定基に対するものである。したがって、修飾語「モノクローナル」は、実質的に均一な抗体の集団から得られるものである、という抗体の特徴を示し、何らかの特定の方法による抗体の製造を求めるものと解釈されるべきではない。例えば、本発明にしたがって用いられるモノクローナル抗体は、これらに限定されるものではないが、ハイブリドーマ法、組換えDNA法、ファージディスプレイ法、ヒト免疫グロブリン遺伝子座の全部または一部を含んだトランスジェニック動物を利用する方法を含む、様々な手法によって作成されてよく、モノクローナル抗体を作製するためのそのような方法および他の例示的な方法は、本明細書に記載されている。 The term "monoclonal antibody" as used herein refers to an antibody obtained from a substantially uniform population of antibodies. That is, the individual antibodies that make up the population are possible mutant antibodies (eg, mutant antibodies that contain naturally occurring mutations, or mutant antibodies that occur during the production of monoclonal antibody preparations, such variants are usually few. Except for the amount present), it binds to the same and / or the same epitope. In contrast to polyclonal antibody preparations, which typically contain different antibodies against different determinants (epitopes), each monoclonal antibody in the monoclonal antibody preparation is for a single determinant on the antigen. Therefore, the modifier "monoclonal" should not be construed as requiring the production of an antibody by any particular method, indicating the characteristic of the antibody that it is obtained from a substantially homogeneous population of antibodies. For example, the monoclonal antibody used according to the present invention is not limited to these, but is a hybridoma method, a recombinant DNA method, a phage display method, and a transgenic animal containing all or a part of the human immunoglobulin locus. It may be made by a variety of methods, including methods that utilize, such methods and other exemplary methods for making monoclonal antibodies are described herein.

 「天然型抗体」は、天然に生じる様々な構造を伴う免疫グロブリン分子のことをいう。例えば、天然型IgG抗体は、ジスルフィド結合している2つの同一の軽鎖と2つの同一の重鎖から構成される約150,000ダルトンのヘテロ四量体糖タンパク質である。N末端からC末端に向かって、各重鎖は、可変重鎖ドメインまたは重鎖可変ドメインとも呼ばれる可変領域 (VH) を有し、それに3つの定常ドメイン(CH1、CH2、およびCH3)が続く。同様に、N末端からC末端に向かって、各軽鎖は、可変軽鎖ドメインまたは軽鎖可変ドメインとも呼ばれる可変領域 (VL) を有し、それに定常軽鎖 (CL) ドメインが続く。抗体の軽鎖は、その定常ドメインのアミノ酸配列に基づいて、カッパ(κ)およびラムダ(λ)と呼ばれる、2つのタイプの1つに帰属させられてよい。 "Natural antibody" refers to an immunoglobulin molecule with various naturally occurring structures. For example, a native IgG antibody is a heterotetrameric glycoprotein of approximately 150,000 daltons composed of two identical disulfide-bonded light chains and two identical heavy chains. From the N-terminus to the C-terminus, each heavy chain has a variable region (VH), also known as a variable heavy chain domain or heavy chain variable domain, followed by three constant domains (CH1, CH2, and CH3). Similarly, from the N-terminus to the C-terminus, each light chain has a variable region (VL), also called a variable light chain domain or a light chain variable domain, followed by a stationary light chain (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain.

 用語「キメラ」抗体は、重鎖および/または軽鎖の一部分が特定の供給源または種に由来する一方で、重鎖および/または軽鎖の残りの部分が異なった供給源または種に由来する抗体のことをいう。 The term "chimeric" antibody is one in which a portion of the heavy chain and / or light chain is derived from a particular source or species, while the rest of the heavy chain and / or light chain is derived from a different source or species. It refers to an antibody.

 抗体の「クラス」は、抗体の重鎖に備わる定常ドメインまたは定常領域のタイプのことをいう。抗体には5つの主要なクラスがある:IgA、IgD、IgE、IgG、およびIgMである。そして、このうちいくつかはさらにサブクラス(アイソタイプ)に分けられてもよい。例えば、IgG1、IgG2、IgG3、IgG4、IgA1、およびIgA2である。異なるクラスの免疫グロブリンに対応する重鎖定常ドメインを、それぞれ、α、δ、ε、γ、およびμと呼ぶ。 The "class" of an antibody refers to the type of constant domain or constant region in the heavy chain of an antibody. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM. And some of them may be further divided into subclasses (isotypes). For example, IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. Heavy chain constant domains corresponding to different classes of immunoglobulins are referred to as α, δ, ε, γ, and μ, respectively.

 本発明の一つの実施態様としての定常領域とは、好ましくは抗体定常領域であり、より好ましくはIgG1、IgG2、IgG3、IgG4型の抗体定常領域であり、さらにより好ましくはヒトIgG1、IgG2、IgG3、IgG4型の抗体定常領域である。また本発明の別の一つの実施態様としての定常領域とは、好ましくは重鎖定常領域であり、より好ましくはIgG1、IgG2、IgG3、IgG4型の重鎖定常領域であり、さらにより好ましくはヒトIgG1、IgG2、IgG3、IgG4型の重鎖定常領域である。ヒトIgG1定常領域、ヒトIgG2定常領域、ヒトIgG3定常領域およびヒトIgG4定常領域のアミノ酸配列は公知である。ヒトIgG1、ヒトIgG2、ヒトIgG3、ヒトIgG4抗体の定常領域としては、遺伝子多型による複数のアロタイプ配列がSequences of proteins of immunological interest, NIH Publication No.91-3242に記載されているが、本発明においてはそのいずれであっても良い。なお本発明のアミノ酸が改変された定常領域は、本発明のアミノ酸変異を含むものである限り、他のアミノ酸変異や修飾を含んでもよい。 The constant region as one embodiment of the present invention is preferably an antibody constant region, more preferably an IgG1, IgG2, IgG3, IgG4 type antibody constant region, and even more preferably a human IgG1, IgG2, IgG3. , IgG4 type antibody constant region. Further, the constant region as another embodiment of the present invention is preferably a heavy chain constant region, more preferably an IgG1, IgG2, IgG3, IgG4 type heavy chain constant region, and even more preferably a human. It is a heavy chain constant region of IgG1, IgG2, IgG3, and IgG4 types. The amino acid sequences of human IgG1 constant region, human IgG2 constant region, human IgG3 constant region and human IgG4 constant region are known. As constant regions of human IgG1, human IgG2, human IgG3, and human IgG4 antibodies, a plurality of allotype sequences due to gene polymorphisms are described in Sequences of proteins of immunological interest, NIH Publication No. 91-3242. In any of these cases. The constant region in which the amino acid of the present invention is modified may contain other amino acid mutations and modifications as long as it contains the amino acid mutation of the present invention.

 「ヒンジ領域」という用語は、野生型抗体重鎖においてCH1ドメインおよびCH2ドメインを連結する、例えばEUナンバリングシステムによれば216位あたりから230位あたりまでの、またはKabatナンバリングシステムによれば226位あたりから243位あたりまでの、抗体重鎖ポリペプチド部分を意味する。天然型IgG抗体において、ヒンジ領域におけるEUナンバリング220位のシステイン残基は、抗体軽鎖における214位のシステイン残基とジスルフィド結合を形成することが知られている。さらに、2つの抗体重鎖の間では、ヒンジ領域におけるEUナンバリング226位のシステイン残基どうし、および229位のシステイン残基どうしがジスルフィド結合を形成することが知られている。本明細書におけるヒンジ領域は、野生型のほか、野生型においてアミノ酸残基の置換、付加、または欠失させた改変体も包含する。 The term "hinge region" refers to linking the CH1 and CH2 domains in a wild-type antibody heavy chain, eg, from around 216 to 230 according to the EU numbering system, or around 226 according to the Kabat numbering system. It means the antibody heavy chain polypeptide portion from to the 243rd position. In a native IgG antibody, the cysteine residue at position 220 of the EU numbering in the hinge region is known to form a disulfide bond with the cysteine residue at position 214 in the antibody light chain. Furthermore, it is known that between the two antibody heavy chains, the cysteine residue at EU numbering 226 and the cysteine residue at 229 in the hinge region form a disulfide bond. The hinge region herein includes the wild-type as well as variants in which amino acid residues have been substituted, added, or deleted in the wild-type.

 本明細書で用語「Fc領域」は、少なくとも定常領域の一部分を含む免疫グロブリン重鎖のC末端領域を定義するために用いられる。この用語は、天然型配列のFc領域および変異体Fc領域を含む。一態様において、ヒトIgG重鎖Fc領域はCys226から、またはPro230から、重鎖のカルボキシル末端まで延びる。ただし、Fc領域のC末端のリジン (Lys447) またはグリシン‐リジン(Gly446-Lys447)は、存在していてもしていなくてもよい。本明細書では別段特定しない限り、Fc領域または定常領域中のアミノ酸残基の番号付けは、Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD 1991 に記載の、EUナンバリングシステム(EUインデックスとも呼ばれる)にしたがう。 As used herein, the term "Fc region" is used to define the C-terminal region of an immunoglobulin heavy chain that contains at least part of the constant region. The term includes the Fc region of a native sequence and the mutant Fc region. In one embodiment, the human IgG heavy chain Fc region extends from Cys226 or from Pro230 to the carboxyl end of the heavy chain. However, the C-terminal lysine (Lys447) or glycine-lysine (Gly446-Lys447) in the Fc region may or may not be present. Unless otherwise specified herein, the numbering of amino acid residues in the Fc region or constant region is Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Follow the EU numbering system (also known as the EU index) described in MD 1991.

 用語「可変領域」または「可変ドメイン」は、抗体を抗原へと結合させることに関与する、抗体の重鎖または軽鎖のドメインのことをいう。天然型抗体の重鎖および軽鎖の可変ドメイン(それぞれVHおよびVL)は、通常、各ドメインが4つの保存されたフレームワーク領域 (FR) および3つの超可変領域 (HVR) を含む、類似の構造を有する。(例えば、Kindt et al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007) 参照。)1つのVHまたはVLドメインで、抗原結合特異性を与えるに充分であろう。さらに、ある特定の抗原に結合する抗体は、当該抗原に結合する抗体からのVHまたはVLドメインを使ってそれぞれVLまたはVHドメインの相補的ライブラリをスクリーニングして、単離されてもよい。例えばPortolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991) 参照。 The term "variable region" or "variable domain" refers to the heavy or light chain domain of an antibody involved in binding the antibody to an antigen. The heavy and light chain variable domains of native antibodies (VH and VL, respectively) are similar, with each domain usually containing four conserved framework regions (FR) and three hypervariable regions (HVR). Has a structure. (See, for example, Kindt al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007).) One VH or VL domain may be sufficient to confer antigen binding specificity. In addition, antibodies that bind to a particular antigen may be isolated by screening complementary libraries of VL or VH domains using the VH or VL domains from antibodies that bind to that antigen, respectively. See, for example, Portolano et al., J. Immunol. 150: 880-887 (1993); Clarkson et al., Nature 352: 624-628 (1991).

 本明細書で用いられる用語「超可変領域」または「HVR」は、配列において超可変であり(「相補性決定領域」または「CDR」(complementarity determining region))、および/または構造的に定まったループ(「超可変ループ」)を形成し、および/または抗原接触残基(「抗原接触」)を含む、抗体の可変ドメインの各領域のことをいう。通常、抗体は6つのHVRを含む:VHに3つ(H1、H2、H3)、およびVLに3つ(L1、L2、L3)である。本明細書での例示的なHVRは、以下のものを含む:
 (a) アミノ酸残基26-32 (L1)、50-52 (L2)、91-96 (L3)、26-32 (H1)、53-55 (H2)、および96-101 (H3)のところで生じる超可変ループ (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987));
 (b) アミノ酸残基24-34 (L1)、50-56 (L2)、89-97 (L3)、31-35b (H1)、50-65 (H2)、 および95-102 (H3)のところで生じるCDR (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991));
 (c) アミノ酸残基27c-36 (L1)、46-55 (L2)、89-96 (L3)、30-35b (H1)、47-58 (H2)、および93-101 (H3) のところで生じる抗原接触 (MacCallum et al. J. Mol. Biol. 262: 732-745 (1996));ならびに、
 (d) HVRアミノ酸残基46-56 (L2)、47-56 (L2)、48-56 (L2)、49-56 (L2)、26-35 (H1)、26-35b (H1)、49-65 (H2)、93-102 (H3)、および94-102 (H3)を含む、(a)、(b)、および/または(c)の組合せ。
 別段示さない限り、HVR残基および可変ドメイン中の他の残基(例えば、FR残基)は、本明細書では上記のKabatらにしたがって番号付けされる。
As used herein, the terms "hypervariable region" or "HVR" are hypervariable in sequence ("complementarity determining regions" or "CDRs" (complementarity determining regions)) and / or are structurally defined. Refers to each region of the variable domain of an antibody that forms a loop (“hypervariable loop”) and / or contains antigen contact residues (“antigen contact”). Usually, the antibody contains 6 HVRs: 3 for VH (H1, H2, H3) and 3 for VL (L1, L2, L3). Illustrative HVRs herein include:
(a) At amino acid residues 26-32 (L1), 50-52 (L2), 91-96 (L3), 26-32 (H1), 53-55 (H2), and 96-101 (H3) The resulting hypervariable loop (Chothia and Lesk, J. Mol. Biol. 196: 901-917 (1987));
(b) At amino acid residues 24-34 (L1), 50-56 (L2), 89-97 (L3), 31-35b (H1), 50-65 (H2), and 95-102 (H3). The resulting CDR (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991));
(c) At amino acid residues 27c-36 (L1), 46-55 (L2), 89-96 (L3), 30-35b (H1), 47-58 (H2), and 93-101 (H3). Antigen contact that occurs (MacCallum et al. J. Mol. Biol. 262: 732-745 (1996));
(d) HVR Amino Acid Residues 46-56 (L2), 47-56 (L2), 48-56 (L2), 49-56 (L2), 26-35 (H1), 26-35b (H1), 49 A combination of (a), (b), and / or (c), including -65 (H2), 93-102 (H3), and 94-102 (H3).
Unless otherwise indicated, HVR residues and other residues in the variable domain (eg, FR residues) are numbered herein according to Kabat et al., Supra.

 「フレームワーク」または「FR」は、超可変領域 (HVR) 残基以外の、可変ドメイン残基のことをいう。可変ドメインのFRは、通常4つのFRドメイン:FR1、FR2、FR3、およびFR4からなる。それに応じて、HVRおよびFRの配列は、通常次の順序でVH(またはVL)に現れる:FR1-H1(L1)-FR2-H2(L2)-FR3-H3(L3)-FR4。 "Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. A variable domain FR usually consists of four FR domains: FR1, FR2, FR3, and FR4. Correspondingly, the sequences of HVR and FR usually appear in VH (or VL) in the following order: FR1-H1 (L1) -FR2-H2 (L2) -FR3-H3 (L3) -FR4.

 用語「全長抗体」、「完全抗体」、および「全部抗体」は、本明細書では相互に交換可能に用いられ、天然型抗体構造に実質的に類似した構造を有する、または本明細書で定義するFc領域を含む重鎖を有する抗体のことをいう。 The terms "full-length antibody," "complete antibody," and "whole antibody" are used interchangeably herein and have a structure substantially similar to that of a native antibody, or are defined herein. An antibody having a heavy chain containing an Fc region.

 用語「宿主細胞」、「宿主細胞株」、および「宿主細胞培養物」は、相互に交換可能に用いられ、外来核酸を導入された細胞(そのような細胞の子孫を含む)のことをいう。宿主細胞は「形質転換体」および「形質転換細胞」を含み、これには初代の形質転換細胞および継代数によらずその細胞に由来する子孫を含む。子孫は、親細胞と核酸の内容において完全に同一でなくてもよく、変異を含んでいてもよい。オリジナルの形質転換細胞がスクリーニングされたまたは選択された際に用いられたものと同じ機能または生物学的活性を有する変異体子孫も、本明細書では含まれる。 The terms "host cell", "host cell line", and "host cell culture" are used interchangeably and refer to cells into which foreign nucleic acids have been introduced, including progeny of such cells. .. Host cells include "transformants" and "transformants", which include primary transformants and progeny derived from those cells regardless of the number of passages. The offspring do not have to be exactly the same in the content of the parent cell and nucleic acid and may contain mutations. Variant progeny with the same function or biological activity as those used when the original transformed cells were screened or selected are also included herein.

 本明細書で用いられる用語「ベクター」は、それが連結されたもう1つの核酸を増やすことができる、核酸分子のことをいう。この用語は、自己複製核酸構造としてのベクター、および、それが導入された宿主細胞のゲノム中に組み入れられるベクターを含む。あるベクターは、自身が動作的に連結された核酸の、発現をもたらすことができる。そのようなベクターは、本明細書では「発現ベクター」とも称される。 As used herein, the term "vector" refers to a nucleic acid molecule that can augment another nucleic acid to which it is linked. The term includes a vector as a self-replicating nucleic acid structure and a vector incorporated into the genome of the host cell into which it has been introduced. Certain vectors can result in the expression of nucleic acids to which they are operably linked. Such vectors are also referred to herein as "expression vectors."

 「ヒト抗体」は、ヒトもしくはヒト細胞によって産生された抗体またはヒト抗体レパートリーもしくは他のヒト抗体コード配列を用いる非ヒト供給源に由来する抗体のアミノ酸配列に対応するアミノ酸配列を備える抗体である。このヒト抗体の定義は、非ヒトの抗原結合残基を含むヒト化抗体を、明確に除外するものである。 A "human antibody" is an antibody having an amino acid sequence corresponding to the amino acid sequence of an antibody produced by a human or human cell or an antibody derived from a non-human source using a human antibody repertoire or other human antibody coding sequence. This definition of human antibody explicitly excludes humanized antibodies that contain non-human antigen-binding residues.

 「ヒト化」抗体は、非ヒトHVRからのアミノ酸残基およびヒトFRからのアミノ酸残基を含む、キメラ抗体のことをいう。ある態様では、ヒト化抗体は、少なくとも1つ、典型的には2つの可変ドメインの実質的にすべてを含み、当該可変領域においては、すべてのもしくは実質的にすべてのHVR(例えばCDR)は非ヒト抗体のものに対応し、かつ、すべてのもしくは実質的にすべてのFRはヒト抗体のものに対応する。ヒト化抗体は、任意で、ヒト抗体に由来する抗体定常領域の少なくとも一部分を含んでもよい。抗体(例えば、非ヒト抗体)の「ヒト化された形態」は、ヒト化を経た抗体のことをいう。 A "humanized" antibody is a chimeric antibody that contains an amino acid residue from a non-human HVR and an amino acid residue from a human FR. In some embodiments, the humanized antibody comprises substantially all of at least one, typically two variable domains, in which all or substantially all HVRs (eg, CDRs) are non-existent. Corresponds to those of human antibodies, and all or substantially all FRs correspond to those of human antibodies. The humanized antibody may optionally include at least a portion of the antibody constant region derived from the human antibody. The "humanized form" of an antibody (eg, a non-human antibody) refers to an antibody that has undergone humanization.

 一態様において、抗原結合分子は、遺伝子組換え技術を用いて産生させた組換え型抗原結合分子である。(例えば、Borrebaeck CAK and Larrick JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990 参照)。組換え型抗原結合分子は、それをコードするDNAをハイブリドーマ、または抗原結合分子を産生する感作リンパ球等の抗原結合分子産生細胞からクローニングし、適当なベクターに組み込んで、これを宿主に導入し産生させることにより得ることができる。 In one embodiment, the antigen-binding molecule is a recombinant antigen-binding molecule produced using a gene recombination technique. (See, for example, Borrebaeck CAK and Larrick JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). The recombinant antigen-binding molecule clones the DNA encoding it from an antigen-binding molecule-producing cell such as a hybridoma or a sensitized lymphocyte that produces an antigen-binding molecule, incorporates it into an appropriate vector, and introduces it into a host. It can be obtained by producing it.

 さらに、抗原結合分子は、抗体に限定されず、アプタマーであってよい。抗原結合分子が抗体断片である場合における抗体断片としては、ダイアボディ(diabody;Db)、線状抗体、VHH(variable domain of heavy chain of heavy chain antibody)、一本鎖抗体(以下、scFvとも記載する)分子などが含まれる。ここで、「Fv」断片は最小の抗体断片であり、完全な抗原認識部位と結合部位を含む。「Fv」断片は1つの重(H)鎖可変領域(VH)および軽(L)鎖可変領域(VL)が非共有結合により強く連結されたダイマー(VH-VLダイマー)である。各可変領域の3つの相補鎖決定領域(complementarity determining region;CDR)が相互作用し、VH-VLダイマーの表面に抗原結合部位を形成する。6つのCDRが抗体に抗原結合部位を付与している。しかしながら、1つの可変領域(または、抗原に特異的な3つのCDRのみを含むFvの半分)であっても、全結合部位よりも親和性は低いが、抗原を認識し、結合する能力を有する。 Furthermore, the antigen-binding molecule is not limited to an antibody and may be an aptamer. When the antigen-binding molecule is an antibody fragment, the antibody fragment includes a diabody (Db), a linear antibody, a VHH (variable domain of heavy chain of heavy chain antibody), and a single chain antibody (hereinafter, also referred to as scFv). Includes molecules and the like. Here, the "Fv" fragment is the smallest antibody fragment and contains the complete antigen recognition and binding sites. The "Fv" fragment is a dimer (V H- V L dimer) in which one heavy (H) chain variable region (V H ) and a light (L) chain variable region (V L ) are strongly linked by a non-covalent bond. .. The three complementarity determining regions (CDRs) of each variable region interact to form an antigen-binding site on the surface of the V H - VL dimer. Six CDRs confer an antigen binding site on the antibody. However, even one variable region (or half of the Fv containing only three antigen-specific CDRs) has the ability to recognize and bind antigens, albeit with lower affinity than the full binding site. ..

 また、Fab断片(F(ab)とも呼ばれる)はさらに、L鎖の定常領域およびH鎖の定常領域(CH1)を含む。Fab'断片は、抗体のヒンジ領域からの1またはそれ以上のシステインを含むH鎖CH1領域のカルボキシ末端由来の数残基を付加的に有する点でFab断片と異なっている。Fab'-SHとは、定常領域の1またはそれ以上のシステイン残基が遊離のチオール基を有するFab'を示すものである。F(ab')断片は、F(ab')2ペプシン消化物のヒンジ部のシステインにおけるジスルフィド結合の切断により製造される。化学的に結合されたその他の抗体断片も当業者には知られている。 The Fab fragment (also referred to as F (ab)) further contains a constant region of the L chain and a constant region of the H chain (CH1). The Fab'fragment differs from the Fab fragment in that it additionally has a few residues from the carboxy terminus of the H chain CH1 region containing one or more cysteines from the hinge region of the antibody. Fab'-SH refers to Fab'in which one or more cysteine residues in the constant region have a free thiol group. The F (ab') fragment is produced by cleavage of the disulfide bond in cysteine at the hinge of the F (ab') 2 pepsin digest. Other chemically bound antibody fragments are also known to those of skill in the art.

 ダイアボディは、遺伝子融合により構築された二価(bivalent)の抗体断片を指す(Holliger P et al., Proc.Natl.Acad.Sci.USA 90: 6444-6448 (1993)、EP404,097号、WO93/11161号等)。ダイアボディは、2本のポリペプチド鎖から構成されるダイマーであり、ポリペプチド鎖は各々、同じ鎖中でL鎖可変領域(VL)及びH鎖可変領域(VH)が、互いに結合できない位に短い、例えば、5残基程度のリンカーにより結合されている。同一ポリペプチド鎖上にコードされるVLとVHとは、その間のリンカーが短いため単鎖可変領域フラグメントを形成することが出来ず二量体を形成するため、ダイアボディは2つの抗原結合部位を有することとなる。 Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger P et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP404,097, WO 93/11161, etc.). A diabody is a dimer composed of two polypeptide chains, in which the L chain variable region ( VL ) and the H chain variable region (V H ) cannot bind to each other in the same chain, respectively. It is bound by a linker that is short in position, for example, about 5 residues. Since V L and V H encoded on the same polypeptide chain cannot form a single chain variable region fragment due to the short linker between them and form a dimer, the diabody binds two antigens. Will have a site.

 一本鎖抗体またはscFv抗体断片には、抗体のVHおよびVL領域が含まれ、これらの領域は単一のポリペプチド鎖中に存在する。一般に、FvポリペプチドはさらにVHおよびVL領域の間にポリペプチドリンカーを含んでおり、これによりscFvは、抗原結合のために必要な構造を形成することができる(scFvの総説については、Pluckthun『The Pharmacology of Monoclonal Antibodies』Vol.113(Rosenburg and Moore ed (Springer Verlag, New York) pp.269-315, 1994)を参照)。本発明におけるリンカーは、その両端に連結された抗体可変領域の発現を阻害するものでなければ特に制限されない。 A single chain antibody or scFv antibody fragment contains the V H and VL regions of the antibody, which are located within a single polypeptide chain. In general, Fv polypeptides also contain a polypeptide linker between the V H and VL regions, which allows scFv to form the structure required for antigen binding (for a review of scFv, Pluckthun. See The Pharmacology of Monoclonal Antibodies, Vol.113 (Rosenburg and Moore ed (Springer Verlag, New York) pp.269-315, 1994). The linker in the present invention is not particularly limited as long as it does not inhibit the expression of the antibody variable region linked to both ends thereof.

 IgGタイプ二重特異性抗体はIgG抗体を産生するハイブリドーマ二種を融合することによって生じるhybrid hybridoma(quadroma)によって分泌させることが出来る(Milstein C et al. Nature 1983, 305: 537-540)。また目的の二種のIgGを構成するL鎖及びH鎖の遺伝子、合計4種の遺伝子を細胞に導入することによって共発現させることによって分泌させることが出来る。 IgG-type bispecific antibodies can be secreted by hybridomas (quadromas) produced by fusing two hybridomas that produce IgG antibodies (Milstein C et al. Nature 1983, 305: 537-540). In addition, it can be secreted by co-expressing a total of four genes, L chain and H chain genes constituting the two target IgGs, into cells.

 しかし、これらの方法で産生されるIgGのH鎖とL鎖の組合せは理論上10通りにもなる。10種類のIgGから目的の組み合わせのH鎖L鎖からなるIgGを精製することは困難である。さらに目的の組み合わせのものの分泌量も理論上著しく低下するため、大きな培養規模が必要になり、製造上のコストはさらに増大する。 However, there are theoretically 10 combinations of H chain and L chain of IgG produced by these methods. It is difficult to purify an IgG consisting of H chains and L chains of the desired combination from 10 types of IgG. In addition, the amount of secretion of the desired combination is theoretically significantly reduced, which requires a large culture scale and further increases the manufacturing cost.

 この際H鎖のCH3領域に適当なアミノ酸置換を施すことによってH鎖についてヘテロな組合せのIgGを優先的に分泌させることも出来る(Ridgway JB et al. Protein Engineering 1996, 9: 617-621、Merchant AM et al. Nature Biotechnology 1998, 16: 677-681)。 At this time, by appropriately substituting an appropriate amino acid in the CH3 region of the H chain, IgG in a heterogeneous combination for the H chain can be preferentially secreted (Ridgway JB et al. Protein Engineering 1996, 9: 617-621, Merchant). AM et al. Nature Biotechnology 1998, 16: 677-681).

 また、L鎖に関しては、H鎖可変領域に比べてL鎖可変領域の多様性が低いことから、両H鎖に結合能を与え得る共通のL鎖が得られることが期待される。この共通L鎖と両H鎖遺伝子を細胞に導入することによってIgGを発現させることで効率の良い二重特異性IgGの発現が可能となる(Nature Biotechnology. 1998, 16, 677-681)。しかし任意に2種の抗体を選んだ場合、同じL鎖を含む可能性は低く上記のアイデアを実行することは困難であり、任意の異なるH鎖に対応し高い結合能を示す共通L鎖を選択する方法も提案されている(WO2004/065611)。上記変異(Nature Biotechnology. 1998, 16, 677-681) CH3を有するH鎖は、相手側のH鎖が存在しない場合ほとんど分泌されない。この特徴を利用してまず右腕のL鎖とH鎖を誘導発現させ、これを止めた後、左腕のL鎖とH鎖を誘導発現させることによって目的の組み合わせのIgGの発現比率を高めることが出来る(PCT/JP2004/008585)。
 Fab’を化学的に架橋することによっても二重特異性抗原結合分子を作製し得る。例えば一方の抗体から調製したFab’をo-PDM(ortho-phenylenedi-maleimide)にてマレイミド化し、これともう一方の抗体から調製したFab’を反応させることにより、異なる抗体由来Fab’同士を架橋させ二重特異性 F(ab’)2を作製することが出来る(Keler T et al. Cancer Research 1997, 57: 4008-4014)。またFab’-チオニトロ安息香酸(TNB)誘導体とFab’-チオール(SH)等の抗体断片を化学的に結合する方法も知られている(Brennan M et al. Science 1985, 229: 81-83)。
Further, regarding the L chain, since the diversity of the L chain variable region is lower than that of the H chain variable region, it is expected that a common L chain capable of imparting a binding ability to both H chains can be obtained. Efficient bispecific IgG can be expressed by expressing IgG by introducing this common L-chain and both H-chain genes into cells (Nature Biotechnology. 1998, 16, 677-681). However, if two types of antibodies are arbitrarily selected, it is unlikely that they will contain the same L chain, and it is difficult to carry out the above idea. A method of selection has also been proposed (WO2004 / 065611). The H chain having the above mutation (Nature Biotechnology. 1998, 16, 677-681) CH3 is hardly secreted in the absence of the other H chain. Utilizing this feature, it is possible to increase the expression ratio of IgG in the desired combination by first inducing and expressing the L and H chains of the right arm, stopping this, and then inducing and expressing the L and H chains of the left arm. Yes (PCT / JP2004 / 008585).
Bispecific antigen-binding molecules can also be made by chemically cross-linking Fab'. For example, Fab'prepared from one antibody is maleimided with o-PDM (ortho-phenylenedi-maleimide), and Fab'prepared from the other antibody is reacted with this to crosslink Fab'derived from different antibodies. Bispecific F (ab') 2 can be produced (Keler T et al. Cancer Research 1997, 57: 4008-4014). Also known is a method of chemically binding a Fab'-thionitrobenzoic acid (TNB) derivative to an antibody fragment such as Fab'-thiol (SH) (Brennan M et al. Science 1985, 229: 81-83). ..

 化学架橋の代りにFos, Junなどに由来するロイシンジッパーを用いることも出来る。Fos, Junはホモダイマーも形成するが、ヘテロダイマーを優先的に形成することを利用する。Fosロイシンジッパーを付加したFab’とJunのそれを付加したもう一方のFab’を発現調製する。温和な条件で還元した単量体Fab’-Fos, Fab’-Junを混合し反応させることによって二重特異性 F(ab’)2が形成できる(Kostelny SA et al. J of Immunology, 1992, 148: 1547-53)。この方法はFab’には限定されず、scFv, Fvなどにおいても応用可能である。 Leucine zippers derived from Fos, Jun, etc. can be used instead of chemical cross-linking. Fos and Jun also form homodimers, but take advantage of the preferential formation of heterodimers. The expression of Fab'with the Fos leucine zipper and the other Fab'with the addition of that of Jun are prepared. Bispecific F (ab') 2 can be formed by mixing and reacting the reduced monomers Fab'-Fos and Fab'-Jun under mild conditions (Kostelny SA et al. J of Immunology, 1992, 148: 1547-53). This method is not limited to Fab', but can also be applied to scFv, Fv, etc.

 ダイアボディにおいても二重特異性抗原結合分子を作製し得る。二重特異性ダイアボディは二つのcross-over scFv断片のヘテロダイマーである。つまり二種の抗体A,B由来のVHとVLを5残基前後の比較的短いリンカーで結ぶことによって作製されたVH(A)-VL(B), VH(B)-VL(A)を用いてヘテロダイマーを構成することによって出来る(Holliger P et al. Proc of the National Academy of Sciences of the USA 1993, 90: 6444-6448)。 A bispecific antigen-binding molecule can also be prepared in the diabody. The bispecific diabody is a heterodimer of two cross-over scFv fragments. In other words, V H (A) -V L (B), V H (B) -produced by linking V H and V L derived from two types of antibodies A and B with a relatively short linker of about 5 residues. It can be done by constructing a heterodimer using V L (A) (Holliger P et al. Proc of the National Academy of Sciences of the USA 1993, 90: 6444-6448).

 この際、二種のscFvを15残基程度の柔軟な比較的長いリンカーで結ぶ(一本鎖ダイアボディ:Kipriyanov SM et al. J of Molecular Biology. 1999, 293: 41-56)、適当なアミノ酸置換(knobs-into-holes: Zhu Z et al. Protein Science. 1997, 6: 781-788)を行うことによって目的の構成を促進させることも出来る。 At this time, two kinds of scFv are connected by a flexible and relatively long linker of about 15 residues (single-chain diabody: Kipriyanov SM et al. J of Molecular Biology. 1999, 293: 41-56), and an appropriate amino acid. Substitution (knobs-into-holes: Zhu Z et al. Protein Science. 1997, 6: 781-788) can also be used to promote the desired configuration.

 二種のscFvを15残基程度の柔軟な比較的長いリンカーで結ぶことによって作製できるsc(Fv)2も二重特異性抗原結合分子となり得る(Mallender WD et al. J of Biological Chemistry, 1994, 269: 199-206)。 Sc (Fv) 2, which can be produced by linking two types of scFv with a flexible and relatively long linker of about 15 residues, can also be a bispecific antigen-binding molecule (Mallender WD et al. J of Biological Chemistry, 1994, 269: 199-206).

 抗体修飾物としては、例えば、ポリエチレングリコール(PEG)等の各種分子と結合した抗体を挙げることができる。本発明の抗体修飾物においては、結合される物質は限定されない。このような抗体修飾物を得るには、得られた抗体に化学的な修飾を施すことによって得ることができる。これらの方法はこの分野において既に確立されている。 Examples of antibody modifications include antibodies bound to various molecules such as polyethylene glycol (PEG). In the antibody modified product of the present invention, the substance to be bound is not limited. Such antibody modifications can be obtained by chemically modifying the obtained antibody. These methods have already been established in this field.

 本発明の抗原結合分子は、ヒト抗体、マウス抗体、ラット抗体など、その由来は限定されない。またキメラ抗体やヒト化抗体などの遺伝子改変抗体でもよい。 The origin of the antigen-binding molecule of the present invention is not limited, such as human antibody, mouse antibody, and rat antibody. Further, a gene-modified antibody such as a chimeric antibody or a humanized antibody may be used.

 ヒト抗体の取得方法は既に知られており、例えば、ヒト抗体遺伝子の全てのレパートリーを有するトランスジェニック動物を目的の抗原で免疫することで目的のヒト抗体を取得することができる(国際特許出願公開番号WO 93/12227, WO 92/03918,WO 94/02602, WO 94/25585,WO 96/34096, WO 96/33735参照)。 A method for obtaining a human antibody is already known. For example, a human antibody of interest can be obtained by immunizing a transgenic animal having the entire repertoire of human antibody genes with the antigen of interest (publication of international patent application). See numbers WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735).

 遺伝子改変抗体は、既知の方法を用いて製造することができる。具体的には、たとえばキメラ抗体は、免疫動物の抗体のH鎖、およびL鎖の可変領域と、ヒト抗体のH鎖およびL鎖の定常領域からなる抗体である。免疫動物由来の抗体の可変領域をコードするDNAを、ヒト抗体の定常領域をコードするDNAと連結し、これを発現ベクターに組み込んで宿主に導入し産生させることによって、キメラ抗体を得ることができる。 The genetically modified antibody can be produced using a known method. Specifically, for example, a chimeric antibody is an antibody consisting of a variable region of H chain and L chain of an immune animal antibody and a constant region of H chain and L chain of a human antibody. A chimeric antibody can be obtained by ligating a DNA encoding a variable region of an antibody derived from an immune animal with a DNA encoding a constant region of a human antibody, incorporating this into an expression vector, introducing it into a host, and producing it. ..

 ヒト化抗体は、再構成(reshaped)ヒト抗体とも称される改変抗体である。ヒト化抗体は、免疫動物由来の抗体のCDRを、ヒト抗体の相補性決定領域へ移植することによって構築される。その一般的な遺伝子組換え手法も知られている。 A humanized antibody is a modified antibody that is also called a reshaped human antibody. Humanized antibodies are constructed by transplanting the CDRs of antibodies from immune animals into the complementarity determining regions of human antibodies. The general gene recombination method is also known.

 具体的には、マウス抗体のCDRとヒト抗体のフレームワーク領域(framework region;FR)を連結するように設計したDNA配列を、末端部にオーバーラップする部分を有するように作製した数個のオリゴヌクレオチドからPCR法により合成する。得られたDNAを、ヒト抗体定常領域をコードするDNAと連結し、次いで発現ベクターに組み込んで、これを宿主に導入し産生させることにより得られる(欧州特許出願公開番号EP 239400、国際特許出願公開番号WO 96/02576参照)。CDRを介して連結されるヒト抗体のFRは、相補性決定領域が良好な抗原結合部位を形成するものが選択される。必要に応じ、再構成ヒト抗体の相補性決定領域が適切な抗原結合部位を形成するように抗体の可変領域におけるフレームワーク領域のアミノ酸を置換してもよい(Sato K et al, Cancer Research 1993, 53: 851-856)。また、様々なヒト抗体由来のフレームワーク領域に置換してもよい(国際特許出願公開番号WO 99/51743参照)。 Specifically, several oligos prepared by preparing a DNA sequence designed to link the CDR of a mouse antibody and the framework region (FR) of a human antibody so as to have an overlapping portion at the terminal portion. It is synthesized from nucleotides by the PCR method. It is obtained by ligating the obtained DNA with the DNA encoding the human antibody constant region, then incorporating it into an expression vector, introducing it into a host and producing it (European Patent Application Publication No. EP 239400, International Patent Application Publication). See number WO 96/02576). The FR of the human antibody linked via CDR is selected so that the complementarity determining regions form a good antigen-binding site. If desired, the amino acids in the framework regions of the variable region of the antibody may be replaced so that the complementarity determining regions of the reconstituted human antibody form the appropriate antigen binding site (Sato K et al, Cancer Research 1993, 53: 851-856). It may also be replaced with framework regions derived from various human antibodies (see International Patent Application Publication No. WO 99/51743).

 一態様としては、酵素および該酵素の触媒反応を受け得る基質の両方を認識して該酵素が該基質を加水分解するが、血液凝固第V因子、血液凝固第VIII因子、組織因子(TF), トロンボモデュリン(TM)、プロテインS(PS)、プロテインZ(PZ)補体C4b、補体制御タンパクH因子(complement regulatory Factor H)、membrane cofactor Protein(MCP)、complement receptor1(CR1)からなる群から選択される少なくとも一の補因子あるいはヘパリンの機能を代替しない二重特異性抗原結合分子が提供される。別の一態様においては、酵素および該酵素の触媒反応を受け得る基質の両方を認識するが、天然に存在する血液凝固系補因子の機能を代替しない二重特異性抗原結合分子が提供される。これらの態様における、酵素は、生体で起こる化学反応に対する触媒活性を有する分子である限り、特に制限されない。酵素は、プロテアーゼ、アミラーゼ、セルラーゼ、リパーゼが例示される。プロテアーゼには、血液凝固系のセリンプロテアーゼが例示される。特定の態様において、酵素は血液凝固系酵素であり得る。血液凝固系酵素であるセリンプロテアーゼとして、例えばF.XII(a)、F.XI(a)、F.IX(a)、F.X(a)、F.VII(a)、F.IIaが挙げられる。なお、F.XII(a)はF.XII及び/またはF.XIIaを意味し、他も同様である。 In one embodiment, the enzyme hydrolyzes the substrate by recognizing both the enzyme and the substrate capable of undergoing a catalytic reaction of the enzyme, but blood coagulation factor V, blood coagulation factor VIII, tissue factor (TF). , From Thrombomodulin (TM), Protein S (PS), Protein Z (PZ) Complement C4b, Complement Regulatory Factor H, membrane cofactor Protein (MCP), complement receptor1 (CR1) A bispecific antigen-binding molecule that does not replace the function of at least one complement or heparin selected from the group is provided. In another aspect, a bispecific antigen-binding molecule that recognizes both the enzyme and the substrate that can be catalyzed by the enzyme but does not replace the function of naturally occurring coagulation cofactors is provided. .. The enzyme in these embodiments is not particularly limited as long as it is a molecule having catalytic activity for a chemical reaction occurring in a living body. Examples of the enzyme include protease, amylase, cellulase, and lipase. Examples of proteases include serine proteases of the blood coagulation system. In certain embodiments, the enzyme can be a blood coagulation enzyme. Examples of serine proteases that are blood coagulation enzymes include F.XII (a), F.XI (a), F.IX (a), FX (a), F.VII (a), and F.IIa. .. In addition, F.XII (a) means F.XII and / or F.XIIa, and the same applies to others.

 本願でいう前記酵素の触媒反応を受け得る基質とは、強制的に近接させた場合に酵素の触媒反応が起こる基質を言う。酵素の触媒反応が起こることは、基質を加水分解させることを意味する。基質の加水分解は、本願の二重特異性抗原結合分子がない場合と比較して、基質が加水分解されていればよい。基質の加水分解反応は、例えばProtein Assey Kit II(Bio-Red)を用いて、Bradford法等によって二重特異性抗原結合分子添加後の継時的なタンパク質量の測定で比較できる。あるいは、二重特異性抗原結合分子の添加前後のタンパク質を用いたSDS-PAGE分析やウエスタン・ブロッティング(WB)によって測定できる。あるいは二重特異性抗原結合分子の添加前後のタンパク質の質量分析(MALDI-TOFMS等)を行うことやN末端アミノ酸配列分析(プロテインシーケンス)を行うことで測定できる。基質の加水分解は例えば、血液凝固第X因子または血液凝固第IX因子の活性化が挙げられる。基質の活性化は該酵素・該基質を含む反応系を用い、該抗原結合分子を加えることによる該酵素活性(基質分解能)の上昇を指標とすることができ、例えば 該酵素、該基質、活性化された基質の合成基質、リン脂質、Ca2+から成る測定系(in vitro 酵素反応測定系)で、該酵素による該基質活性化促進活性で評価できる。その結果を以って、該活性を有する二重特異性抗原結合分子として、原則本測定系で該酵素添加群のみ0.1以上の該酵素による該基質活性化促進活性を示したものを選択できる。なお、ここでいう該酵素による該基質活性化促進活性とは、抗原結合分子溶液の合成基質添加30分後の吸光度の値で測定することができる。 The substrate capable of undergoing the catalytic reaction of the enzyme referred to in the present application means a substrate in which the catalytic reaction of the enzyme occurs when the enzyme is forcibly brought close to each other. The catalytic reaction of the enzyme means that the substrate is hydrolyzed. Hydrolysis of the substrate may be performed as long as the substrate is hydrolyzed as compared with the case where the bispecific antigen-binding molecule of the present application is absent. Substrate hydrolysis reactions can be compared by measuring the amount of protein over time after the addition of a bispecific antigen-binding molecule by the Bradford method or the like using, for example, Protein Assey Kit II (Bio-Red). Alternatively, it can be measured by SDS-PAGE analysis or Western blotting (WB) using a protein before and after the addition of the bispecific antigen-binding molecule. Alternatively, it can be measured by performing mass spectrometry (MALDI-TOFMS, etc.) of the protein before and after the addition of the bispecific antigen-binding molecule, or by performing N-terminal amino acid sequence analysis (protein sequence). Substrate hydrolysis includes, for example, activation of blood coagulation factor X or blood coagulation factor IX. The activation of the substrate can be performed by using the enzyme and the reaction system containing the substrate, and the increase in the enzyme activity (substrate resolution) by adding the antigen-binding molecule can be used as an index. For example, the enzyme, the substrate, and the activity. It is a measurement system (in vitro enzyme reaction measurement system) consisting of a synthetic substrate, phospholipids, and Ca2 + of the converted substrate, and can be evaluated by the substrate activation promoting activity by the enzyme. Based on the results, as a bispecific antigen-binding molecule having the activity, in principle, a molecule showing the substrate activation promoting activity by the enzyme of 0.1 or more only in the enzyme-added group can be selected in this measurement system. The substrate activation promoting activity by the enzyme referred to here can be measured by the value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.

 「該酵素による該基質の活性化を促進する」とはin vitro 酵素反応測定系における該基質活性化促進活性が発色基質溶液添加30分後の吸光度0.1以上を示すことを指す。in vitro 酵素反応測定系は、各酵素、各基質で測定可能であり、例えば実施例2に示すようにF.XIaによるF.X活性化促進活性として、発色基質溶液添加30分間後の吸光度の値として0.1以上の場合には該酵素による該基質の活性化を促進するとして評価できる。 "Promoting the activation of the substrate by the enzyme" means that the substrate activation promoting activity in the in vitro enzyme reaction measurement system shows an absorbance of 0.1 or more 30 minutes after the addition of the chromogenic substrate solution. The in vitro enzyme reaction measurement system can be measured with each enzyme and each substrate. For example, as shown in Example 2, as the FX activation promoting activity by F.XIa, as the absorbance value 30 minutes after the addition of the color-developing substrate solution. When it is 0.1 or more, it can be evaluated as promoting the activation of the substrate by the enzyme.

 酵素および基質が血液凝固線溶関連因子である態様における酵素(「血液凝固酵素」とも呼ぶ)、および基質(「血液凝固基質」とも呼ぶ)の具体例としては、例えば、以下の表1の組合せを挙げることができる。 Specific examples of the enzyme (also referred to as "blood coagulation enzyme") and the substrate (also referred to as "blood coagulation substrate") in the embodiment in which the enzyme and the substrate are blood coagulation / fibrinolysis-related factors are, for example, the combinations shown in Table 1 below. Can be mentioned.

Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001

 二重特異性抗原結合分子を得る方法は特に制限されず、どのような方法で取得されてもよい。例えば、酵素A及び基質Bに対する二重特異性抗原結合分子を得る場合、酵素A、基質Bそれぞれを免疫動物に免疫し、抗酵素A抗体及び抗基質B抗体を取得する。その後、抗酵素A抗体のH鎖可変領域とL鎖可変領域及び抗基質B抗体のH鎖可変領域とL鎖可変領域を含む二重特異性抗原結合分子を作製する。ここで、抗酵素A抗体と抗基質B抗体はそれぞれ複数種得られていることが望ましく、これらを用いてなるべく多くの組合せの二重特異性抗原結合分子を作製することが好ましい。二重特異性抗原結合分子を作製後、基質Bを活性化させる抗原結合分子を選択する。 The method for obtaining the bispecific antigen-binding molecule is not particularly limited, and any method may be used for obtaining the bispecific antigen-binding molecule. For example, when a bispecific antigen-binding molecule for enzyme A and substrate B is obtained, each of enzyme A and substrate B is immunized against an immunized animal to obtain an anti-enzyme A antibody and an anti-substrate B antibody. Then, a bispecific antigen-binding molecule containing the H-chain variable region and the L-chain variable region of the anti-enzyme A antibody and the H-chain variable region and the L-chain variable region of the anti-substrate B antibody is prepared. Here, it is desirable that a plurality of types of anti-enzyme A antibody and anti-substrate B antibody are obtained, and it is preferable to prepare as many combinations of bispecific antigen-binding molecules as possible using these. After preparing the bispecific antigen-binding molecule, the antigen-binding molecule that activates substrate B is selected.

 酵素あるいは基質に対する抗原結合分子は、当業者に公知の方法により得ることができる。例えば、免疫動物に対して抗原を免疫することにより調製することができる。動物を免疫する抗原としては、免疫原性を有する完全抗原と、免疫原性を有さない不完全抗原(ハプテンを含む)が挙げられる。免疫に際しては、抗原結合分子が結合する酵素あるいは基質、もしくはそれらを発現する核酸を、上記抗原(免疫原)として使用する。免疫する動物として、例えば、マウス、ラット、ハムスター、モルモット、ウサギ、ニワトリまたはアカゲザル等を用いることができる。これら動物に対して、抗原を免疫することは、当業者においては、周知の方法によって行うことができる。好ましくは、免疫された動物または該動物の細胞から抗体のL鎖およびH鎖の可変領域の回収を行う。この操作は、当業者においては一般的に公知の技術を用いて行うことができる。抗原によって免疫された動物は、とりわけ脾臓細胞において該抗原に対する抗体を発現する。従って、例えば、免疫された動物の脾臓細胞からmRNAを調製し、該動物の可変領域に対応するプライマーを用いて、RT-PCRによりL鎖およびH鎖の可変領域の回収を行うことができる。 The antigen-binding molecule to the enzyme or substrate can be obtained by a method known to those skilled in the art. For example, it can be prepared by immunizing an immune animal with an antigen. Examples of the antigen that immunizes an animal include a complete antigen having immunogenicity and an incomplete antigen (including a hapten) having no immunogenicity. In immunization, an enzyme or substrate to which an antigen-binding molecule binds, or a nucleic acid expressing them is used as the above-mentioned antigen (immunogen). As the immunized animal, for example, mice, rats, hamsters, guinea pigs, rabbits, chickens, rhesus monkeys and the like can be used. Immunization of these animals with an antigen can be performed by a method well known to those skilled in the art. Preferably, the variable regions of the L and H chains of the antibody are recovered from the immunized animal or the cells of the animal. This operation can be performed using a technique generally known to those skilled in the art. Animals immunized with an antigen express antibodies against that antigen, especially in spleen cells. Therefore, for example, mRNA can be prepared from the spleen cells of an immunized animal, and the variable regions of L and H chains can be recovered by RT-PCR using the primers corresponding to the variable regions of the animal.

 詳細には、動物に酵素、基質それぞれを免疫する。免疫原とする酵素、基質は、蛋白質全体、もしくは該蛋白質の部分ペプチドであってもよい。また、動物を免疫するのに用いる免疫原としては、場合により抗原となるものを他の分子に結合させ可溶性抗原とすることも可能であり、また、場合によりそれらの断片を用いてもよい。 Specifically, immunize animals with enzymes and substrates. The enzyme or substrate used as an immunogen may be the whole protein or a partial peptide of the protein. Further, as the immunogen used for immunizing an animal, it is possible to bind an antigen to another molecule to obtain a soluble antigen, and in some cases, fragments thereof may be used.

 免疫されたマウスの脾臓から脾細胞を単離し、マウスミエローマ細胞と融合し、ハイブリドーマを作製する。抗原に結合するハイブリドーマをそれぞれ選択し、可変領域に対応するプライマーなどを用いてRT-PCRにてL鎖、H鎖の可変領域を回収することが出来る。CDRに対応するプライマー、CDRよりも多様性の低いフレームワークに対応するプライマー、あるいはシグナル配列とCH1もしくはL鎖定常領域(CL)に対応するプライマーを用いることもできる。
 あるいは、当業者公知の方法であるB細胞クローニング技術(Proc Natl Acad Sci U S A. 1996;93(15):7843-7848.;WO2008/045140;およびWO2009/113742)によって生成されうるが、これらに限定されない。
Spleen cells are isolated from the spleen of immunized mice and fused with mouse myeloma cells to produce hybridomas. Hybridomas that bind to the antigen can be selected, and the variable regions of the L and H chains can be recovered by RT-PCR using primers and the like corresponding to the variable regions. Primers that correspond to CDRs, frameworks that are less diverse than CDRs, or primers that correspond to the signal sequence and the CH1 or L chain constant region ( CL ) can also be used.
Alternatively, they can be produced by B cell cloning techniques known to those of skill in the art (Proc Natl Acad Sci US A. 1996; 93 (15): 7843-7848 .; WO2008 / 045140; and WO2009 / 113742). Not limited.

 あるいは、免疫された動物の脾細胞からmRNAを抽出し、可変領域付近に対応するプライマーを用いてRT-PCRにてL鎖、H鎖可変領域のcDNAを回収する。また、in vitroにおいてリンパ球を免疫することもできる。これを用いてscFvもしくはFabを提示するライブラリーを構築する。パンニングによって抗原結合抗体クローンを濃縮・クローン化し、可変領域を得ることが出来る。この際、ヒトや免疫していない動物の末梢血単核球、脾臓、扁桃腺などに由来するmRNAを材料とする同様のライブラリーを用いてスクリーニングを行うことも可能である。 Alternatively, mRNA is extracted from the splenocytes of an immunized animal, and cDNA in the L-chain and H-chain variable regions is recovered by RT-PCR using a primer corresponding to the vicinity of the variable region. It can also immunize lymphocytes in vitro. Use this to build a library that presents scFv or Fab. Antigen-binding antibody clones can be concentrated and cloned by panning to obtain variable regions. At this time, it is also possible to perform screening using a similar library using mRNA derived from peripheral blood mononuclear cells, spleen, tonsils, etc. of humans and non-immune animals.

 その可変領域を用いて抗原結合分子発現ベクターを作製する。抗酵素抗原結合分子発現ベクターと抗基質抗原結合分子発現ベクターを同一の細胞に導入し、抗原結合分子を発現させることにより二重特異性抗原結合分子を得ることができる。 An antigen-binding molecule expression vector is prepared using the variable region. A bispecific antigen-binding molecule can be obtained by introducing an anti-enzyme antigen-binding molecule expression vector and an anti-substrate antigen-binding molecule expression vector into the same cell and expressing the antigen-binding molecule.

 本発明における抗原結合分子の選択は、例えば、以下のような方法により行うことができる。
(1)該酵素・該基質を含む反応系を用い、該抗原結合分子を加えることによる該酵素活性(基質分解能)の上昇を指標とし、選択する。
(2)該酵素・該基質・該補因子が関わる生体機能を測定するあるいは模倣する系(例えば、血漿凝固測定系)を用い、該酵素・該基質・該補因子のうち少なくとも1つの非存在条件下にて該抗原結合分子を加えることによる機能回復活性を指標とし、選択する。
The antigen-binding molecule in the present invention can be selected by, for example, the following method.
(1) A reaction system containing the enzyme / substrate is used, and an increase in the enzyme activity (substrate resolution) due to the addition of the antigen-binding molecule is used as an index for selection.
(2) Using a system that measures or mimics the biological functions associated with the enzyme, the substrate, and the cofactor (for example, a plasma coagulation measurement system), at least one of the enzyme, the substrate, and the cofactor is absent. The function recovery activity by adding the antigen-binding molecule under the conditions is used as an index for selection.

 得られた抗原結合分子は、均一にまで精製することができる。抗原結合分子の分離、精製は通常の蛋白質で使用されている分離、精製方法を使用すればよい。例えばアフィニティークロマトグラフィー等のクロマトグラフィーカラム、フィルター、限外濾過、塩析、透析、SDSポリアクリルアミドゲル電気泳動、等電点電気泳動等を適宜選択、組合せれば、抗体を分離、精製することができる(Antibodies : A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988) が、これらに限定されるものではない。アフィニティークロマトグラフィーに用いるカラムとしては、プロテインAカラム、プロテインGカラムなどが挙げられる。 The obtained antigen-binding molecule can be purified to a uniform level. For the separation and purification of the antigen-binding molecule, the separation and purification methods used for ordinary proteins may be used. For example, an antibody can be separated and purified by appropriately selecting and combining a chromatography column such as affinity chromatography, a filter, ultrafiltration, salting out, dialysis, SDS polyacrylamide gel electrophoresis, isoelectric focusing, and the like. It can be done (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988), but it is not limited to these. Examples of the column used for affinity chromatography include a protein A column and a protein G column.

 本発明の二重特異性抗原結合分子は、例えば酵素および基質の組合せが、血液凝固線溶関連因子のF.XIa、及びF.Xである場合には、好ましくは、抗F.XIa抗体における可変領域と、抗F.X抗体における可変領域とを含む構造を有する。 The bispecific antigen-binding molecule of the present invention preferably has a variable region in an anti-F.XIa antibody when, for example, the combination of enzyme and substrate is the blood coagulation / fibrinolysis-related factors F.XIa and FX. And a variable region in the anti-FX antibody.

 二重特異性抗原結合分子は以下の方法で作製できる。例えば酵素および基質の組合せがF.XIa及びF.Xの場合、市販のF.XIa及びF.Xをそれぞれマウスの皮下に免疫した。抗体価の上昇した免疫マウスの脾臓から脾細胞を単離し、マウスミエローマ細胞と融合し、ハイブリドーマを作製した。抗原(F.XIa、F.X)に結合するハイブリドーマをそれぞれ選択し、可変領域に対応するプライマーを用いてRT-PCRにてL鎖、H鎖の可変領域を回収した。L鎖可変領域はCLを含むL鎖発現ベクターに、H鎖可変領域はH鎖定常領域を含むH鎖発現ベクターにそれぞれ組み込んだ。また、この免疫マウスの脾臓からmRNAを抽出し、可変領域に対応するプライマーを用いてRT-PCRにてL鎖、H鎖可変領域のcDNAを回収した。これら可変領域を用いてscFvを提示するファージライブラリーを構築した。パンニングによって抗原結合抗体クローンを濃縮・クローン化し、その可変領域を用いて抗原結合分子発現ベクターを作製した。あるいは当業者公知の方法でB細胞クローニング技術によって生成されうる。抗F.XIa抗体(H鎖、L鎖)の発現ベクターとF.X抗体(H鎖、L鎖)の発現ベクターを同一の細胞に導入し、抗原結合分子を発現させることにより二重特異性抗原結合分子を得た。 The bispecific antigen-binding molecule can be prepared by the following method. For example, when the combination of enzyme and substrate was F.XIa and FX, commercially available F.XIa and FX were subcutaneously immunized in mice, respectively. Spleen cells were isolated from the spleen of immune mice with elevated antibody titers and fused with mouse myeloma cells to prepare hybridomas. Hybridomas that bind to antigens (F.XIa, FX) were selected, and the variable regions of L and H chains were recovered by RT-PCR using primers corresponding to the variable regions. L chain variable region to the L chain expression vector containing a C L, H chain variable region incorporating the respective H chain expression vector containing an H chain constant region. In addition, mRNA was extracted from the spleen of this immune mouse, and cDNA in the L-chain and H-chain variable regions was recovered by RT-PCR using primers corresponding to the variable regions. A phage library that presents scFv was constructed using these variable regions. An antigen-binding antibody clone was concentrated and cloned by panning, and an antigen-binding molecule expression vector was prepared using the variable region. Alternatively, it can be produced by a B cell cloning technique by a method known to those skilled in the art. Bispecific antigen binding by introducing an anti-F.XIa antibody (H chain, L chain) expression vector and an FX antibody (H chain, L chain) expression vector into the same cell and expressing an antigen-binding molecule. Obtained a molecule.

 得られた二重特異性抗原結合分子に関しては、F.XIa、F.X、F.Xaの合成基質(S-2222)、リン脂質から成る測定系で、F.XIaによるF.X活性化促進活性を評価した。その結果を以って、有する二重特異性抗原結合分子として、原則本測定系で0.1以上のF.XIaによるF.X活性化促進活性を示したものを選択した。なお、ここでいうF.XIaによるF.X活性化促進活性とは、抗原結合分子溶液の合成基質添加30分後の吸光度の値である。 Regarding the obtained bispecific antigen-binding molecule, the FX activation promoting activity by F.XIa was evaluated by a measurement system consisting of F.XIa, FX, F.Xa synthetic substrate (S-2222), and phospholipid. bottom. Based on the results, as the bispecific antigen-binding molecule to have, in principle, a molecule showing F.X activation promoting activity by F.XIa of 0.1 or more in this measurement system was selected. The F.X activation promoting activity by F.XIa referred to here is a value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.

 上記で選択された二重特異性抗原結合分子あるいはその類縁の二重特異性抗原結合分子に関しては、血液凝固因子欠乏ヒト血漿を用いた凝固時間測定系を用い、凝固回復能を測定した。その結果、抗体無添加時に比べて、凝固時間を短縮する二重特異性抗原結合分子を得た。ここでいう凝固時間は、血液凝固第九因子欠乏ヒト血漿を用いた活性化部分トロンボプラスチン時間を測定したものである。それら二重特異性抗原結合分子の中で、最も好ましい二重特異性抗原結合分子は60秒以上の凝固時間短縮能を有していた。 For the bispecific antigen-binding molecule selected above or its related bispecific antigen-binding molecule, the coagulation recovery ability was measured using a coagulation time measurement system using blood coagulation factor-deficient human plasma. As a result, a bispecific antigen-binding molecule having a shorter coagulation time than when no antibody was added was obtained. The coagulation time referred to here is a measurement of the activated partial thromboplastin time using human plasma deficient in blood coagulation factor IX. Among these bispecific antigen-binding molecules, the most preferable bispecific antigen-binding molecule had the ability to shorten the coagulation time by 60 seconds or more.

 本発明の抗原結合分子は、特に制限されず、表1に記載のいずれかの組合せの酵素及び基質の両方を認識する二重特異性抗原結合分子が挙げられる。 The antigen-binding molecule of the present invention is not particularly limited, and examples thereof include bispecific antigen-binding molecules that recognize both the enzyme and the substrate of any combination shown in Table 1.

 本発明の抗原結合分子は、表1に記載の活性化血液凝固第X因子(F.Xa)と血液凝固第X因子 (F.X)との組み合わせ、活性化血液凝固第XI因子(F.XIa)と血液凝固第XI因子(F.XIa)との組み合わせの場合には、同じ重鎖、軽鎖を持つ単一特異性抗原結合分子であってもよく、より好ましくは抗体である。 The antigen-binding molecule of the present invention is a combination of activated blood coagulation factor X (F.Xa) and blood coagulation factor X (FX) shown in Table 1, and activated blood coagulation factor XI (F.XIa). In the case of the combination of blood coagulation factor XI (F.XIa), it may be a monospecific antigen-binding molecule having the same heavy chain and light chain, and more preferably an antibody.

 本発明で開示されている可変領域を用いて全長抗体を作製する場合、定常領域は特に制限されず、当業者に公知の定常領域を用いることが可能であり、例えば、Sequences of proteins of immunological interest, (1991), U.S. Department of Health and Human Services. Public Health Service National Institutes of Healthや、An efficient route to human bispecific IgG, (1998). Nature Biotechnology vol. 16, 677-681、等に記載されている定常領域を用いることができる。 When a full-length antibody is produced using the variable region disclosed in the present invention, the constant region is not particularly limited, and constant regions known to those skilled in the art can be used. For example, Sequences of proteins of immunological interest. , (1991), US Department of Health and Human Services. Public Health Service National Institutes of Health, An efficiency route to human bispecific IgG, (1998). Nature Biotechnology vol. 16, 677-681, etc. Constant regions can be used.

 本発明における抗原結合分子の1つの態様としては、抗原結合分子は少なくとも生体に発現するペプチド性補因子またはヘパリンの機能を代替しないが、酵素と該酵素の触媒反応を受け得る基質の関係において補因子様の機能を果たすことから、ある補因子、酵素、基質の活性(機能)低下や欠損に起因する疾病に対して、有効な薬剤となることが期待される。本発明の抗原結合分子が結合する酵素及び基質が血液凝固線溶関連因子である場合には、上記疾病として、例えば、出血、出血を伴う疾患、もしくは出血に起因する疾患等を挙げることができる。特に、F.VIII/F.VIIIa、F.IX/F.IXa、F.XI/F.XIaの機能低下や欠損は、血友病と呼ばれる出血異常症を引き起こすことで知られる。 In one aspect of the antigen-binding molecule in the present invention, the antigen-binding molecule does not at least replace the function of a peptide cofactor or heparin expressed in the body, but complements the relationship between the enzyme and a substrate capable of undergoing a catalytic reaction of the enzyme. Since it fulfills factor-like functions, it is expected to be an effective drug for diseases caused by decreased activity (function) or deficiency of certain cofactors, enzymes, and substrates. When the enzyme and substrate to which the antigen-binding molecule of the present invention binds are blood coagulation / fibrinolysis-related factors, examples of the above-mentioned diseases include bleeding, diseases associated with bleeding, and diseases caused by bleeding. .. In particular, functional deterioration or deficiency of F.VIII / F.VIIIa, F.IX / F.IXa, and F.XI / F.XIa is known to cause hemorrhagic disorders called hemophilia.

 血友病のうち、先天性のF.VIII/F.VIIIa機能低下または欠損による出血異常症は血友病Aと呼ばれ、F.IX/F.IXa機能低下または欠損による出血異常症は血友病Bと呼ばれる。血友病A患者が出血した場合はF.VIII製剤の補充療法が行われ、血友病B患者が出血した場合はF.IX製剤の補充療法が行われる。また、激しい運動や遠足の当日、頻回に関節内出血を来たす場合、あるいは重症血友病に分類される場合には、これらの製剤の予防投与が行われることがある。 Among hemophilia, bleeding dysfunction due to congenital F.VIII / F.VIIIa dysfunction or deficiency is called hemophilia A, and bleeding dysfunction due to F.IX / F.IXa dysfunction or deficiency is blood. Called friendship B. If hemophilia A patients bleed, F.VIII replacement therapy is given, and if hemophilia B patients bleed, F.IX replacement therapy is given. In addition, on the day of strenuous exercise or excursion, if frequent intra-articular bleeding occurs, or if it is classified as severe hemophilia, prophylactic administration of these preparations may be performed.

 F.VIII製剤の血中半減期は短く、約12 ~ 16時間程度である。それ故、継続的な予防のためには、F.VIII製剤を週に3回程度投与する必要がある。これは、F.VIII活性として、概ね1%以上を維持することに相当する。同様に、標準的な半減期を有するF.IX製剤は、週に2回程度投与する必要がある。また、出血時の補充療法においても、出血が軽度な場合を除き、再出血を防ぎ、完全な止血を行うため、一定期間、F.VIII製剤やF.IX製剤を定期的に追加投与する必要がある。 The half-life of F.VIII preparation in blood is short, about 12 to 16 hours. Therefore, for continuous prophylaxis, it is necessary to administer the F.VIII preparation about three times a week. This corresponds to maintaining approximately 1% or more of F.VIII activity. Similarly, the F.IX preparation with a standard half-life should be administered about twice a week. In addition, in replacement therapy for bleeding, it is necessary to administer additional F.VIII and F.IX products regularly for a certain period of time in order to prevent rebleeding and completely stop bleeding, except when the bleeding is mild. There is.

 また、F.VIII製剤及びF.IX製剤は、静脈内に投与される。静脈内投与実施には、技術的な困難さが存在する。特に年少の患者に対する投与においては、投与に用いられる静脈が細い故、困難さが一層増す。 In addition, the F.VIII preparation and the F.IX preparation are administered intravenously. There are technical difficulties in performing intravenous administration. Especially in the case of administration to younger patients, the difficulty is further increased because the veins used for administration are thin.

 前述の、F.VIII製剤及びF.IX製剤の予防投与や、出血の際の緊急投与においては、多くの場合、家庭療法・自己注射が用いられる。頻回投与の必要性と、投与の際の技術的困難さは、投与に際し患者に苦痛を与えるだけでなく、家庭療法・自己注射の普及を妨げる要因となっている。
 従って、現存の血液凝固第VIII因子製剤及び第IX因子製剤に比し、投与間隔が広い薬剤、あるいは投与が簡単な薬剤が、強く求められていた。
In the above-mentioned prophylactic administration of F.VIII and F.IX preparations and emergency administration in the event of bleeding, home remedies and self-injection are often used. The need for frequent administration and the technical difficulty of administration not only cause pain to patients during administration, but also hinder the spread of home remedies and self-injection.
Therefore, there has been a strong demand for a drug having a wider administration interval or an easy-to-administer drug as compared with the existing blood coagulation factor VIII and factor IX preparations.

 さらに、血友病A患者、特に重症血友病A患者には、インヒビターと呼ばれるF.VIIIに対する抗体が発生する場合がある。同様に、血友病B患者には、インヒビターと呼ばれるF.IXに対する抗体が発生する場合がある。インヒビターが発生すると、F.VIII製剤またはF.IX製剤の効果がインヒビターにより妨げられる。その結果、患者に対する止血管理が非常に困難になる。 Furthermore, hemophilia A patients, especially severe hemophilia A patients, may develop antibodies against F.VIII called inhibitors. Similarly, patients with hemophilia B may develop antibodies to F.IX called inhibitors. When the inhibitor is generated, the effect of the F.VIII or F.IX preparation is hindered by the inhibitor. As a result, hemostasis management for the patient becomes very difficult.

 このような血友病Aインヒビター患者が出血を来たした場合は、通常、大量のF.VIII製剤を用いる中和療法か、複合体製剤(complex concentrate)あるいはF.VIIa製剤を用いるバイパス療法が、実施される。しかしながら、中和療法では、大量のF.VIII製剤の投与が、逆に、インヒビター(抗F.VIII抗体)力価を上げてしまう場合がある。また、バイパス療法では、複合体製剤やF.VIIa製剤の短い血中半減期(約2 ~ 8時間)が問題となっている。その上、それらの作用機序が、F.VIII/F.VIIIaの機能、すなわちF.IXaによるF.X活性化を触媒する機能に非依存的であるため、場合によっては、止血機構をうまく機能させられず、不応答になってしまうケースがある。そのため、血友病Aインヒビター患者では、非インヒビター血友病A患者に比し、十分な止血効果を得られない場合が多いのである。血友病Bインヒビター患者についても同様である。 When such hemophilia A inhibitor patients develop bleeding, neutralization therapy using a large amount of F.VIII preparation, or bypass therapy using a complex concentrate or F.VIIa preparation is usually performed. , Will be implemented. However, in neutralization therapy, administration of a large amount of the F.VIII preparation may conversely increase the titer of the inhibitor (anti-F.VIII antibody). In bypass therapy, the short half-life (about 2 to 8 hours) of complex preparations and F.VIIa preparations has become a problem. Moreover, their mechanism of action is independent of the function of F.VIII / F.VIIIa, that is, the function of catalyzing FX activation by F.IXa, which in some cases makes the hemostatic mechanism work well. There are cases where it is not possible and the response becomes unresponsive. Therefore, hemophilia A inhibitor patients often do not have a sufficient hemostatic effect as compared to non-inhibitor hemophilia A patients. The same is true for patients with hemophilia B inhibitors.

 従って、インヒビターの存在に左右されず、且つF.VIII/F.VIIIaまたはF.IX/F.IXaの機能を代替する薬剤が、強く求められていた。 Therefore, there has been a strong demand for a drug that is independent of the presence of the inhibitor and that replaces the function of F.VIII / F.VIIIa or F.IX / F.IXa.

 さて、(i)投与間隔が広く、(ii)投与が簡単であり、(iii)インヒビターの存在に左右されず、(iv) F.VIII/F.VIIIaまたはF.IX/F.IXa非依存的にその機能を代替する医薬品の創製には、抗体を利用する方法が考えられる。抗体の血中半減期は、一般に、比較的長く、数日から数週間である。また、抗体は、一般に、皮下投与後に血中に移行することが知られている。すなわち、一般に抗体医薬品は、上記の(i)、(ii)を満たしている。 Now, (i) the dosing interval is wide, (ii) the dosing is easy, (iii) independent of the presence of the inhibitor, and (iv) F.VIII / F.VIIIa or F.IX / F.IXa independent. A method using an antibody can be considered for the creation of a drug that replaces the function. The half-life of an antibody in blood is generally relatively long, ranging from days to weeks. In addition, it is generally known that the antibody is transferred to the blood after subcutaneous administration. That is, in general, antibody drugs satisfy the above (i) and (ii).

 本発明では、本発明の抗原結合分子を有効成分として含有する医薬組成物を提供する。例えば、本発明の抗原結合分子が表1に記載の組合せの酵素及び基質の両方を認識する抗原結合分子のうち、該血液凝固因子欠乏血漿において血液凝固反応を促進する抗原結合分子である場合には、該抗原結合分子は、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防あるいは治療のための医薬品(医薬組成物)もしくは薬剤となることが期待される。 The present invention provides a pharmaceutical composition containing the antigen-binding molecule of the present invention as an active ingredient. For example, when the antigen-binding molecule of the present invention is an antigen-binding molecule that recognizes both the enzymes and substrates of the combinations shown in Table 1 and promotes the blood coagulation reaction in the blood coagulation factor-deficient plasma. The antigen-binding molecule is expected to be a drug (pharmaceutical composition) or drug for preventing or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding.

 治療または予防目的で使用される本発明の抗原結合分子を有効成分として含む医薬組成物は、必要に応じて、それらに対して不活性な適当な薬学的に許容される担体、媒体等と混和して製剤化することができる。例えば、滅菌水や生理食塩水、安定剤、賦形剤、酸化防止剤(アスコルビン酸等)、緩衝剤(リン酸、クエン酸、他の有機酸等)、防腐剤、界面活性剤(PEG、Tween等)、キレート剤(EDTA等)、結合剤等を挙げることができる。また、その他の低分子量のポリペプチド、血清アルブミン、ゼラチンや免疫グロブリン等の蛋白質、グリシン、グルタミン、アスパラギン、アルギニン及びリシン等のアミノ酸、多糖及び単糖等の糖類や炭水化物、マンニトールやソルビトール等の糖アルコールを含んでいてもよい。注射用の水溶液とする場合には、例えば生理食塩水、ブドウ糖やその他の補助薬を含む等張液、例えば、D-ソルビトール、D-マンノース、D-マンニトール、塩化ナトリウムが挙げられ、適当な溶解補助剤、例えばアルコール(エタノール等)、ポリアルコール(プロピレングリコール、PEG等)、非イオン性界面活性剤(ポリソルベート80、HCO-50)等と併用してもよい。 The pharmaceutical composition containing the antigen-binding molecule of the present invention used for therapeutic or prophylactic purposes as an active ingredient is optionally mixed with an appropriate pharmaceutically acceptable carrier, medium or the like which is inert to them. Can be formulated. For example, sterile water, physiological saline, stabilizers, excipients, antioxidants (ascorbic acid, etc.), buffers (phosphate, citric acid, other organic acids, etc.), preservatives, surfactants (PEG, etc.) Tween etc.), chelating agent (EDTA etc.), binder and the like can be mentioned. In addition, other low molecular weight polypeptides, serum albumin, proteins such as gelatin and immunoglobulin, amino acids such as glycine, glutamine, asparagine, arginine and lysine, sugars and carbohydrates such as polysaccharides and monosaccharides, sugars such as mannitol and sorbitol. It may contain alcohol. Examples of an aqueous solution for injection include physiological saline, isotonic solutions containing glucose and other adjuvants, such as D-sorbitol, D-mannitol, D-mannitol, and sodium chloride, which are appropriately dissolved. Auxiliary agents such as alcohol (ethanol, etc.), polyalcohol (propylene glycol, PEG, etc.), nonionic surfactant (polysorbate 80, HCO-50), etc. may be used in combination.

 また、必要に応じ本発明の抗原結合分子をマイクロカプセル(ヒドロキシメチルセルロース、ゼラチン、ポリ[メチルメタクリル酸]等のマイクロカプセル)に封入したり、コロイドドラッグデリバリーシステム(リポソーム、アルブミンミクロスフェア、マイクロエマルジョン、ナノ粒子及びナノカプセル等)とすることもできる("Remington's Pharmaceutical Science 16th edition", Oslo Ed. (1980)等参照)。さらに、薬剤を徐放性の薬剤とする方法も公知であり、本発明の抗原結合分子に適用し得る(Langer et al., J.Biomed.Mater.Res. 15: 267-277 (1981); Langer, Chemtech. 12: 98-105 (1982);米国特許第3,773,919号;欧州特許出願公開(EP)第58,481号; Sidman et al., Biopolymers 22: 547-556 (1983);EP第133,988号)。 In addition, if necessary, the antigen-binding molecule of the present invention can be encapsulated in microcapsules (microcapsules of hydroxymethyl cellulose, gelatin, poly [methyl methacrylate], etc.), or a colloidal drug delivery system (liposomes, albumin microspheres, microemulsions, etc.). Nanoparticles and nanocapsules, etc.) (see "Remington's Pharmaceutical Science 16th edition", Oslo Ed. (1980), etc.). Furthermore, a method of making a drug a sustained release drug is also known and can be applied to the antigen-binding molecule of the present invention (Langer et al., J. Biomed. Mater. Res. 15: 267-277 (1981); Langer, Chemtech. 12: 98-105 (1982); US Patent No. 3,773,919; European Patent Application Publication (EP) No. 58,481; Sidman et al., Biopolymers 22: 547-556 (1983); EP No. 133,988) ..

 本発明の抗原結合分子または組成物は、血液凝固第VIII因子と併用することができる。血液凝固第VIII因子はヒトの血液から作られたものであっても、遺伝子組み換えにより作られたものであってもよい。本発明の抗原結合分子または組成物は、血液凝固第VIII因子と同時に投与してもよく、または、時期をずらして投与してもよい。また、本発明の抗原結合分子または医薬組成物と血液凝固第VIII因子を組み合わせたキットとして実施してもよい。さらに、本発明の抗原結合分子または医薬組成物と血液凝固第VIII因子を併用する場合は、いずれかを単独で用いる場合に比べて、所望により各々の投与量を少なくすることも可能である。 The antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor VIII. Blood coagulation factor VIII may be made from human blood or genetically modified. The antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor VIII, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor VIII. Furthermore, when the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with blood coagulation factor VIII, it is possible to reduce the dose of each if desired, as compared with the case where either of them is used alone.

 本発明の抗原結合分子または組成物は、血液凝固第IX因子と併用することができる。血液凝固第IX因子はヒトの血液から作られたものであっても、遺伝子組み換えにより作られたものであってもよい。本発明の抗原結合分子または組成物は、血液凝固第IX因子と同時に投与してもよく、または、時期をずらして投与してもよい。また、本発明の抗原結合分子または医薬組成物と血液凝固第IX因子を組み合わせたキットとして実施してもよい。さらに、本発明の抗原結合分子または医薬組成物と血液凝固第IX因子を併用する場合は、いずれかを単独で用いる場合に比べて、所望により各々の投与量を少なくすることも可能である。 The antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor IX. Blood coagulation factor IX may be made from human blood or genetically modified. The antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor IX, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor IX. Furthermore, when the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with blood coagulation factor IX, it is possible to reduce the dose of each if desired, as compared with the case where either of them is used alone.

 本発明の抗原結合分子または組成物は、血液凝固第XI因子と併用することができる。血液凝固第XI因子はヒトの血液から作られたものであっても、遺伝子組み換えにより作られたものであってもよい。本発明の抗原結合分子または組成物は、血液凝固第XI因子と同時に投与してもよく、または、時期をずらして投与してもよい。また、本発明の抗原結合分子または医薬組成物と血液凝固第XI因子を組み合わせたキットとして実施してもよい。さらに、本発明の抗原結合分子または医薬組成物と血液凝固第XI因子を併用する場合は、いずれかを単独で用いる場合に比べて、所望により各々の投与量を少なくすることも可能である。 The antigen-binding molecule or composition of the present invention can be used in combination with blood coagulation factor XI. Blood coagulation Factor XI may be made from human blood or genetically modified. The antigen-binding molecule or composition of the present invention may be administered at the same time as blood coagulation factor XI, or may be administered at staggered times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with blood coagulation factor XI. Furthermore, when the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with factor XI for blood coagulation, it is possible to reduce the dose of each dose as desired, as compared with the case where either one is used alone.

 本発明の抗原結合分子または組成物は、バイパス製剤と併用することができる。バイパス製剤はヒトの血液から作られたものであっても、遺伝子組み換えにより作られたものであってもよく、例えば血漿由来活性型プロトロンビン複合体製剤(APCC製剤)、遺伝子組換え型活性化第VII因子製剤(rF.VIIa製剤)、乾燥濃縮人血液凝固第X因子加活性化第VII因子製剤(F.VIIa/F.X製剤)である。本発明の抗原結合分子または組成物は、バイパス製剤と同時に投与してもよく、または、時期をずらして投与してもよい。また、本発明の抗原結合分子または医薬組成物とバイパス製剤を組み合わせたキットとして実施してもよい。さらに、本発明の抗原結合分子または医薬組成物とバイパス製剤を併用する場合は、いずれかを単独で用いる場合に比べて、所望により各々の投与量を少なくすることも可能である。 The antigen-binding molecule or composition of the present invention can be used in combination with a bypass preparation. The bypass preparation may be made from human blood or by gene recombination, for example, plasma-derived active prothrombin complex preparation (APCC preparation), gene recombination type activation first. Factor VII preparation (rF.VIIa preparation) and dry concentrated human blood coagulation factor X-activated factor VII preparation (F.VIIa / FX preparation). The antigen-binding molecule or composition of the present invention may be administered at the same time as the bypass preparation, or may be administered at different times. Further, it may be carried out as a kit in which the antigen-binding molecule or pharmaceutical composition of the present invention is combined with a bypass preparation. Furthermore, when the antigen-binding molecule or pharmaceutical composition of the present invention is used in combination with the bypass preparation, it is possible to reduce the dose of each, if desired, as compared with the case where either of them is used alone.

 本発明の医薬組成物の投与量は、剤型の種類、投与方法、患者の年齢や体重、患者の症状、疾患の種類や進行の程度等を考慮して、最終的には医師の判断により適宜決定されるものであるが、一般に大人では、1日当たり、0.1~2000mgを1~数回に分けて投与することができる。より好ましくは1~1000mg/日、更により好ましくは50~500mg/日、最も好ましくは100~300mg/日である。これらの投与量は患者の体重や年齢、投与方法などにより変動するが、当業者であれば適当な投与量を適宜選択することが可能である。投与期間も、患者の治癒経過等に応じて適宜決定することが好ましい。 The dose of the pharmaceutical composition of the present invention is finally determined by a doctor in consideration of the type of dosage form, administration method, age and weight of the patient, symptoms of the patient, type of disease and degree of progression, etc. Although it is determined as appropriate, in general, for adults, 0.1 to 2000 mg can be administered in 1 to several divided doses per day. It is more preferably 1 to 1000 mg / day, even more preferably 50 to 500 mg / day, and most preferably 100 to 300 mg / day. These doses vary depending on the body weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose. It is preferable that the administration period is also appropriately determined according to the healing course of the patient and the like.

 また、本発明の抗原結合分子をコードする遺伝子を遺伝子治療用ベクターに組込み、遺伝子治療を行うことも考えられる。投与方法としては、nakedプラスミドによる直接投与の他、リポソーム等にパッケージングするか、レトロウィルスベクター、アデノウィルスベクター、ワクシニアウィルスベクター、ポックスウィルスベクター、アデノウィルス関連ベクター、HVJベクター等の各種ウィルスベクターとして形成するか(Adolph『ウィルスゲノム法』, CRC Press, Florid (1996)参照)、または、コロイド金粒子等のビーズ担体に被覆(WO93/17706等)して投与することができる。また、本発明の抗原結合分子をコードする核酸は直接生体に投与してもよく、またエレクトロポレーション法により直接生体に投与してもよい。例えば、本発明の抗原結合分子をコードするmRNAに生体内でのmRNAの安定性を高めるための化学修飾を施し、当該mRNAをヒトに直接投与し、生体内で本発明の抗原結合分子を発現させる方法により本発明の抗原結合分子を投与しうる(EP2101823B、WO2013/120629参照)。しかしながら、生体内において抗原結合分子が発現され、その作用を発揮できる限りいかなる方法により投与してもよい。好ましくは、適当な非経口経路(静脈内、腹腔内、皮下、皮内、脂肪組織内、乳腺組織内、吸入または筋肉内の経路を介して注射、注入、またはガス誘導性粒子衝撃法(電子銃等による)、点鼻薬等粘膜経路を介する方法等)により十分な量が投与される。ex vivoにおいてリポソームトランスフェクション、粒子衝撃法(米国特許第4,945,050号)、またはウィルス感染を利用して血液細胞及び骨髄由来細胞等に投与して、該細胞を動物に再導入することにより本発明の抗原結合分子をコードする遺伝子を投与してもよい。 It is also conceivable to incorporate the gene encoding the antigen-binding molecule of the present invention into a gene therapy vector to perform gene therapy. As an administration method, in addition to direct administration with a naked plasmid, it is packaged in liposomes or the like, or formed as various virus vectors such as retrovirus vector, adenovirus vector, vaccinia virus vector, poxvirus vector, adenovirus-related vector, and HVJ vector. (See Adolph "Virus Genome Method", CRC Press, Florid (1996)), or it can be administered by coating it on a bead carrier such as colloidal gold particles (WO93 / 17706, etc.). Further, the nucleic acid encoding the antigen-binding molecule of the present invention may be directly administered to a living body, or may be directly administered to a living body by an electroporation method. For example, the mRNA encoding the antigen-binding molecule of the present invention is chemically modified to enhance the stability of the mRNA in vivo, and the mRNA is directly administered to humans to express the antigen-binding molecule of the present invention in vivo. The antigen-binding molecule of the present invention can be administered by the method of causing (see EP2101823B, WO2013 / 120629). However, as long as the antigen-binding molecule is expressed in vivo and exerts its action, it may be administered by any method. Preferably, injection, infusion, or gas-induced particle impact (electron) is performed via a suitable parenteral route (intravenous, intraperitoneal, subcutaneous, intradermal, intra-adipose tissue, intramammary gland tissue, inhalation or intramuscular route). A sufficient amount is administered by (with a gun, etc.), a method via a mucosal route such as a nasal spray, etc.). The present invention is produced by administering to blood cells, bone marrow-derived cells, etc. using liposome transfection, particle impact method (US Patent No. 4,945,050), or virus infection in ex vivo, and reintroducing the cells into animals. A gene encoding an antigen-binding molecule may be administered.

 また本発明は、本発明の抗原結合分子もしくは組成物を投与する工程を含む、出血、出血を伴う疾患、または出血に起因する疾患の予防および/または治療するための方法を提供する。抗原結合分子もしくは組成物の投与は、例えば、前記の方法により実施することができる。
 一態様において、出血、出血を伴う疾患、または出血に起因する疾患は、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患(例えば、血友病B)である。特定の態様において、当該疾患は、血液凝固第IX因子もしくは活性化血液凝固第IX因子に対するインヒビターが出現している疾患である。特定の態様において、本発明の抗原結合分子または組成物は、血液凝固第IX因子または活性化血液凝固第IX因子に対するインヒビターを保有する対象(インヒビター保有患者)に投与される。特定の態様において、本発明の方法は、血液凝固第IX因子を投与する工程をさらに含む。
 別の態様において、出血、出血を伴う疾患、または出血に起因する疾患は、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患(例えば、血友病A、後天性血友病、またはフォンビルブランド病)である。特定の態様において、当該疾患は、血液凝固第第VIII因子もしくは活性化血液凝固第第VIII因子に対するインヒビターが出現している疾患である。特定の態様において、本発明の抗原結合分子または組成物は、血液凝固第第VIII因子または活性化血液凝固第第VIII因子に対するインヒビターを保有する対象(インヒビター保有患者)に投与される。特定の態様において、本発明の方法は、血液凝固第VIII因子を投与する工程をさらに含む。
 別の態様において、出血、出血を伴う疾患、または出血に起因する疾患は、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患(例えば、血友病C、または後天性血友病)である。特定の態様において、当該疾患は、血液凝固第XI因子もしくは活性化血液凝固第XI因子に対するインヒビターが出現している疾患である。特定の態様において、本発明の抗原結合分子または組成物は、血液凝固第XI因子または活性化血液凝固第XI因子に対するインヒビターを保有する対象(インヒビター保有患者)に投与される。特定の態様において、本発明の方法は、血液凝固第XI因子を投与する工程をさらに含む。
The present invention also provides a method for preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding, which comprises the step of administering the antigen-binding molecule or composition of the present invention. Administration of the antigen-binding molecule or composition can be carried out, for example, by the method described above.
In one aspect, bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to decreased or deficient activity of blood coagulation factor IX and / or activated blood coagulation factor IX (eg,). , Hemophilia B). In certain embodiments, the disease is a disease in which an inhibitor of blood coagulation factor IX or activated blood coagulation factor IX has emerged. In certain embodiments, the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor IX or activated blood coagulation factor IX. In certain embodiments, the method of the invention further comprises the step of administering blood coagulation factor IX.
In another embodiment, bleeding, a disease associated with bleeding, or a disease resulting from bleeding is a disease that develops and / or progresses due to decreased or deficient activity of hemocoagulation factor VIII and / or activated hemocoagulation factor VIII. For example, hemophilia A, acquired hemophilia, or Fonville brand disease). In certain embodiments, the disease is a disease in which an inhibitor of blood coagulation factor VIII or activated blood coagulation factor VIII has emerged. In certain embodiments, the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor VIII or activated blood coagulation factor VIII. In certain embodiments, the method of the invention further comprises the step of administering blood coagulation factor VIII.
In another embodiment, bleeding, a disease associated with bleeding, or a disease resulting from bleeding is a disease that develops and / or progresses due to decreased or deficient activity of blood coagulation factor XI and / or activated blood coagulation factor XI. For example, hemophilia C, or acquired hemophilia). In a particular embodiment, the disease is a disease in which an inhibitor of blood coagulation factor XI or activated blood coagulation factor XI appears. In certain embodiments, the antigen-binding molecule or composition of the invention is administered to a subject (patient carrying an inhibitor) who carries an inhibitor of blood coagulation factor XI or activated blood coagulation factor XI. In certain embodiments, the method of the invention further comprises the step of administering blood coagulation factor XI.

 また本発明は、本発明の抗原結合分子の、本発明の(医薬)組成物の製造のための使用に関する。 The present invention also relates to the use of the antigen-binding molecule of the present invention for the production of the (pharmaceutical) composition of the present invention.

 さらに本発明は、少なくとも本発明の抗原結合分子もしくは組成物を含む、上記方法に用いるためのキットを提供する。該キットには、その他、注射筒、注射針、薬学的に許容される媒体、アルコール綿布、絆創膏、または使用方法を記載した指示書等をパッケージしておくこともできる。一態様において、本発明のキットは、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのものである。特定の態様において、本発明のキットは、本発明の抗原結合分子もしくは組成物に加えて、血液凝固第IX因子または第VIII因子を含む。 Furthermore, the present invention provides a kit for use in the above method, which comprises at least the antigen-binding molecule or composition of the present invention. The kit may also be packaged with a syringe, a needle, a pharmaceutically acceptable medium, an alcohol cotton cloth, an adhesive plaster, or instructions describing how to use it. In one aspect, the kit of the present invention is for use in a method of preventing and / or treating bleeding, a disease associated with bleeding, or a disease caused by bleeding. In certain embodiments, the kit of the invention comprises a blood coagulation factor IX or factor VIII in addition to the antigen binding molecule or composition of the invention.

 別の局面において、本発明は、表1に記載のいずれかの組合せの酵素及び基質の両方を認識する二重特異性抗原結合分子を用いて血液凝固を促進する方法に関する。当該局面の一態様において、本発明の血液凝固促進方法は、表1に記載のいずれかの組合せの酵素及び基質の両方を認識する二重特異性抗原結合分子を投与する工程を含む。抗原結合分子の投与は、例えば、前記の投与方法により実施することができる。前記局面の特定の態様において、本発明の血液凝固促進方法に用いられる二重特異性抗原結合分子は、活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子である。 In another aspect, the present invention relates to a method of promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both the enzyme and the substrate of any combination shown in Table 1. In one aspect of this aspect, the method for promoting blood coagulation of the present invention comprises the step of administering a bispecific antigen-binding molecule that recognizes both the enzyme and the substrate of any combination shown in Table 1. The administration of the antigen-binding molecule can be carried out by, for example, the above-mentioned administration method. In a particular aspect of the above aspect, the bispecific antigen binding molecule used in the method for promoting blood coagulation of the present invention is a bispecific antigen that recognizes both activated blood coagulation factor XI and blood coagulation factor X. It is a binding molecule.

 さらにまた別の局面において、本発明は、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法に関する。当該局面の一態様において、本発明のスクリーニング方法は、(1)試験物質と、表1に記載のいずれかの組合せの酵素との結合を評価する工程、および(2)試験物質と、表1に記載の当該組合せの基質との結合を評価する工程を含む。特定の態様において、本発明のスクリーニング方法は、(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および(2)試験物質と血液凝固第X因子との結合を評価する工程を含む。特定の態様において、本発明のスクリーニング方法は、表1に記載のいずれかの組合せの酵素及び基質の両方と結合する試験物質を、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質の候補として選択する工程をさらに含む。 In yet another aspect, the present invention relates to a method for screening a substance effective for the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding. In one aspect of this aspect, the screening method of the present invention comprises (1) a step of evaluating the binding of the test substance to any combination of enzymes shown in Table 1, and (2) the test substance and Table 1. Includes a step of evaluating the binding of the combination to the substrate according to. In certain embodiments, the screening methods of the invention assess (1) the step of assessing the binding of the test substance to activated blood coagulation factor XI, and (2) assessing the binding of the test substance to blood coagulation factor X. Includes the process of In certain embodiments, the screening methods of the present invention use a test substance that binds to both the enzyme and substrate of any of the combinations listed in Table 1 to prevent bleeding, bleeding-related diseases, or diseases caused by bleeding. / Or further includes the step of selecting as a candidate for a therapeutically effective substance.

 「試験物質」は、特に限定されるものではなく、例えば天然化合物、有機化合物、無機化合物、核酸、タンパク質、ペプチド等の単一物質、並びに化合物ライブラリー、核酸ライブラリー、ペプチドライブラリー、遺伝子ライブラリーの発現産物、細胞抽出物、細胞培養上清、発酵微生物産生物、海洋生物抽出物、植物抽出物、原核細胞抽出物、真核単細胞抽出物もしくは動物細胞抽出物等を挙げることができる。上記試験物質は必要に応じて適宜標識して用いることができる。標識としては、例えば、放射標識、蛍光標識等を挙げることができる。また、上記試験物質に加えて、これらの試験物質を複数種混合した混合物も含まれる。 The "test substance" is not particularly limited, and includes, for example, a single substance such as a natural compound, an organic compound, an inorganic compound, a nucleic acid, a protein, and a peptide, as well as a compound library, a nucleic acid library, a peptide library, and a gene live. Examples thereof include rally expression products, cell extracts, cell culture supernatants, fermented microbial products, marine organism extracts, plant extracts, prokucleic acid cell extracts, eukaryotic single cell extracts, animal cell extracts and the like. The test substance can be appropriately labeled and used as needed. Examples of the label include a radial label, a fluorescent label and the like. In addition to the above test substances, a mixture of a plurality of these test substances is also included.

 さらにまた別の局面において、本発明は、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法に関する。当該局面の一態様において、本発明の品質試験方法は、(1)試験物質もしくは試験組成物と、表1に記載のいずれかの組合せの酵素との結合を評価する工程、および(2)試験物質もしくは試験組成物と、表1に記載の当該組合せの基質との結合を評価する工程を含む。特定の態様において、本発明の品質試験方法は、(1)試験物質もしくは試験組成物と活性化血液凝固第XI因子との結合を評価する工程、および(2)試験物質もしくは試験組成物と血液凝固第X因子との結合を評価する工程を含む。特定の態様において、本発明の品質試験法は、表1に記載のいずれかの組合せの酵素及び基質の両方と結合する試験物質もしくは組成物を、出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物としての品質を評価する工程をさらに含む。 In yet another aspect, the present invention relates to a quality test method for a substance or composition effective in the prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding. In one aspect of this aspect, the quality test method of the present invention comprises (1) a step of evaluating the binding of a test substance or test composition to an enzyme of any combination shown in Table 1, and (2) a test. The step of evaluating the binding between the substance or the test composition and the substrate of the combination shown in Table 1 is included. In a particular embodiment, the quality test method of the present invention comprises (1) a step of evaluating the binding of the test substance or test composition to activated blood coagulation factor XI, and (2) the test substance or test composition and blood. Includes the step of assessing binding to coagulation factor X. In certain embodiments, the quality test method of the invention results in bleeding, a disease associated with bleeding, or bleeding that binds a test substance or composition that binds to both of the enzymes and substrates of any of the combinations listed in Table 1. It further comprises the step of assessing the quality of the substance or composition as effective for the prevention and / or treatment of the disease.

 「試験組成物」は、特に限定されるものではなく、例えば医薬品、試薬などが挙げられる。 The "test composition" is not particularly limited, and examples thereof include pharmaceuticals and reagents.

 (1)試験物質と活性化血液凝固第XI因子との結合を評価する工程としては、例えば
、ELISA法(Enzyme-Linked Immuno Sorbent Assay)、表面プラズモン共鳴(Surface plasmon resonance:SPR)法、例えばBiacore シリーズ(GE Healthcare)を用いた測定、バイオセンサー技術(BLI法)を用いた生体分子間相互作用解析システム、例えばOctetシステム(Fortebio社)によって評価できる。
(1) Examples of the steps for evaluating the binding between the test substance and the activated blood coagulation factor XI include an ELISA method (Enzyme-Linked Immuno Sorbent Assay) and a surface plasmon resonance (SPR) method, for example, Biacore. It can be evaluated by measurement using a series (GE Healthcare) and biosensor interaction analysis system using biosensor technology (BLI method), for example, Octet system (Fortebio).

 (2)試験物質と血液凝固第X因子との結合を評価する工程としては、例えば、ELISA法(Enzyme-Linked Immuno Sorbent Assay)、表面プラズモン共鳴(Surface plasmon resonance:SPR)法、例えばBiacore シリーズ(GE Healthcare)を用いた測定、バイオセンサー技術(BLI法)を用いた生体分子間相互作用解析システム、例えばOctetシステム(Fortebio社)によって評価できる。 (2) Examples of the steps for evaluating the binding between the test substance and blood coagulation factor X include an ELISA method (Enzyme-Linked ImmunoSorbent Assay) and a surface plasmon resonance (SPR) method, for example, the Biacore series (Biacore series). It can be evaluated by measurement using GE Healthcare) and biosensor interaction analysis system using biosensor technology (BLI method), for example, Octet system (Fortebio).

 (3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程としては、例えば、該酵素、該基質、活性化された基質の合成基質、リン脂質、Ca2+から成る測定系で、該酵素による該基質活性化促進活性で評価できる。その結果を以って、該活性を有する二重特異性抗原結合分子として、原則本測定系で該酵素添加群のみ0.1以上の該酵素による該基質活性化促進活性を示したものを選択できる。なお、ここでいう該酵素による該基質活性化促進活性とは、抗原結合分子溶液の合成基質添加30分後の吸光度の値で測定することができる。 (3) Activation As a step of evaluating the activation reaction of blood coagulation factor X by factor XI using a test substance, for example, the enzyme, the substrate, the synthetic substrate of the activated substrate, phosphorus It is a measurement system consisting of lipid and Ca2 +, and can be evaluated by the substrate activation promoting activity by the enzyme. Based on the results, as a bispecific antigen-binding molecule having the activity, in principle, a molecule showing the substrate activation promoting activity by the enzyme of 0.1 or more only in the enzyme-added group can be selected in this measurement system. The substrate activation promoting activity by the enzyme referred to here can be measured by the value of the absorbance 30 minutes after the addition of the synthetic substrate of the antigen-binding molecular solution.

 なお本明細書において引用された全ての先行技術文献は、参照として本明細書に組み入れられる。 All prior art documents cited herein are incorporated herein by reference.

 以下、本発明を実施例により具体的に説明するが、本発明はこれら実施例に制限されるものではない。 Hereinafter, the present invention will be specifically described with reference to Examples, but the present invention is not limited to these Examples.

〔実施例1〕抗F.XIa, F.X 二重特異性抗体の調製
 抗F.XIa抗体調製のために、以下の抗体の可変領域配列を用いた:WO2010080623A2に記載されている14E11(本明細書内配列番号:重鎖可変領域配列番号:1, 軽鎖可変領域配列番号:2);WO2013167669の配列番号165、166に記載されているF.XIa抗体(本明細書内でF11B1と記載:重鎖可変領域配列番号:11, 軽鎖可変領域配列番号12);WO2018145533の配列番号105、106に記載されているF.XIa抗体21F12(本明細書内でF11B2と記載:重鎖可変領域配列番号:13, 軽鎖可変領域配列番号14);およびWO2013167669の配列番号29、30に記載されているF.XIa抗体(本明細書内でF11B3と記載:重鎖可変領域配列番号:15, 軽鎖可変領域配列番号16)。抗F.X抗体調製のために、以下の抗体の可変領域配列を用いた:WO2005035756A1に記載されているSB04由来のF10B1(本明細書内配列番号:重鎖可変領域配列番号:3, 軽鎖可変領域配列番号:4);WO2019065795A1に記載されているJ327の重鎖可変領域配列とJNL095の軽鎖可変領域の配列に二つのアミノ酸置換を導入した配列からなるF10B2 (本明細書内配列番号:重鎖可変領域配列番号:5, 軽鎖可変領域配列番号:6);WO2018098363の配列番号427、615に記載されているF.X抗体BIIB-12-917 (本明細書内でF10B3と記載:重鎖可変領域配列番号:17, 軽鎖可変領域配列番号18);WO2019096874の配列番号4、5に記載されているF.X抗体mAb 00916 (本明細書内では軽鎖の1残基目Alaを除去し F10B4と記載:重鎖可変領域配列番号:19, 軽鎖可変領域配列番号20);およびWO2019096874の配列番号6、7に記載されているF.X抗体mAb 13F62 (本明細書内でF10B5と記載:重鎖可変領域配列番号:21, 軽鎖可変領域配列番号22)。いずれの抗体の可変領域配列も重鎖定常領域あるいは軽鎖定常領域配列と連結させ、抗体配列全長をコードする遺伝子を含む発現ベクターを構築した。発現ベクターをExpi293細胞 (Thermo Fisher Scientific)に、一過性に導入し抗体の発現を行った。得られた培養上清から、Protein A等を用いたアフィニティ精製により当業者公知の方法で精製した。さらに、これらの抗F.XIa抗体と抗F.X抗体からなる二重特異性抗体を当業者公知の方法で調製した。各抗体の可変領域と定常領域の配列番号の対応は表2の通りであり、それぞれの単一特異性抗体の名称と二重特異性抗体の名称も表2の通りに命名した。
[Example 1] Preparation of anti-F.XIa, FX bispecific antibody For the preparation of anti-F.XIa antibody, the following variable region sequences of the antibody were used: 14E11 described in WO2010080623A2 (the present specification). Internal SEQ ID NO:: Heavy Chain Variable Region SEQ ID NO: 1, Light Chain Variable Region SEQ ID NO: 2); F.XIa antibody set forth in WO2013167669 SEQ ID NOs: 165 and 166 (described herein as F11B1: heavy) Chain variable region SEQ ID NO: 11, light chain variable region SEQ ID NO: 12); F.XIa antibody 21F12 (described herein as F11B2) set forth in WO2018145533 SEQ ID NOs: 105 and 106: heavy chain variable region SEQ ID NO: : 13, light chain variable region SEQ ID NO: 14); and F.XIa antibody set forth in WO2013167669 SEQ ID NOs: 29, 30 (described herein as F11B3: heavy chain variable region SEQ ID NO: 15, light chain Variable region SEQ ID NO: 16). The following antibody variable region sequences were used for anti-FX antibody preparation: F10B1 from SB04 described in WO2005035756A1 (SEQ ID NO:: heavy chain variable region SEQ ID NO: 3, light chain variable region herein). SEQ ID NO: 4); F10B2 consisting of a sequence in which two amino acid substitutions are introduced into the heavy chain variable region sequence of J327 and the light chain variable region sequence of JNL095 described in WO2019065795A1 (SEQ ID NO:: heavy chain in the present specification). Variable region SEQ ID NO: 5, Light chain variable region SEQ ID NO: 6); FX antibody BIIB-12-917 (described herein as F10B3: heavy chain variable region) described in SEQ ID NOs: 427 and 615 of WO2018098363. SEQ ID NO: 17, light chain variable region SEQ ID NO: 18); FX antibody mAb 00916 described in SEQ ID NOs: 4 and 5 of WO2019096874 (in the present specification, the first residue Ala of the light chain is removed and described as F10B4. : Heavy chain variable region SEQ ID NO: 19, light chain variable region SEQ ID NO: 20); and FX antibody mAb 13F62 described in SEQ ID NOs: 6 and 7 of WO2019096874 (described herein as F10B5: heavy chain variable region). SEQ ID NO: 21, light chain variable region SEQ ID NO: 22). The variable region sequence of each antibody was linked to the heavy chain constant region or light chain constant region sequence to construct an expression vector containing a gene encoding the full length of the antibody sequence. The expression vector was transiently introduced into Expi293 cells (Thermo Fisher Scientific) to express the antibody. The obtained culture supernatant was purified by a method known to those skilled in the art by affinity purification using Protein A or the like. Furthermore, bispecific antibodies consisting of these anti-F.XIa antibodies and anti-FX antibodies were prepared by methods known to those skilled in the art. The correspondence between the sequence numbers of the variable region and the constant region of each antibody is shown in Table 2, and the names of the respective monospecific antibodies and bispecific antibodies are also named as shown in Table 2.

Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002

〔実施例2〕 in vitro酵素反応測定系によるF.XIa、F.X二重特異性抗体の、F.XIaによるF.X活性化促進活性の測定
 作製した抗F.XIa, F.X二重特異性抗体の、F.XIaによるF.X活性化促進活性の有無を、合成発色基質を用いたin vitro酵素反応測定系を用いて評価した。具体的には以下の手順で測定を行い、反応は全て室温で行った。48 ng/mLのF.XIa(Enzyme Research Laboratories)5 μLと各濃度の抗体溶液5 μLの混合液を384穴プレート中で30分間インキュベーションした。さらにその混合液に、22.9 μg/mLのF.X(Enzyme Research Laboratories)5 μLを添加し、酵素反応を開始させた。60分間反応させたのち、50μM Aprotinin(Sigma Aldrich)5 μLを加えることにより酵素反応を停止させた。続いて、F.Xaにより呈色する合成発色基質溶液5 μLをそれぞれのウェルに加え、30分後の波長405 nmにおける吸光度をSpectraMax340PC(Molecular Devices)により測定した。F.XIa、F.X二重特異性抗体の、F.XIaによるF.X活性化促進活性は、発色基質溶液添加30分間後の吸光度の値として表した[図3、図5]。なお、抗体の濃度は酵素反応中の溶液中濃度として示した。
評価の結果、F.XIa及びF.Xを含むin vitro試験系において、二重特異性抗体を添加した群では吸光度で示されるF.Xa量の抗体濃度依存的な上昇が確認された。一方、吸光度の上昇は二重特異性抗体の代わりに単一特異性抗体を添加した群においては確認されなかった。以上より、F.XIa, F.X二重特異性抗体はin vitro 酵素反応測定系においてF.XIa及びF.X両者への結合依存的にF.X活性化を促進することが示唆された。
 抗体溶液の溶媒には、0.1 %ウシ血清アルブミンを含むトリス緩衝生理食塩液(以下、TBSBと称す)を用いた。また、F.XIa及びF.Xの溶媒には1.5 mM CaCl2及び4.0 μMのリン脂質(Sysmex)を含むTBSB(以下、TBCPと称す)を用いた。発色基質溶液は、精製水を用いて調製した1.47 mg/mLの発色基質S-2222(Chromogenix)溶液を用いた。
[Example 2] Measurement of FX activation promoting activity by F.XIa of F.XIa and FX bispecific antibody by in vitro enzyme reaction measurement system The presence or absence of FX activation promoting activity by F.XIa was evaluated using an in vitro enzyme reaction measurement system using a synthetic color-developing substrate. Specifically, the measurement was carried out according to the following procedure, and all the reactions were carried out at room temperature. A mixture of 4 μL of 48 ng / mL F.XIa (Enzyme Research Laboratories) and 5 μL of antibody solution at each concentration was incubated in a 384-well plate for 30 minutes. Further, 5 μL of FX (Enzyme Research Laboratories) of 22.9 μg / mL was added to the mixed solution to initiate an enzymatic reaction. After reacting for 60 minutes, the enzyme reaction was stopped by adding 5 μL of 50 μM Aprotinin (Sigma Aldrich). Subsequently, 5 μL of a synthetic color-developing substrate solution colored by F.Xa was added to each well, and the absorbance at a wavelength of 405 nm after 30 minutes was measured by SpectraMax340PC (Molecular Devices). The FX activation promoting activity of F.XIa and FX bispecific antibodies by F.XIa was expressed as the absorbance value 30 minutes after the addition of the coloring substrate solution [Figs. 3 and 5]. The antibody concentration is shown as the concentration in the solution during the enzymatic reaction.
As a result of the evaluation, in the in vitro test system containing F.XIa and FX, an antibody concentration-dependent increase in the amount of F.Xa indicated by absorbance was confirmed in the group to which the bispecific antibody was added. On the other hand, no increase in absorbance was confirmed in the group to which the monospecific antibody was added instead of the bispecific antibody. From the above, it was suggested that F.XIa and FX bispecific antibodies promote FX activation in an in vitro enzyme reaction measurement system in a binding-dependent manner to both F.XIa and FX.
As the solvent of the antibody solution, Tris-buffered physiological saline containing 0.1% bovine serum albumin (hereinafter referred to as TBSB) was used. In addition, TBSB (hereinafter referred to as TBCP) containing 1.5 mM CaCl2 and 4.0 μM phospholipid (Sysmex) was used as the solvent for F.XIa and FX. As the color-developing substrate solution, a 1.47 mg / mL color-developing substrate S-2222 (Chromogenix) solution prepared using purified water was used.

〔実施例3〕 活性化部分トロンボプラスチン時間(APTT)による血漿凝固活性の測定
 血液凝固反応は複数のセリンプロテアーゼによる逐次的基質活性化反応である。F.X活性化促進活性を有する抗F.XIa,F.X二重特異性抗体が、血友病B血漿中においても同反応を促進し凝固能を是正するか明らかにするため、F.IX欠乏血漿を用いて臨床の凝固機能診断として広く用いられる指標である活性化部分トロンボプラスチン時間(APTT)に対する同抗体の影響を検討した。APTT測定は当業者公知の方法で実施し、具体的には以下の手順で実施した。TBSBを用いて各濃度に調製した抗体溶液、あるいは各濃度に調製したF.IX(クリスマシン、日本血液製剤機構)15 μL及びF.IX欠乏血漿(Sysmex)135 μLを混合し、抗体を含む血漿サンプルを調製した。血漿サンプル50 μL及びAPTT試薬(Sysmex)50 μLの混合液を37℃で190秒間加温した。凝固反応は0.02 M塩化カルシウム液(Sysmex)50 μLを同混合液に加えることにより開始させた。凝固時間は透過度の低下が最大低下時の50%となる時間を基準としてCS-2000i(Sysmex)を用いて測定し、APTTとして示した[図4、図6]。なお、抗体濃度は抗体を含む血漿サンプル中の濃度として示した。
 評価の結果、F.IX欠乏血漿において、F.IX添加群と同様に、二重特異性抗体添加群ではAPTT短縮効果を示した。以上の結果から抗F.XIa,F.X二重特異性抗体は、F.IX欠乏血漿において凝固促進作用を有することが示され、抗F.XIa,F.X二重特異性抗体は血友病Bの治療薬として臨床で用いられるF.IXと同様に血友病B患者血漿の凝固能を是正する可能性が示唆された。
[Example 3] Measurement of plasma coagulation activity by activated partial thromboplastin time (APTT) The blood coagulation reaction is a sequential substrate activation reaction by a plurality of serine proteases. In order to clarify whether anti-F.XIa, FX bispecific antibody having FX activation promoting activity promotes the same reaction in hemophilia B plasma and corrects coagulation ability, F.IX deficient plasma is used. The effect of the antibody on activated partial thromboplastin time (APTT), which is a widely used index for clinical diagnosis of coagulation function, was investigated. APTT measurement was carried out by a method known to those skilled in the art, and specifically, the following procedure was carried out. An antibody solution prepared at each concentration using TBSB, or 15 μL of F.IX (Chris Machine, Japan Blood Products Organization) prepared at each concentration and 135 μL of F.IX-deficient plasma (Sysmex) are mixed and contains an antibody. Plasma samples were prepared. A mixture of 50 μL of plasma sample and 50 μL of APTT reagent (Sysmex) was heated at 37 ° C. for 190 seconds. The coagulation reaction was initiated by adding 50 μL of 0.02 M calcium chloride solution (Sysmex) to the mixture. The coagulation time was measured using CS-2000i (Sysmex) based on the time when the decrease in permeability was 50% of the maximum decrease, and was shown as APTT [Figs. 4 and 6]. The antibody concentration is shown as the concentration in the plasma sample containing the antibody.
As a result of the evaluation, in F.IX-deficient plasma, the APTT shortening effect was shown in the bispecific antibody-added group as in the F.IX-added group. From the above results, it was shown that the anti-F.XIa, FX bispecific antibody has a coagulation-promoting effect in F.IX-deficient plasma, and the anti-F.XIa, FX bispecific antibody is associated with hemophilia B. It was suggested that it may correct the coagulation ability of plasma of hemophilia B patients as well as F.IX used clinically as a therapeutic drug.

他の血液凝固因子への応用可能性
 酵素の基質特異性はトロンビンやF.XIaに代表されるようにエキソサイトによる標的基質への結合によって与えられるものも多い(Mol Aspects Med. 2008 Aug; 29(4): 203-254., Thromb Res. 2018 Jan; 161: 94-105., J Thromb Haemost. 2005;3(1):54-67.)。したがって、上述のF.XIa、F.X二重特異性抗体による酵素反応促進ならびに血液凝固能に対する結果に基づき、酵素Aによる基質Bの反応を促進するような高分子ポリペプチドである補因子が生体に存在しないような組み合わせの酵素と基質でも、二重特異性抗原結合分子によって両者を強制的に近接させることによって反応を促進できると考えられる。
Applicability to other blood coagulation factors Substrate specificity of enzymes is often conferred by binding to target substrates by exosites, as represented by thrombin and F.XIa (Mol Aspects Med. 2008 Aug; 29). (4): 203-254., Thromb Res. 2018 Jan; 161: 94-105., J Thromb Haemost. 2005; 3 (1): 54-67.). Therefore, based on the results of the above-mentioned F.XIa and FX bispecific antibodies for promoting the enzyme reaction and blood coagulation ability, a cofactor, which is a high molecular weight polypeptide that promotes the reaction of the substrate B with the enzyme A, is present in the living body. It is considered that even a combination of enzyme and substrate that does not exist can promote the reaction by forcibly bringing them close to each other by a bispecific antigen-binding molecule.

血液凝固系における他の凝固因子に対する適用
 血液凝固反応は,血漿中のセリンプロテアーゼ凝固因子前駆体の逐次的活性化により生成したトロンビンがフィブリノーゲンをフィブリンに変換することにより完結する。主要な血液凝固カスケードの模式図を図2に示した(Blood. 2010;115(13):2569-2577.を参考にして作製)。F.XII(a)、F.XI(a)、F.IX(a)、F.X(a)、F.VII(a)、F.IIaはセリンプロテアーゼであり、図2に記載されるようにF.XIIaはF.XIを、F.XIaはF.IXを、F.IXaはF.Xを、F.VIIaはF.Xを、F.Xaはプロトロンビンをそれぞれ活性化して凝固反応を進める。
Application to other coagulation factors in the blood coagulation system The blood coagulation reaction is completed by the conversion of fibrinogen to fibrin by thrombin produced by the sequential activation of the serine protease coagulation factor precursor in plasma. A schematic diagram of the major blood coagulation cascades is shown in Figure 2 (created with reference to Blood. 2010; 115 (13): 2569-2577.). F.XII (a), F.XI (a), F.IX (a), FX (a), F.VII (a), F.IIa are serine proteases, as shown in FIG. F.XIIa activates F.XI, F.XIa activates F.IX, F.IXa activates FX, F.VIIa activates FX, and F.Xa activates prothrombin to proceed with the coagulation reaction.

 血友病は図2におけるF.VIIIa, F.IX, F.XIが先天的、あるいは後天的に活性低下することによって引き起こされる出血性疾患である。一方で疾患状態を引き起こしている血液凝固因子以外の血液凝固因子は正常な機能を有している。したがって、これらの正常な活性を有する血液凝固因子に本コンセプトを適用することで、血友病状態の血漿で内因性凝固カスケードを正常化させる、あるいは外因系カスケードの反応を促進して出血傾向を抑制することができる。
 具体的には、表1に記載したa~hの酵素と基質の組合せに結合する二重特異性抗原結合分子を作製することで血友病における出血傾向を抑制することができる。
Hemophilia is a hemorrhagic disease caused by a congenital or acquired decrease in activity of F.VIIIa, F.IX, and F.XI in Fig. 2. On the other hand, blood coagulation factors other than the blood coagulation factor causing the disease state have a normal function. Therefore, by applying this concept to these normally active blood coagulation factors, the endogenous coagulation cascade can be normalized in hemophiliac plasma, or the reaction of the extrinsic cascade can be promoted to promote bleeding tendency. It can be suppressed.
Specifically, the bleeding tendency in hemophilia can be suppressed by preparing a bispecific antigen-binding molecule that binds to the combination of the enzymes a to h shown in Table 1 and the substrate.

 本開示の二重特異性抗原結合分子は、血液凝固の促進に寄与し、ひいては出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有用であり得る。 The bispecific antigen-binding molecule of the present disclosure contributes to the promotion of blood coagulation and may be useful for the prevention and / or treatment of bleeding, diseases associated with bleeding, or diseases caused by bleeding.

Claims (29)

 以下の(a)~(h)のいずれかに記載の二重特異性抗原結合分子:
(a)活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(b)活性化血液凝固第X因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(c)活性化血液凝固第VII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(d)活性化血液凝固第VII因子-Tissue Factor complexと血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(e)活性化血液凝固第XII因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子;
(f)Thrombinと血液凝固第X因子の両方を認識する二重特異性抗原結合分子; 
(g)活性化血液凝固第XII因子と血液凝固第IX因子の両方を認識する二重特異性抗原結合分子;または
(h)活性化血液凝固第XI因子と血液凝固第XI因子の両方を認識する二重特異性抗原結合分子。
The bispecific antigen-binding molecule according to any one of (a) to (h) below:
(a) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X;
(b) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor X and blood coagulation factor X;
(c) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor VII and blood coagulation factor X;
(d) Activated blood coagulation factor VII-A bispecific antigen-binding molecule that recognizes both Tissue Factor complex and blood coagulation factor X;
(e) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor X;
(f) A bispecific antigen-binding molecule that recognizes both Thrombin and blood coagulation factor X;
(g) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XII and blood coagulation factor IX; or
(h) A bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor XI.
 請求項1に記載の二重特異性抗原結合分子であって、該酵素による該基質の活性化を促進する二重特異性抗原結合分子。 The bispecific antigen-binding molecule according to claim 1, which promotes activation of the substrate by the enzyme.  二重特異性抗体である、請求項1または2に記載の二重特異性抗原結合分子。 The bispecific antigen-binding molecule according to claim 1 or 2, which is a bispecific antibody.  請求項1~3のいずれかに記載の抗原結合分子および薬学的に許容される担体を含む、組成物。 A composition comprising the antigen-binding molecule according to any one of claims 1 to 3 and a pharmaceutically acceptable carrier.  出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に用いられる医薬組成物である、請求項4に記載の組成物。 The composition according to claim 4, which is a pharmaceutical composition used for prevention and / or treatment of bleeding, a disease associated with bleeding, or a disease caused by bleeding.  出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患である、請求項5に記載の組成物。 Claim 5 that bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activated blood coagulation factor IX. The composition according to.  血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Bである、請求項6に記載の組成物。 The composition according to claim 6, wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor IX and / or activation blood coagulation factor IX is hemophilia B.  血液凝固第IX因子および/または活性化血液凝固第IX因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第IX因子および/または活性化血液凝固第IX因子に対するインヒビターが出現している疾患である、請求項6に記載の組成物。 Blood coagulation factor IX and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor IX, and inhibitors against blood coagulation factor IX and / or activated blood coagulation factor IX appear The composition according to claim 6, which is a disease.  出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患である、請求項5に記載の組成物。 Claim 5 that bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor VIII and / or activated blood coagulation factor VIII. The composition according to.  血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病A、後天性血友病、またはフォンビルブランド病である、請求項9に記載の組成物。 Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII are hemophilia A, acquired hemophilia, or von Willebrand's disease. The composition according to claim 9.  血液凝固第VIII因子および/または活性化血液凝固第VIII因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第VIII因子および/または活性化血液凝固第VIII因子に対するインヒビターが出現している疾患である、請求項9に記載の組成物。 Blood coagulation factor VIII and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor VIII appear as inhibitors of blood coagulation factor VIII and / or activated blood coagulation factor VIII The composition according to claim 9, which is a disease.  出血、出血を伴う疾患、もしくは出血に起因する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患である、請求項5に記載の組成物。 Claim 5 that bleeding, a disease associated with bleeding, or a disease caused by bleeding is a disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activated blood coagulation factor XI. The composition according to.  血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血友病Cである、請求項12に記載の組成物。 The composition according to claim 12, wherein the disease that develops and / or progresses due to a decrease or deficiency in the activity of blood coagulation factor XI and / or activation of blood coagulation factor XI is hemophilia C.  血液凝固第XI因子および/または活性化血液凝固第XI因子の活性の低下ないし欠損によって発症および/または進展する疾患が、血液凝固第XI因子および/または活性化血液凝固第XI因子に対するインヒビターが出現している疾患である、請求項12に記載の組成物。 Blood coagulation factor XI and / or activation Diseases that develop and / or progress due to decreased or deficient activity of blood coagulation factor XI appear as inhibitors of blood coagulation factor XI and / or activated blood coagulation factor XI The composition according to claim 12, which is a disease.  請求項1~3のいずれかに記載の二重特異性抗原結合分子の、請求項3~13のいずれかに記載した組成物の製造のための使用。 Use of the bispecific antigen-binding molecule according to any one of claims 1 to 3 for producing the composition according to any one of claims 3 to 13.  少なくとも請求項1~3のいずれかに記載の二重特異性抗原結合分子、または請求項3に記載の組成物を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を予防および/または治療する方法に用いるためのキット。 Preventing and / or preventing bleeding, bleeding-related diseases, or diseases caused by bleeding, which comprises at least the bispecific antigen-binding molecule according to any one of claims 1 to 3 or the composition according to claim 3. A kit for use in therapeutic methods.  少なくとも請求項1~3のいずれかに記載の二重特異性抗原結合分子、または請求項3に記載の組成物を含み、かつ血液凝固第IX因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第IX因子と併用して、予防および/または治療する方法に用いるためのキット。 Bleeding, a disease associated with bleeding, or bleeding, which comprises at least the bispecific antigen-binding molecule according to any one of claims 1 to 3 or the composition according to claim 3 and contains blood coagulation factor IX. A kit for use in methods of preventing and / or treating diseases caused by blood coagulation factor IX in combination.  少なくとも請求項1~3のいずれかに記載の二重特異性抗原結合分子、または請求項3に記載の組成物を含み、かつ血液凝固第VIII因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第VIII因子と併用して、予防および/または治療する方法に用いるためのキット。 Bleeding, a disease associated with bleeding, or bleeding, which comprises at least the bispecific antigen-binding molecule according to any one of claims 1 to 3 or the composition according to claim 3 and contains blood coagulation factor VIII. A kit for use in methods of preventing and / or treating diseases caused by blood coagulation factor VIII in combination.  少なくとも請求項1~3のいずれかに記載の二重特異性抗原結合分子、または請求項3に記載の組成物を含み、かつ血液凝固第XI因子を含む、出血、出血を伴う疾患、もしくは出血に起因する疾患を、血液凝固第XI因子と併用して、予防および/または治療する方法に用いるためのキット。 Bleeding, bleeding-related disease, or bleeding containing at least the bispecific antigen-binding molecule according to any one of claims 1 to 3 or the composition according to claim 3 and containing blood coagulation factor XI. A kit for use in methods of preventing and / or treating diseases caused by blood coagulation factor XI in combination.  少なくとも請求項1~3のいずれかに記載の二重特異性抗原結合分子、または請求項3に記載の組成物を含み、かつ出血、出血を伴う疾患、もしくは出血に起因する疾患を、バイパス製剤と併用して、予防および/または治療する方法に用いるためのキット。 A bypass preparation for bleeding, a disease associated with bleeding, or a disease caused by bleeding, which contains at least the bispecific antigen-binding molecule according to any one of claims 1 to 3 or the composition according to claim 3. A kit for use in prophylactic and / or therapeutic methods in combination with.  活性化血液凝固第XI因子と血液凝固第X因子の両方を認識する二重特異性抗原結合分子を用いて血液凝固を促進する方法。 A method of promoting blood coagulation using a bispecific antigen-binding molecule that recognizes both activated blood coagulation factor XI and blood coagulation factor X.  出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質のスクリーニング方法であって、
(1)試験物質と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質と血液凝固第X因子との結合を評価する工程
を含む、スクリーニング方法。
A screening method for substances that are effective in the prevention and / or treatment of bleeding, bleeding-related diseases, or diseases caused by bleeding.
A screening method comprising (1) evaluating the binding between the test substance and activated blood coagulation factor XI, and (2) evaluating the binding between the test substance and blood coagulation factor X.
 さらに以下の工程を含む、請求項22に記載のスクリーニング方法:
(3)試験物質を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
The screening method according to claim 22, further comprising the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance.
 出血、出血を伴う疾患、もしくは出血に起因する疾患の予防および/または治療に有効な物質もしくは組成物の品質試験方法であって、
(1)試験物質もしくは試験組成物と活性化血液凝固第XI因子との結合を評価する工程、および
(2)試験物質もしくは試験組成物と血液凝固第X因子との結合を評価する工程
を含む、品質試験方法。
A quality test method for a substance or composition that is effective in the prevention and / or treatment of bleeding, bleeding-related diseases, or diseases caused by bleeding.
Includes (1) a step of evaluating the binding of the test substance or test composition to activated blood coagulation factor XI, and (2) a step of evaluating the binding of the test substance or test composition to blood coagulation factor X. , Quality test method.
 さらに以下の工程を含む、請求項24に記載の品質試験方法:
(3)試験物質もしくは試験組成物を用いて活性化血液凝固第XI因子による血液凝固第X因子の活性化反応を評価する工程。
The quality test method according to claim 24, further comprising the following steps:
(3) A step of evaluating the activation reaction of blood coagulation factor X by activated blood coagulation factor XI using a test substance or a test composition.
請求項1~3のいずれかに記載の二重特異性抗原結合分子をコードする核酸。 A nucleic acid encoding the bispecific antigen-binding molecule according to any one of claims 1 to 3. 請求項26に記載の核酸が挿入されたベクター。 A vector into which the nucleic acid according to claim 26 is inserted. 請求項26に記載の核酸または請求項27に記載のベクターを含む細胞。 A cell comprising the nucleic acid according to claim 26 or the vector according to claim 27. 請求項28に記載の細胞を培養することにより、請求項1~3のいずれかに記載の二重特異性抗原結合分子を製造する方法。 The method for producing a bispecific antigen-binding molecule according to any one of claims 1 to 3 by culturing the cell according to claim 28.
PCT/JP2021/015683 2020-04-17 2021-04-16 Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition Ceased WO2021210667A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180041295.5A CN115916836A (en) 2020-04-17 2021-04-16 Bispecific antigen binding molecules and compositions related thereto, uses, kits and methods for making compositions

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2020-074338 2020-04-17
JP2020074338A JP2023106635A (en) 2020-04-17 2020-04-17 Bispecific antigen binding molecules and compositions related thereto, uses, kits and methods for producing compositions

Publications (1)

Publication Number Publication Date
WO2021210667A1 true WO2021210667A1 (en) 2021-10-21

Family

ID=78083783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/015683 Ceased WO2021210667A1 (en) 2020-04-17 2021-04-16 Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition

Country Status (4)

Country Link
JP (1) JP2023106635A (en)
CN (1) CN115916836A (en)
TW (1) TW202204421A (en)
WO (1) WO2021210667A1 (en)

Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0058481A1 (en) 1981-02-16 1982-08-25 Zeneca Limited Continuous release pharmaceutical compositions
EP0133988A2 (en) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Regulating peptide-containing pharmaceutical preparations with retarded release, and process for their preparation
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US4945050A (en) 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1993017706A1 (en) 1992-03-11 1993-09-16 Agracetus, Inc. Genetic vaccine for immunodeficiency viruses
WO1994002602A1 (en) 1992-07-24 1994-02-03 Cell Genesys, Inc. Generation of xenogeneic antibodies
WO1994025585A1 (en) 1993-04-26 1994-11-10 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1996002576A1 (en) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin-8
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1999051743A1 (en) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
WO2004065611A1 (en) 2003-01-21 2004-08-05 Chugai Seiyaku Kabushiki Kaisha Method of screening light chain of antibdoy
WO2005035756A1 (en) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
WO2006109592A1 (en) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha Antibody substituting for function of blood coagulation factor viii
WO2008045140A1 (en) 2006-05-19 2008-04-17 Alder Biopharmaceuticals, Inc. Culture method for obtaining a clonal population of antigen-specific b cells
JP2009082141A (en) * 2001-06-28 2009-04-23 Domantis Ltd Dual-specific ligand and use thereof
WO2009113742A1 (en) 2008-03-14 2009-09-17 東ソー株式会社 Method of producing genetically engineered antibody
WO2010080623A2 (en) 2008-12-18 2010-07-15 Oregon Health & Science University Anti-fxi antibodies and methods of use
WO2012067176A1 (en) 2010-11-17 2012-05-24 中外製薬株式会社 Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
WO2013120629A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
WO2013167669A1 (en) 2012-05-10 2013-11-14 Bayer Pharma Aktiengesellschaft Antibodies capable of binding to the coagulation factor xi and/or its activated form factor xia and uses thereof
EP2101823B1 (en) 2007-01-09 2016-11-23 CureVac AG Rna-coded antibody
WO2018098363A2 (en) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Bispecific antibodies binding to coagulation factor ix and coagulation factor x
WO2018145533A1 (en) 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anti-coagulation factor xi antibody
WO2019065795A1 (en) 2017-09-29 2019-04-04 中外製薬株式会社 Multispecific antigen-binding molecule having blood coagulation factor viii (fviii) cofactor function-substituting activity, and pharmaceutical formulation containing said molecule as active ingredient
WO2019096874A1 (en) 2017-11-15 2019-05-23 Novo Nordisk A/S Factor x binders enhancing fx activation
WO2019235420A1 (en) * 2018-06-04 2019-12-12 中外製薬株式会社 Method for detecting complex

Patent Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
EP0058481A1 (en) 1981-02-16 1982-08-25 Zeneca Limited Continuous release pharmaceutical compositions
EP0133988A2 (en) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Regulating peptide-containing pharmaceutical preparations with retarded release, and process for their preparation
US4945050A (en) 1984-11-13 1990-07-31 Cornell Research Foundation, Inc. Method for transporting substances into living cells and tissues and apparatus therefor
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1993017706A1 (en) 1992-03-11 1993-09-16 Agracetus, Inc. Genetic vaccine for immunodeficiency viruses
WO1994002602A1 (en) 1992-07-24 1994-02-03 Cell Genesys, Inc. Generation of xenogeneic antibodies
WO1994025585A1 (en) 1993-04-26 1994-11-10 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1996002576A1 (en) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin-8
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1999051743A1 (en) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
JP2009082141A (en) * 2001-06-28 2009-04-23 Domantis Ltd Dual-specific ligand and use thereof
WO2004065611A1 (en) 2003-01-21 2004-08-05 Chugai Seiyaku Kabushiki Kaisha Method of screening light chain of antibdoy
WO2005035756A1 (en) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Bispecific antibody substituting for functional proteins
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
WO2006109592A1 (en) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha Antibody substituting for function of blood coagulation factor viii
WO2008045140A1 (en) 2006-05-19 2008-04-17 Alder Biopharmaceuticals, Inc. Culture method for obtaining a clonal population of antigen-specific b cells
EP2101823B1 (en) 2007-01-09 2016-11-23 CureVac AG Rna-coded antibody
WO2009113742A1 (en) 2008-03-14 2009-09-17 東ソー株式会社 Method of producing genetically engineered antibody
WO2010080623A2 (en) 2008-12-18 2010-07-15 Oregon Health & Science University Anti-fxi antibodies and methods of use
WO2012067176A1 (en) 2010-11-17 2012-05-24 中外製薬株式会社 Multi-specific antigen-binding molecule having alternative function to function of blood coagulation factor viii
WO2013120629A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
WO2013167669A1 (en) 2012-05-10 2013-11-14 Bayer Pharma Aktiengesellschaft Antibodies capable of binding to the coagulation factor xi and/or its activated form factor xia and uses thereof
WO2018098363A2 (en) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Bispecific antibodies binding to coagulation factor ix and coagulation factor x
WO2018145533A1 (en) 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anti-coagulation factor xi antibody
WO2019065795A1 (en) 2017-09-29 2019-04-04 中外製薬株式会社 Multispecific antigen-binding molecule having blood coagulation factor viii (fviii) cofactor function-substituting activity, and pharmaceutical formulation containing said molecule as active ingredient
WO2019096874A1 (en) 2017-11-15 2019-05-23 Novo Nordisk A/S Factor x binders enhancing fx activation
WO2019235420A1 (en) * 2018-06-04 2019-12-12 中外製薬株式会社 Method for detecting complex

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
"Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
"Remington's Pharmaceutical Science", 1980
"Sequences of proteins of immunological interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES
BLOOD, vol. 115, no. 13, 2010, pages 2569 - 2577
BLOOD, vol. 127, no. 13, 2016, pages 1633 - 1641
BORREBAECK CAKLARRICK JW: "THERAPEUTIC MONOCLONAL ANTIBODIES", 1990, MACMILLAN PUBLISHERS LTD
BRENNAN M ET AL., SCIENCE, vol. 229, 1985, pages 81 - 83
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLARKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
HAEMOPHILIA, vol. 21, no. 4, 2015, pages 477 - 480
HOLLIGER P ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOLLIGER P ET AL., PROC. OF THE NATIONAL ACADEMY OF SCIENCES OF THE USA, vol. 90, 1993, pages 6444 - 6448
J THROMB HAEMOST, vol. 11, no. 12, 2013, pages 2118 - 2127
J THROMB HAEMOST, vol. 16, no. 10, 2018, pages 2044 - 2049
J THROMB HAEMOST, vol. 3, no. 1, 2005, pages 54 - 67
KELER T ET AL., CANCER RESEARCH, vol. 57, 1997, pages 4008 - 4014
KINDT ET AL.: "Kuby Immunology", 2007, W.H. FREEMAN AND CO., pages: 91
KIPRIYANOV SM ET AL., J. OF MOLECULAR BIOLOGY, vol. 293, 1999, pages 41 - 56
KOSTELNY SA ET AL., J. OF IMMUNOLOGY, vol. 148, 1992, pages 1547 - 53
LANGER ET AL., J.BIOMED.MATER.RES., vol. 15, 1981, pages 267 - 277
LANGER, CHEMTECH, vol. 12, 1982, pages 98 - 105
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745
MALLENDER WD ET AL., J. OF BIOLOGICAL CHEMISTRY, vol. 269, 1994, pages 199 - 206
MATAFONOV, A. ET AL.: "Evidence for factor IX-independent roles for factor XIa in blood coagulation", J. THROMB. HAEMOST., vol. 11, 2013, pages 2118 - 2127, XP055293618 *
MERCHANT, AM ET AL.: "An efficient route to human bispecific IgG", NATURE BIOTECHNOLOGY, vol. 16, 1998, pages 677 - 681, XP002141015, DOI: 10.1038/nbt0798-677
MILSTEIN C ET AL., NATURE, vol. 305, 1983, pages 537 - 540
MOL ASPECTS MED, vol. 29, no. 4, August 2008 (2008-08-01), pages 203 - 254
NAT MED, vol. 18, no. 10, October 2012 (2012-10-01), pages 1570 - 4
NEW ENG J MED, vol. 21, 2016, pages 2044 - 2053
PORTOLANO ET AL., J. IMMUNOL., vol. 150, 1993, pages 880 - 887
PROC NATL ACAD SCI USA., vol. 93, no. 15, 1996, pages 7843 - 7848
RIDGWAY, JB ET AL., PROTEIN ENGINEERING, vol. 9, 1996, pages 617 - 621
SATO K ET AL., CANCER RESEARCH, vol. 53, 1993, pages 851 - 856
SEMIN THROMB HEMOST, vol. 44, no. 6, 2018, pages 578 - 589
SIDMAN ET AL., BIOPOLYMERS, vol. 22, 1983, pages 547 - 556
THROMB. RES., vol. 161, January 2018 (2018-01-01), pages 94 - 105
ZHU Z ET AL., PROTEIN SCIENCE, vol. 6, 1997, pages 781 - 788

Also Published As

Publication number Publication date
CN115916836A (en) 2023-04-04
TW202204421A (en) 2022-02-01
JP2023106635A (en) 2023-08-02

Similar Documents

Publication Publication Date Title
US20250270345A1 (en) Antibody substituting for function of blood coagulation factor viii
JP6823677B2 (en) A multispecific antigen-binding molecule that has the function of substituting the function of blood coagulation factor VIII.
US20240052059A1 (en) Medicinal composition usable for preventing and/or treating blood coagulation factor ix abnormality, comprising multispecific antigen binding molecule replacing function of blood coagulation factor viii
RU2339696C2 (en) Bispecific antibody replacing functional proteins
ES2677329T3 (en) Monoclonal antibodies against tissue factor pathway inhibitor (TFPI)
ES2905085T3 (en) Antibodies that can bind to two epitopes on the tissue factor pathway inhibitor (1-161)
ES2765418T3 (en) Monoclonal Antibodies Optimized Against Tissue Factor Pathway Inhibitor (TFPI)
TWI772724B (en) Fixaxfx bispecific antibody with common light chain
US20160297892A1 (en) Novel Methods and Antibodies for Treating Coagulapathy
WO2018234575A1 (en) BISPECIFIC ANTIBODIES OF FACTOR IX AND FACTOR X
CA3018124A1 (en) Monoclonal antibodies against the active site of factor xi and uses thereof
TW202007696A (en) Improved procoagulant antibodies
KR20210153639A (en) bispecific antibody
WO2021210667A1 (en) Bispecific antigen-binding molecule, composition associated therewith, and use, kit, and method for producing composition
WO2008026720A1 (en) Antibody capable of enhancing activation of blood coagulation factor viii
JP6445440B2 (en) Treatment of bleeding disorders with anti-protein C antibodies
TW202039583A (en) Proteinaceous molecules binding factor ixa and factor x
RU2818588C2 (en) Bispecific antibodies
WO2020114615A1 (en) Bispecific antibodies binding factor ixa and factor x
ES2865152T3 (en) Bispecific antibody that replaces functional proteins
WO2020115283A1 (en) Bispecific antibodies binding factor ixa and factor x

Legal Events

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

Ref document number: 21788259

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021788259

Country of ref document: EP

Effective date: 20221117

122 Ep: pct application non-entry in european phase

Ref document number: 21788259

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP