WO2021210667A1 - Molécule de liaison à un antigène bispécifique, composition associée à celle-ci et utilisation, kit et procédé de production de composition - Google Patents

Molécule de liaison à un antigène bispécifique, composition associée à celle-ci et utilisation, kit et procédé de production de 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
Application number
PCT/JP2021/015683
Other languages
English (en)
Japanese (ja)
Inventor
光 古賀
Original Assignee
中外製薬株式会社
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 中外製薬株式会社 filed Critical 中外製薬株式会社
Priority to CN202180041295.5A priority Critical patent/CN115916836A/zh
Publication of WO2021210667A1 publication Critical patent/WO2021210667A1/fr

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

Abstract

La présente invention concerne une molécule de liaison à un antigène bispécifique qui reconnaît deux facteurs spécifiques à la fois parmi divers facteurs liés à la coagulation, ainsi qu'une composition et un kit contenant la molécule de liaison à un antigène bispécifique. Dans un mode de réalisation non limitatif, la molécule de liaison à un antigène bispécifique, la composition et le kit selon la présente invention sont utilisés pour prévenir et/ou traiter une hémorragie, les maladies s'accompagnant d'une hémorragie ou les maladies provoquées par une hémorragie. De plus, la présente invention concerne une méthode d'utilisation de la molécule de liaison à un antigène bispécifique pour favoriser la coagulation. En outre, la présente invention concerne un procédé de criblage de substances qui sont efficaces pour prévenir et/ou traiter une hémorragie, les maladies s'accompagnant d'une hémorragie ou les maladies provoquées par une hémorragie.
PCT/JP2021/015683 2020-04-17 2021-04-16 Molécule de liaison à un antigène bispécifique, composition associée à celle-ci et utilisation, kit et procédé de production de composition WO2021210667A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180041295.5A CN115916836A (zh) 2020-04-17 2021-04-16 双特异性抗原结合分子及其相关组合物、用于制备组合物的应用、试剂盒和方法

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2020-074338 2020-04-17
JP2020074338A JP2023106635A (ja) 2020-04-17 2020-04-17 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

Publications (1)

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

Family

ID=78083783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/015683 WO2021210667A1 (fr) 2020-04-17 2021-04-16 Molécule de liaison à un antigène bispécifique, composition associée à celle-ci et utilisation, kit et procédé de production de composition

Country Status (4)

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

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 (fr) 1981-02-16 1982-08-25 Zeneca Limited Compositions pharmaceutiques pour la libération continue de la substance active
EP0133988A2 (fr) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Préparations pharmaceutiques contenant des peptides régulateurs à libération retardée et procédé pour leur préparation
EP0239400A2 (fr) 1986-03-27 1987-09-30 Medical Research Council Anticorps recombinants et leurs procédés de 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 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1992003918A1 (fr) 1990-08-29 1992-03-19 Genpharm International, Inc. Animaux non humains transgeniques capables de produire des anticorps heterologues
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1993012227A1 (fr) 1991-12-17 1993-06-24 Genpharm International, Inc. Animaux transgeniques non humains capables de produire des anticorps heterologues
WO1993017706A1 (fr) 1992-03-11 1993-09-16 Agracetus, Inc. Vaccin genetique pour virus d'immunodefience
WO1994002602A1 (fr) 1992-07-24 1994-02-03 Cell Genesys, Inc. Production d'anticorps xenogeniques
WO1994025585A1 (fr) 1993-04-26 1994-11-10 Genpharm International, Inc. Animaux transgeniques capables de produire des anticorps heterologues
WO1996002576A1 (fr) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Anticorps humain reconstitue contre l'interleukine-8 humaine
WO1996034096A1 (fr) 1995-04-28 1996-10-31 Abgenix, Inc. Anticorps humains derives de xeno-souris immunisees
WO1996033735A1 (fr) 1995-04-27 1996-10-31 Abgenix, Inc. Anticorps humains derives d'une xenosouris immunisee
WO1999051743A1 (fr) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Anticorps humanise contre le facteur tissulaire humain (tf) et procede de production d'anticorps humanises
WO2004065611A1 (fr) 2003-01-21 2004-08-05 Chugai Seiyaku Kabushiki Kaisha Procede de selection par criblage d'une chaine legere d'anticorps
WO2005035754A1 (fr) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Anticorps a double specificite de substitution de proteine fonctionnelle
WO2005035756A1 (fr) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Anticorps bispecifique substituant des proteines fonctionnelles
WO2006109592A1 (fr) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha Substitution des anticorps de la fonction du facteur viii de coagulation sanguine
WO2008045140A1 (fr) 2006-05-19 2008-04-17 Alder Biopharmaceuticals, Inc. Procédé de culture permettant d'obtenir une population clonée de lymphocytes b spécifiques d'antigène
JP2009082141A (ja) * 2001-06-28 2009-04-23 Domantis Ltd 二重特異性リガンドとその利用
WO2009113742A1 (fr) 2008-03-14 2009-09-17 東ソー株式会社 Procédé de fabrication d'un anticorps produit par génie génétique
WO2010080623A2 (fr) 2008-12-18 2010-07-15 Oregon Health & Science University Anticorps anti-fxi et procédés d'utilisation
WO2012067176A1 (fr) 2010-11-17 2012-05-24 中外製薬株式会社 Molécule de liaison à un antigène multi-spécifique ayant une fonction alternative par rapport à la fonction du facteur viii de coagulation sanguine
WO2013120629A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine thérapeutique codée
WO2013167669A1 (fr) 2012-05-10 2013-11-14 Bayer Pharma Aktiengesellschaft Anticorps capables de lier au facteur de coagulation xi et/ou à sa forme activée, le facteur xia, et utilisations de ceux-ci
EP2101823B1 (fr) 2007-01-09 2016-11-23 CureVac AG Anticorps code par un arn
WO2018098363A2 (fr) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Anticorps bispécifiques se liant au facteur ix de coagulation et au facteur x de coagulation
WO2018145533A1 (fr) 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anticorps anti-facteur xi de coagulation
WO2019065795A1 (fr) 2017-09-29 2019-04-04 中外製薬株式会社 Molécule multispécifique de liaison à l'antigène ayant une activité de substitution de la fonction de cofacteur du facteur viii de coagulation sanguine (fviii), et formulation pharmaceutique contenant ladite molécule en tant que principe actif
WO2019096874A1 (fr) 2017-11-15 2019-05-23 Novo Nordisk A/S Liants de facteur x renforçant l'activation fx
WO2019235420A1 (fr) * 2018-06-04 2019-12-12 中外製薬株式会社 Procédé permettant de détecter un complexe

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 (fr) 1981-02-16 1982-08-25 Zeneca Limited Compositions pharmaceutiques pour la libération continue de la substance active
EP0133988A2 (fr) 1983-08-02 1985-03-13 Hoechst Aktiengesellschaft Préparations pharmaceutiques contenant des peptides régulateurs à libération retardée et procédé pour leur préparation
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 (fr) 1986-03-27 1987-09-30 Medical Research Council Anticorps recombinants et leurs procédés de production
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1992003918A1 (fr) 1990-08-29 1992-03-19 Genpharm International, Inc. Animaux non humains transgeniques capables de produire des anticorps heterologues
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
WO1993012227A1 (fr) 1991-12-17 1993-06-24 Genpharm International, Inc. Animaux transgeniques non humains capables de produire des anticorps heterologues
WO1993017706A1 (fr) 1992-03-11 1993-09-16 Agracetus, Inc. Vaccin genetique pour virus d'immunodefience
WO1994002602A1 (fr) 1992-07-24 1994-02-03 Cell Genesys, Inc. Production d'anticorps xenogeniques
WO1994025585A1 (fr) 1993-04-26 1994-11-10 Genpharm International, Inc. Animaux transgeniques capables de produire des anticorps heterologues
WO1996002576A1 (fr) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Anticorps humain reconstitue contre l'interleukine-8 humaine
WO1996033735A1 (fr) 1995-04-27 1996-10-31 Abgenix, Inc. Anticorps humains derives d'une xenosouris immunisee
WO1996034096A1 (fr) 1995-04-28 1996-10-31 Abgenix, Inc. Anticorps humains derives de xeno-souris immunisees
WO1999051743A1 (fr) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Anticorps humanise contre le facteur tissulaire humain (tf) et procede de production d'anticorps humanises
JP2009082141A (ja) * 2001-06-28 2009-04-23 Domantis Ltd 二重特異性リガンドとその利用
WO2004065611A1 (fr) 2003-01-21 2004-08-05 Chugai Seiyaku Kabushiki Kaisha Procede de selection par criblage d'une chaine legere d'anticorps
WO2005035756A1 (fr) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Anticorps bispecifique substituant des proteines fonctionnelles
WO2005035754A1 (fr) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Anticorps a double specificite de substitution de proteine fonctionnelle
WO2006109592A1 (fr) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha Substitution des anticorps de la fonction du facteur viii de coagulation sanguine
WO2008045140A1 (fr) 2006-05-19 2008-04-17 Alder Biopharmaceuticals, Inc. Procédé de culture permettant d'obtenir une population clonée de lymphocytes b spécifiques d'antigène
EP2101823B1 (fr) 2007-01-09 2016-11-23 CureVac AG Anticorps code par un arn
WO2009113742A1 (fr) 2008-03-14 2009-09-17 東ソー株式会社 Procédé de fabrication d'un anticorps produit par génie génétique
WO2010080623A2 (fr) 2008-12-18 2010-07-15 Oregon Health & Science University Anticorps anti-fxi et procédés d'utilisation
WO2012067176A1 (fr) 2010-11-17 2012-05-24 中外製薬株式会社 Molécule de liaison à un antigène multi-spécifique ayant une fonction alternative par rapport à la fonction du facteur viii de coagulation sanguine
WO2013120629A1 (fr) 2012-02-15 2013-08-22 Curevac Gmbh Acide nucléique comprenant ou codant pour une tige-boucle d'histone et une séquence poly(a) ou un signal de polyadénylation pour augmenter l'expression d'une protéine thérapeutique codée
WO2013167669A1 (fr) 2012-05-10 2013-11-14 Bayer Pharma Aktiengesellschaft Anticorps capables de lier au facteur de coagulation xi et/ou à sa forme activée, le facteur xia, et utilisations de ceux-ci
WO2018098363A2 (fr) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Anticorps bispécifiques se liant au facteur ix de coagulation et au facteur x de coagulation
WO2018145533A1 (fr) 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anticorps anti-facteur xi de coagulation
WO2019065795A1 (fr) 2017-09-29 2019-04-04 中外製薬株式会社 Molécule multispécifique de liaison à l'antigène ayant une activité de substitution de la fonction de cofacteur du facteur viii de coagulation sanguine (fviii), et formulation pharmaceutique contenant ladite molécule en tant que principe actif
WO2019096874A1 (fr) 2017-11-15 2019-05-23 Novo Nordisk A/S Liants de facteur x renforçant l'activation fx
WO2019235420A1 (fr) * 2018-06-04 2019-12-12 中外製薬株式会社 Procédé permettant de détecter un complexe

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 (zh) 2023-04-04
TW202204421A (zh) 2022-02-01
JP2023106635A (ja) 2023-08-02

Similar Documents

Publication Publication Date Title
US20240190997A1 (en) Antibody substituting for function of blood coagulation factor viii
JP6823677B2 (ja) 血液凝固第viii因子の機能を代替する機能を有する多重特異性抗原結合分子
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 (ru) Биспецифическое антитело, заменяющее функциональные белки
ES2677329T3 (es) Anticuerpos monoclonales contra el inhibidor de la ruta del factor tisular (TFPI)
ES2905085T3 (es) Anticuerpos que pueden unirse a dos epítopos en el inhibidor de la vía del factor tisular (1-161)
ES2765418T3 (es) Anticuerpos monoclonales optimizados contra el inhibidor de la ruta del factor tisular (TFPI)
US20160297892A1 (en) Novel Methods and Antibodies for Treating Coagulapathy
TWI772724B (zh) 具有共同輕鏈的FIXaxFX雙專一性抗體
CA3018124A1 (fr) Anticorps monoclonaux contre le site actif du facteur xi et leurs utilisations
TW202007696A (zh) 經改良的促凝血抗體
KR20210153639A (ko) 이중 특이적 항체
WO2021210667A1 (fr) Molécule de liaison à un antigène bispécifique, composition associée à celle-ci et utilisation, kit et procédé de production de composition
JP6445440B2 (ja) 抗プロテインc抗体による出血性疾患の治療
WO2008026720A1 (fr) Anticorps capable d'améliorer l'activation du facteur viii de coagulation sanguine
WO2020114615A1 (fr) Anticorps bispécifiques se liant au facteur ixa et au facteur x
RU2818588C2 (ru) Биспецифические антитела
ES2865152T3 (es) Anticuerpo biespecífico que sustituye a proteínas funcionales
WO2020115283A1 (fr) Anticorps bispécifiques fixant le facteur ixa et le facteur x
TW202039583A (zh) 結合因子IXa及因子X的蛋白分子

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