WO2011013668A1 - Composition pharmaceutique utilisable dans le cadre du traitement d'événements ischémiques - Google Patents

Composition pharmaceutique utilisable dans le cadre du traitement d'événements ischémiques Download PDF

Info

Publication number
WO2011013668A1
WO2011013668A1 PCT/JP2010/062631 JP2010062631W WO2011013668A1 WO 2011013668 A1 WO2011013668 A1 WO 2011013668A1 JP 2010062631 W JP2010062631 W JP 2010062631W WO 2011013668 A1 WO2011013668 A1 WO 2011013668A1
Authority
WO
WIPO (PCT)
Prior art keywords
vegf
inhibitor
receptor
antibody
composition according
Prior art date
Application number
PCT/JP2010/062631
Other languages
English (en)
Japanese (ja)
Inventor
享良 下畑
カーヴァー ローレンス
Original Assignee
国立大学法人新潟大学
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
Priority claimed from JP2010124374A external-priority patent/JP5823671B2/ja
Priority claimed from JP2010124382A external-priority patent/JP5823672B2/ja
Application filed by 国立大学法人新潟大学 filed Critical 国立大学法人新潟大学
Publication of WO2011013668A1 publication Critical patent/WO2011013668A1/fr
Priority to US13/359,281 priority Critical patent/US8652476B2/en
Priority to US14/151,507 priority patent/US9439961B2/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/164Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • A61K38/166Streptokinase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/49Urokinase; Tissue plasminogen activator
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to a pharmaceutical composition for the treatment of ischemic events, specifically comprising a thrombolytic agent and an inhibitor that inhibits signal transduction mediated by the receptor for vascular endothelial growth factor (VEGF),
  • VEGF vascular endothelial growth factor
  • the present invention relates to a pharmaceutical composition for the treatment of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • Serious ischemic events result from local blood flow blockage or ischemia.
  • cerebral infarction the central part of ischemia in the acute phase is irreversible and cell death occurs even when blood flow is resumed, but there is a reversible incomplete ischemic region around it, especially called penumbra .
  • the central part of the ischemia expands unless treated, and the penumbra gradually disappears.
  • the cerebral infarction portion is pathologically enlarged, clinically dysfunctional, and in the worst case, death occurs.
  • the purpose of treatment in the acute phase of cerebral infarction is to restore blood flow in the penumbra. The recovery depends on the degree of ischemia and its duration. That is, how quickly the blood flow to the penumbra is resumed determines the early recovery of cerebral infarction.
  • Tissue-type plasminogen activator (hereinafter sometimes referred to as “t-PA”) is effective as a thrombolytic therapy that reopens the blood supply to the penumbra by dissolving the thrombus causing ischemia. So it is approved as a treatment for the acute phase of ischemic events.
  • administration of t-PA to patients after the acute phase of an ischemic event is not effective.
  • cerebral infarction results in cerebral hemorrhage complications and prognostic exacerbations.
  • the administration of t-PA to patients after a period of time, ie, 3 hours or more after the onset of cerebral infarction is contraindicated.
  • the present invention relates to a therapeutic pharmaceutical composition that can be administered to patients after acute passing of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism without causing complications such as cerebral hemorrhage.
  • the purpose is to provide.
  • VEGF vascular endothelial growth factor
  • cerebral hemorrhage can be achieved by using the thrombolytic agent and an inhibitor that inhibits signal transduction mediated by the VEGF receptor, for example, an antibody against at least one of VEGF and VEGF receptor and other binding factors. It was found that the thrombolytic drug can be administered to patients who have caused severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism without causing complications of the present invention. .
  • a therapeutic pharmaceutical composition or a treatment method using the composition as one embodiment is provided for the treatment of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • the composition comprises a thrombolytic agent and an inhibitor that inhibits signal transduction mediated by at least one receptor for vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • the inhibitor inhibits signal transduction mediated by the VEGF receptor, for example, by inhibiting the binding of VEGF to the receptor for vascular endothelial growth factor (VEGF).
  • composition or method according to ⁇ 1> wherein the inhibitor decreases binding of VEGF to the VEGF receptor.
  • the inhibitor is a specific binding partner for at least one of VEGF and a VEGF receptor.
  • the specific binding partner is at least one of the following (1) to (4).
  • VEGF peptide or small molecule mimic of VEGF peptide that binds to VEGF receptor but does not activate VEGF receptor (4) is used for stimulation of VEGF receptor VEGF receptor peptides or small molecule mimics of VEGF receptor peptides that reduce the effective level of VEGF.
  • ⁇ 5> At least one of an antibody that binds to a VEGF receptor and antagonizes the binding of VEGF to the VEGF receptor, and an antibody that binds to the VEGF and causes the removal of the VEGF from the blood.
  • the composition or method according to ⁇ 4> wherein ⁇ 6>
  • the composition or method according to ⁇ 1> wherein the inhibitor inhibits the release of VEGF from platelets.
  • the inhibitor reduces binding of ADP to adenosine diphosphate (ADP) receptor.
  • ADP adenosine diphosphate
  • the inhibitor is a specific binding partner for at least one of ADP and ADP receptor.
  • composition or method according to ⁇ 8> wherein the specific binding partner is at least one of the following (1) to (4).
  • the inhibitor is at least an inhibitor that interacts with a component of the VEGF receptor signaling pathway, and an inhibitor that interacts with an enzyme that modifies a component of the VEGF receptor signaling pathway
  • the composition or method according to ⁇ 1> which is any one of the above.
  • composition or method according to ⁇ 10> wherein the inhibitor is a tyrosine kinase inhibitor and an agonist of tyrosine phosphatase.
  • the inhibitor is the composition or method according to ⁇ 1>, wherein the inhibitor decreases production of at least one of VEGF and a VEGF receptor.
  • the inhibitor is at least one of an antisense nucleic acid, a small interfering RNA (siRNA), and a ribozyme.
  • siRNA small interfering RNA
  • ribozyme ⁇ 14>
  • the composition or method according to ⁇ 1> wherein the inhibitor is combined with a thrombolytic drug.
  • ⁇ 15> The composition or method according to ⁇ 14>, wherein the inhibitor binds to the thrombolytic drug as a fusion protein.
  • the thrombolytic drug is at least one of urokinase, streptokinase, tissue-type plasminogen activator (t-PA), and analogs thereof.
  • t-PA tissue-type plasminogen activator
  • VEGF receptor is VEGF receptor type 2 (VEGFR-2).
  • an acute phase of an ischemic event including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • the acute phase of the ischemic event is 3 to 6 hours from the onset of the ischemic event.
  • composition or method according to ⁇ 18>, wherein the acute phase of cerebral infarction is within 3 hours from the onset of the cerebral infarction.
  • ⁇ 21> A polyclonal antibody or a monoclonal antibody, wherein the specific binding partner specifically binds to at least one of VEGF and the VEGF receptor, and inhibits the signal transduction of the VEGF, and an antigen of the antibody
  • the specific binding partner binds to at least one of VEGF-A and VEGF-A receptor, and inhibits a signal transduction pathway mediated by VEGF-A receptor ⁇ 3 > To ⁇ 5>.
  • the specific binding partner is the composition or method according to the above ⁇ 22>, wherein the specific binding partner is an anti-VEGF-A neutralizing antibody or a derivative thereof.
  • a polyclonal antibody or a monoclonal antibody, wherein the specific binding partner specifically binds to at least one of ADP and the ADP receptor and inhibits the signal transduction of the ADP, and an antigen of the antibody The composition or method according to any one of ⁇ 7> to ⁇ 9>, wherein the composition or method is selected from the group consisting of a binding fragment, a recombinant antibody or chimeric antibody containing the antigen-binding fragment, and derivatives thereof.
  • the present invention it is possible to solve the above-described problems and achieve the above-mentioned object, and to prevent serious complications including cerebral infarction, myocardial infarction, and pulmonary embolism without causing complications such as cerebral hemorrhage. It is possible to provide a therapeutic pharmaceutical composition that can be administered to a patient after an acute phase of a bloody event.
  • FIG. 1A is a schematic diagram showing a procedure for producing a conventional rat cerebral infarction model.
  • FIG. 1B is a schematic diagram showing a procedure for producing a rat cerebral infarction model in Example 1.
  • FIG. 2A is a photograph of a coronal section of an animal 24 hours after the onset of cerebral infarction due to thrombus injection.
  • FIG. 2B is a photograph of a coronal section of an animal to which t-PA was administered 1 hour after the onset of cerebral infarction due to thrombus injection.
  • FIG. 2C is a photograph of a coronal section of an animal administered with t-PA 4 hours after the onset of cerebral infarction due to thrombus injection.
  • FIG. 1A is a schematic diagram showing a procedure for producing a conventional rat cerebral infarction model.
  • FIG. 1B is a schematic diagram showing a procedure for producing a rat cerebral infarction model in Example 1.
  • FIG. 2A is a photograph of a cor
  • FIG. 4A is a bar graph showing the volume of cerebral infarction of a TTC-stained coronal section 24 hours after the onset of rats administered with t-PA and an anti-VEGF antibody in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis volume of cerebral infarction (mm 3).
  • FIG. 4B is a bar graph showing the volume of edema of a TTC-stained coronal section 24 hours after the onset of rats administered with t-PA and an anti-VEGF antibody in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • FIG. 4C is a bar graph showing the amount of cerebral hemorrhage of a TTC-stained coronal section 24 hours after the onset of rats administered with t-PA and an anti-VEGF antibody in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis cerebral hemorrhage (mg / dL).
  • FIG. 4D is a band graph showing the motor function scale 24 hours after the onset of rats administered with t-PA and an anti-VEGF antibody in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis Motor function scale.
  • FIG. 5A is a bar graph showing the volume of cerebral infarction in a TTC-stained coronal section 24 hours after the onset of rats administered with t-PA and SU1498 in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis cerebral infarction volume (mm 3 ).
  • FIG. 5B is a bar graph showing the volume of edema of TTC-stained coronal sections 24 hours after the onset of rats administered with t-PA and SU1498 in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis volume of edema (mm 3 ).
  • FIG. 5C is a bar graph showing the amount of cerebral hemorrhage of a TTC-stained coronal section 24 hours after the onset of rats administered with t-PA and SU1498 in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis cerebral hemorrhage (mg / dL).
  • FIG. 5D is a band graph showing a motor function scale 24 hours after the onset of rats administered with t-PA and SU1498 in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Vertical axis Motor function scale.
  • the therapeutic pharmaceutical composition of the present invention comprises at least a thrombolytic agent and an inhibitor that inhibits signal transduction mediated by a receptor for vascular endothelial growth factor (VEGF), and if necessary, other Contains the ingredients.
  • VEGF vascular endothelial growth factor
  • thrombolytic agents include serious ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism (“severe ischemic events”, “ischemic disease”, “ischemia”, “ischemic attack”, etc. Can be appropriately selected according to the purpose, for example, tissue type plasminogen activator (t-PA) or Derivatives thereof, urokinase (see Murray V. et al., J Inter Med. 2010, Feb; 267 (2): 191-208), streptokinase, single-chain urokinase-type plasminogen activator (u-PA), desmoteplase And other proteases that act on fibrin.
  • tissue type plasminogen activator t-PA
  • urokinase see Murray V. et al., J Inter Med. 2010, Feb; 267 (2): 191-208
  • streptokinase single-chain urokinase-type plasminogen activator (u-PA)
  • u-PA
  • the thrombolytic drug preferably contains urokinase, tissue-type plasminogen activator (t-PA), and derivatives or analogs thereof from the viewpoint of increasing the success rate of thrombolysis.
  • t-PA tissue-type plasminogen activator
  • the t-PA derivative is not particularly limited and may be appropriately selected depending on the intended purpose.
  • the t-PA includes sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, and other pharmaceutically acceptable products. And the like which are combined with additives and treatment agents.
  • one or several amino acids may be substituted in the amino acid sequence of t-PA.
  • Specific examples of the t-PA derivative include a t-PA derivative in which a part of amino acids are substituted in the amino acid sequence of the t-PA such as monteplase, pamitepase, and reteplase; and the t-PA such as tenecteplase and lanoteplase.
  • a t-PA derivative in which a part of the amino acid is substituted and the sugar chain is further modified.
  • the content of the serum dissolving drug in the therapeutic pharmaceutical composition is not particularly limited and can be appropriately selected depending on the type of the serum dissolving drug.
  • the “acute phase” in the present invention is an early stage of development of a serious ischemic event including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • cerebral infarction cranial nerve function associated with a decrease in cerebral blood flow
  • the disorder refers to a time when recovery is possible only by rapid resumption of blood flow by the thrombolytic drug.
  • the acute phase generally refers to 3 to 6 hours from the onset of infarction, but in cerebral infarction, it is preferably within 3 hours from the onset.
  • Patient in the present invention includes humans but is not limited to humans.
  • VEGF vascular endothelial growth factor
  • the inhibitor that inhibits signal transduction mediated by the VEGF receptor is not particularly limited and may be appropriately selected depending on the intended purpose.
  • Inhibitors that reduce the binding of VEGF to VEGF receptors, inhibitors that inhibit the release of VEGF from platelets, inhibitors that interact with components of the VEGF receptor signaling pathway, components of the VEGF receptor signaling pathway An inhibitor that interacts with an enzyme that modifies VEGF, an inhibitor that reduces the production of at least one of VEGF and VEGF receptors, and the like.
  • the inhibitor that decreases the binding of VEGF to the VEGF receptor is not particularly limited and may be appropriately selected depending on the intended purpose.
  • a specific binding partner for example, a specific binding partner for VEGF and / or VEGF receptor ( And the like, which are sometimes referred to as “binding inhibitors” that inhibit the binding between VEGF and the VEGF receptor.
  • the specific binding partner is not particularly limited as long as it can inhibit the binding between the VEGF and the VEGF receptor, and can be appropriately selected according to the purpose. It is preferably one that specifically binds to at least one of the VEGF receptors. Thereby, the signal transduction mediated by the VEGF receptor can be inhibited.
  • the specific binding partner include a receptor or a ligand that specifically binds to at least one of the VEGF and the VEGF receptor.
  • the receptor or ligand is not particularly limited and may be appropriately selected depending on the intended purpose. Examples thereof include proteins such as antibodies, carbohydrates, nucleic acids, lipids, and other biopolymers.
  • Antibody in the present invention refers to an antibody that is generally defined, and includes a Fab fragment, a single chain Fv structure, a bispecific antibody in which two different Fab fragments are bound to one Fc, and a similar structure.
  • Fab fragment fragment fragment
  • bispecific antibody in which two different Fab fragments are bound to one Fc
  • similar structure See Bauerle PA. Et al., Cancer Res. 2009, Jun 15; 69 (12): 4941-4.
  • the antibody is human in sequence (transgenic animals expressing the human antibody repertoire (Jakobovits A. et al., Nat Biotechnol. 2007, Oct; 25 (10): 1134-43) or a human antibody gene recombination library (Benhar I.
  • Mimics of antibodies can also be used and include, for example, protein families based on fibronectin, transferrin, glutathione transferase, lens etc. (Wurch T. et al., Curr Pharm Biotechnol. 2008). Dec; 9 (6): 502-9).
  • Other mimetics include non-peptide binding factors such as aptamers consisting of nucleic acids (see Guo KT. Et al., Int J Mol Sci. 2008, Apr; 9 (4): 668-78). Similarly, small molecule peptides (see Holtzman JH. Et al., Biochemistry. 2007, Nov 27; 46 (47): 13541-53), peptide mimetics (eg, beta amino acids (Petersson EJ.
  • antibody mimic refers to all binding agents having the same function as the antibody.
  • an antibody mimic can be used instead of an antibody.
  • the inventors focused on antibodies in the embodiments as excellent examples of such binding factors, but the present invention is not limited thereto.
  • the VEGF is a group of glycoproteins involved in angiogenesis and angiogenesis.
  • the VEGF signaling system is activated.
  • severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism
  • the activation of the VEGF signal transduction system promotes the degradation of proteins that make up the vascular wall.
  • cerebral infarction It was confirmed in the present invention that a complication of cerebral hemorrhage occurs.
  • VEGF-A examples include VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, placental growth factor (PIGF) -1, and PIGF-2.
  • PIGF placental growth factor
  • Each member of the VEGF family has several additional subtypes.
  • human VEGF-A has 121 amino acids (VEGF-A 121 ), 165 amino acids (VEGF-A 165 ), 189 (VEGF-A 189 ), 206 (VEGF-A 206 ), 145 (VEGF-A 145 ), 183 (VEGF-A 183 ) and the like are known.
  • human VEGF-B is known to have 167 amino acids (VEGF-B 167 ), 186 amino acids (VEGF-B 186 ), and the like.
  • the specific binding partner that specifically binds to VEGF may bind to any of the VEGF families.
  • the specific binding partner that specifically binds to VEGF is not particularly limited and may be appropriately selected depending on the purpose.
  • a polyclonal antibody or a monoclonal antibody that recognizes VEGF, an antigen-binding fragment of the antibody A chimeric antibody or a recombinant antibody (hereinafter sometimes referred to as “anti-VEGF antibody” or the like) containing the antigen-binding fragment, or a derivative thereof, a part of a recombinant VEGF receptor (Chu QS. Expert) Opin Biol Ther. 2009, Feb; 9 (2): 263-71), a VEGF variant that competitively binds to VEGF receptor for VEGF but does not activate the VEGF receptor (Sieffle G.
  • the specific binding partner that specifically binds to VEGF is preferably a monoclonal antibody, and the anti-VEGF-A neutralizing antibody is a VEGF receptor of VEGF-A that is involved in vascular disruption during angiogenesis. It is more preferable in that the binding to can be efficiently inhibited.
  • the method for producing a specific binding partner that specifically binds to VEGF is not particularly limited and may be appropriately selected depending on the intended purpose. Examples thereof include a gene recombination method and a synthesis method. Moreover, you may use a commercial item.
  • the specific binding partner that specifically binds to VEGF may be at least one of the anti-VEGF antibody and the like and derivatives thereof, such as polyethylene glycol, other pharmaceutically acceptable additives, Other components such as a treatment agent may be combined or added.
  • the content of other components in the specific binding partner that specifically binds to VEGF is not particularly limited and may be appropriately selected depending on the purpose.
  • the polyclonal antibody is injected into any animal host of mammals (eg, mouse, rat, rabbit, sheep or goat) or birds (eg, chicken) using the VEGF or a fragment thereof as an immunogen.
  • mammals eg, mouse, rat, rabbit, sheep or goat
  • birds eg, chicken
  • an excellent immune response may be induced when linked to a carrier protein such as bovine serum albumin or keyhole limpet hemocyanine.
  • the immunogen is preferably injected into the animal host according to a predetermined schedule incorporating one or more booster immunizations.
  • the immunogen may be injected into the animal host in a mixture with complete or incomplete Freund's adjuvant or other immunopotentiators.
  • the polyclonal antibody is purified from the antiserum by, for example, affinity chromatography using VEGF bound to an appropriate solid support or a fragment thereof, and binding between VEGF and the VEGF receptor is inhibited, In some cases, it has been confirmed that inhibition of binding can inhibit VEGF signaling.
  • the polyclonal antibody include rabbit anti-rat VEGF antibody IgG (RB-222, 19 kDa to 22 kDa) prepared using human recombinant VEGF 165 as an immunogen. The RB-222 can recognize VEGF165 and VEGF121.
  • the monoclonal antibodies may be prepared using the technique of Kohler and Milstein (Eur. J. Immunol. 6: 511-519 (1976)) and improved techniques thereof. These methods involve the preparation of immortal cell lines that can produce antibodies with the desired specificity.
  • the immortal cell line may be prepared from spleen cells derived from an animal host immunized by the same method as the method for producing the polyclonal antibody.
  • the spleen cells are immortalized by various methods to prepare an immortalized cell line capable of producing an antibody.
  • the spleen cells are immortalized by, for example, fusion with myeloma cells derived from the same or different species of the immunized animal. Various fusion techniques known to those skilled in the art may be used.
  • the spleen cells and myeloma cells are mixed with a nonionic surfactant for several minutes and then plated at a low concentration in a selective medium that supports the growth of hybrid cells but does not support the growth of myeloma cells.
  • a preferred selection technique uses HAT (hypoxanthine, aminopterin, thymidine) selection. Hybrid colonies are usually observed after a sufficient time of about 1 to 2 weeks. A single colony is selected, and the single colony is cultured in a medium such as HAT (hypoxanthine, aminopterin, thymidine-added medium), and the culture supernatant is tested for binding activity to the VEGF and these fragments.
  • HAT hypoxanthine, aminopterin, thymidine
  • a hybridoma clone that stably produces a large amount of highly reactive and specific antibody is selected.
  • Monoclonal antibodies may be isolated from the supernatants of colonies of cell lines derived from selected growing hybridoma clones.
  • various techniques may be used to improve yield, such as injecting the hybridoma cell line into the peritoneal cavity of a suitable vertebrate host such as a mouse.
  • the monoclonal antibody may be recovered from the hybridoma cell ascites or blood. Contaminants such as impure proteins from cell debris may be removed from the antibody by conventional techniques such as chromatography, gel filtration, precipitation and extraction.
  • the monoclonal antibody is, for example, bevacizumab obtained by genetically recombining the mouse monoclonal antibody against VEGF, or a Fab fragment of bevacizumab, and genetic modification is performed so that the binding to VEGF is further strengthened.
  • anti-VEGF-A neutralizing antibody of a monoclonal antibody preparation such as Ranibizumab.
  • the monoclonal antibody preparation has already been clinically applied to malignant tumors and has been confirmed to be safe for humans.
  • the antigen-binding fragment of the antibody refers to the part of the antibody that participates in antigen binding.
  • the antigen binding site is formed by amino acid residues in the variable (V) region at the N-terminus of the heavy (H) chain and light (L) chain.
  • the antigen-binding fragment of the antibody includes, in addition to the Fab fragment or F (ab ′) 2 fragment obtained by degrading an intact polyclonal antibody or monoclonal antibody with the proteolytic enzyme papain or pepsin, respectively, Includes Fv fragments containing non-covalent VH and VL region heterodimers containing antigen binding sites that retain much of the binding capacity.
  • the recombinant antibody may be prepared by expression cloning of an antibody gene including transformation into a suitable bacterial host, transfection into a suitable mammalian cell host, and the like.
  • the recombinant antibody can be prepared in large quantities using, for example, gene expression systems derived from prokaryotes and eukaryotes.
  • the chimeric antibody is a fusion protein supported by a constant domain of a homologous or heterologous antibody so that the antigen-binding site of the recombinant antibody can specifically bind to VEGF.
  • the chimeric antibody includes a short chain variable region antibody (scFv) comprising an antibody heavy chain variable region (VH) operably linked to an antibody light chain variable region (VL), camelidae (Camelidae, camel, dromedary, A camel heavy chain antibody (HCAb) or its heavy chain variable region domain (VHH), which is a class of IgG without light chain produced by animals (including llamas).
  • the derivative of the anti-VEGF antibody or the like having binding inhibitory activity between the VEGF and the VEGF receptor is not particularly limited and may be appropriately selected depending on the purpose.
  • the anti-VEGF antibody or Examples include chains, oligonucleotides, polynucleotides, polyethylene glycols, and other pharmaceutically acceptable additives and treatment agents bound thereto.
  • RNA aptamer pegaptanib that binds to the exon 7 portion of the VEGF gene and inhibits the production of VEGF.
  • sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, and other pharmaceutically acceptable additives and treatment agents may be added to the anti-VEGF antibody and the like.
  • These sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, additives and treatment agents are not particularly limited and can be appropriately selected depending on the purpose.
  • the VEGF receptor is a kind of receptor tyrosine kinase, and is involved in expression of actions such as promotion of proliferation and migration of vascular endothelial cells by the ligand VEGF.
  • the VEGF receptor includes VEGFR-1 (sometimes referred to as Flt-1), VEGFR-2 (sometimes referred to as KDR and Flk-1), and VEGFR-3 (sometimes referred to as Flt-4). ), Soluble VEGFR-1, soluble VEGFR-2, soluble VEGFR-3, and the like are known.
  • the VEGF family binds to a specific receptor, VEGF-A binds to VEGFR-1 and VEGFR-2, VEGF-B, PlGF-1, and PlGF-2 bind to VEGFR1, VEGF-C and VEGF-D Binds to VEGFR-2 and VEGFR-3, and VEGF-E binds to VEGFR2.
  • the specific binding partner that specifically binds to the VEGF receptor may bind to any of the VEGF receptors.
  • the specific binding partner that specifically binds to the VEGF receptor is not particularly limited and may be appropriately selected depending on the purpose.
  • the VEGF analog, the VEGF antagonist, and the VEGF receptor A polyclonal or monoclonal antibody that recognizes, an aptamer, an antibody that antagonizes the binding of VEGF to the VEGF receptor, an antibody that binds to the VEGF and causes the removal of the VEGF from the blood, an antigen-binding fragment of these antibodies, A chimeric antibody or a recombinant antibody comprising the antigen-binding fragment (hereinafter sometimes referred to as “anti-VEGFR antibody etc.”), derivatives thereof, VEGF receptor that binds to VEGF receptor but does not activate VEGF receptor, Small molecule mimics of peptides or VEGF peptides Reducing the effective level of VEGF to be used for stimulation of VEGF receptor, VEGF receptor peptide or VEGF receptor small molecule mimetic peptide
  • the specific binding partner that specifically binds to the VEGF receptor is preferably a monoclonal antibody, more preferably an anti-VEGFR-1 neutralizing antibody or an anti-VEGFR-2 antibody.
  • the “small molecule mimic of VEGF peptide” means a molecule that is smaller than a peptide consisting of the complete form of VEGF and that has the same function as the VEGF peptide.
  • the “small mimic of VEGF receptor peptide” means a molecule that is smaller than a peptide consisting of the complete form of VEGF receptor and that has the same function as the VEGF receptor peptide.
  • the specific binding partner that specifically binds to the VEGF receptor may be at least one of the anti-VEGFR antibody and the like, or a derivative thereof, such as polyethylene glycol and other pharmaceutically acceptable additions. Other components such as agents and treatment agents may be combined or added.
  • the content of other components in the specific binding partner that specifically binds to the VEGF receptor is not particularly limited and can be appropriately selected depending on the purpose.
  • polyclonal antibody, monoclonal antibody, antigen-binding fragment can be produced in the same manner as the polyclonal antibody, monoclonal antibody, and antigen-binding fragment that recognize VEGF using the VEGF receptor or these fragments as an immunogen. it can.
  • the recombinant antibody can be produced in the same manner as the recombinant antibody recognizing VEGF.
  • the chimeric antibody recognizes the VEGF except that it is a fusion protein supported by the constant domain of the same or different antibody so that the antigen binding site of the recombinant antibody can specifically bind to the VEGF receptor. Examples thereof include antibodies similar to the chimeric antibody.
  • the derivative of the anti-VEGFR antibody or the like having binding inhibitory activity between the VEGF and the VEGF receptor is not particularly limited and may be appropriately selected depending on the purpose.
  • the anti-VEGF antibody or the like Examples include chains, oligonucleotides, polynucleotides, polyethylene glycols, and other pharmaceutically acceptable additives and treatment agents bound thereto.
  • sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, and other pharmaceutically acceptable additives and treatment agents may be added to the anti-VEGF antibody and the like.
  • examples of the additive, treating agent, sugar chain, oligonucleotide, polynucleotide, polyethylene glycol, and the like are the same as those of the anti-VEGF antibody.
  • These sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, additives and treatment agents are not particularly limited and can be appropriately selected depending on the purpose.
  • the inhibitor that inhibits the release of VEGF from the platelet is not particularly limited and may be appropriately selected depending on the purpose.
  • an adenosine diphosphate (ADP) receptor Bambace NM
  • the inhibitor for reducing the binding of ADP to the ADP receptor is not particularly limited and may be appropriately selected depending on the purpose.
  • a specific binding partner for at least one of ADP and ADP receptor ( And the like, which are sometimes referred to as “binding inhibitors” that inhibit the binding between ADP and the ADP receptor).
  • the specific binding partner is not particularly limited as long as it specifically binds to at least one of the ADP and the ADP receptor and can inhibit the binding between the ADP and the ADP receptor.
  • ADP receptor competitive inhibitor, polyclonal or monoclonal antibody that recognizes ADP or ADP receptor, aptamer binds to ADP receptor but does not activate ADP receptor ADP peptide or ADP peptide small molecule mimic, ADP binding, ADP-binding ADP receptor peptide or ADP receptor peptide small molecule mimic, and the like.
  • the “small molecule mimic of ADP peptide” means a molecule that is smaller than a peptide consisting of the complete form of ADP and that has the same function as the ADP peptide.
  • the “small molecule mimic of the ADP receptor peptide” means a molecule that is smaller than a peptide consisting of the complete form of the ADP receptor and that has the same function as the ADP receptor peptide.
  • Inhibitor that interacts with components of VEGF receptor signaling pathway Inhibitor that interacts with enzymes that modify components of VEGF receptor signaling pathway >> At least one of an inhibitor that interacts with a component of the VEGF receptor signaling pathway and an inhibitor that interacts with an enzyme that modifies a component of the VEGF receptor signaling pathway (hereinafter referred to as “VEGF receptor signaling inhibition”). May be referred to as “agents.”) As bleeding caused by administering t-PA to patients after acute phase of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism Any VEGF receptor-mediated signal transduction inhibitor may be used provided that it can be inhibited.
  • the component of the VEGF receptor signal transduction pathway is not particularly limited and can be appropriately selected according to the purpose.
  • PLC ⁇ phospholipase
  • PLC protein kinase C
  • Raf map kinase kinase
  • MEK map kinase kinase
  • ERK Extracellular signal-regulated kinase
  • PI3K PI3 kinase
  • PDK1 pyruvate dehydrogenase kinase
  • Akt Akt and the like.
  • the enzyme that modifies the components of the VEGF receptor signaling pathway is not particularly limited and may be appropriately selected depending on the intended purpose.
  • the VEGF receptor signaling inhibitor is capable of functioning other biomolecules, such as receptor kinases, provided that the side effects are in an acceptable range for treatment of patients who have caused ischemic events. Examples include those that inhibit other enzyme activities.
  • VEGF receptor signaling inhibitor examples include SU1498 ((E) -3- (3,5-Diisopropyl-4-hydroxyphenyl) -2-((3-phenyl-n-propyl) amino- carbonyl) acrylonitrile), SU5614 (5-Chloro-3-((3,5-dimethylpyrrol-2-yl) methylene) -2-indolinone), SU11248 (N- (2- (diethylamino) ethyl) -5-(( Z)-(5-fluoro-1,2-dihydro-2-oxo-3H-indol-3-ylidine) methyl) -2,4-dimethyl-1H-pyrrole-3-carbamide), A D2171 (4-((4-Fluoro-2-methyl-1H-indol-5-yl) oxy) -6-methyl-7- (3- (pyrrolidin-1-yl) propoxy) quinazoline), PTK
  • An agonist of tyrosine phosphatase can also reduce signal transduction via VEGF receptor tyrosine kinase (see Xu D. et al., Front Biosci. 2008, May 1; 13: 4925-32). These may be used alone or in combination of two or more.
  • the VEGF receptor signaling inhibitors include SU1498, SU5416, SU11248, AZD2171, PTK787 / ZK222584, sorafenib, GW78660B, and other VEGFR-2 kinase inhibitor preparations that are involved in angiogenesis. This is preferable in that it does not affect VEGFR-1-positive cells known to do.
  • VEGFR-2 kinase inhibitor preparation include cediranib (AZD2171), sunitinib (SU11248), varatinib (PTK787 / ZK222584), sorafenib (pazobib), pazobib Is mentioned.
  • ⁇ Inhibitor that reduces production of at least one of VEGF and VEGF receptor >> There is no restriction
  • siRNA small molecule interference RNA
  • the therapeutic pharmaceutical composition may use the thrombolytic agent and an inhibitor that inhibits signal transduction mediated by the VEGF receptor, It may be a single factor having both a thrombolytic effect and an inhibitory effect on signal transduction mediated by the VEGF receptor.
  • Such factors include, for example, the Fab as the thrombolytic drug (Siller-Matula JM et al., Br J Pharmacol, 2010 Feb 1; 159 (3), 502-17, Epub 2009 Dec 24), Bispecific antibodies (fusion proteins) with Fab as an inhibitor can be made. According to this bispecific antibody, Fab as the thrombolytic agent acts on von Willebrand factor (vWF), and Fab as the inhibitor acts on VEGF or VEGF receptor.
  • vWF von Willebrand factor
  • the thrombolytic drug and the inhibitor may be a directly bound fusion protein (Baeuler PA et al., Curr Opin Mol There, 2009, Feb; 11 (1): 22-30).
  • the von Willebrand factor is a high-molecular glycoprotein that is produced in vascular endothelial cells and bone marrow megakaryocytes and is present in plasma, intravascular subcutaneous tissue and platelets.
  • the content of the inhibitor in the therapeutic pharmaceutical composition is not particularly limited and may be appropriately selected depending on the type of the inhibitor.
  • ⁇ Other ingredients> there is no restriction
  • excipients such as lactose, sucrose, sodium chloride, glucose, starch, calcium carbonate, kaolin, microcrystalline cellulose, silicic acid; water, ethanol, propanol , Simple syrup, glucose solution, starch solution, gelatin solution, carboxymethylcellulose, hydroxypropylcellulose, hydroxypropyl starch, methylcellulose, ethylcellulose, shellac, calcium phosphate, polyvinylpyrrolidone, etc .; dry starch, sodium alginate, agar powder, hydrogen carbonate Disintegrating agents such as sodium, calcium carbonate, sodium lauryl sulfate, stearic acid monoglyceride, lactose; lubricants such as purified talc,
  • a flavoring / flavoring agent such as sucrose, orange peel, citric acid or tartaric acid
  • a buffering agent such as sodium citrate
  • a stable agent such as tragacanth, gum arabic and gelatin And the like.
  • pH adjusting agents and buffers such as sodium citrate, sodium acetate, and sodium phosphate; sodium pyrosulfite, EDTA, thioglycolic acid, thiolactic acid, etc.
  • the therapeutic pharmaceutical composition may contain a sugar chain, an oligonucleotide, a polynucleotide, and the like. These sugar chains, oligonucleotides, polynucleotides, polyethylene glycol, additives and treatment agents are not particularly limited and can be appropriately selected depending on the purpose. There is no restriction
  • the administration timing of the therapeutic pharmaceutical composition is not particularly limited and may be appropriately selected according to the purpose. However, after the onset of a serious ischemic event including cerebral infarction, myocardial infarction, and pulmonary embolism 3 hours or more are preferable, and 3 to 6 hours are more preferable.
  • the therapeutic pharmaceutical composition is advantageous in that it can be administered to patients after an acute phase of an ischemic event, and further, it can improve complications such as cerebral hemorrhage and prognostic exacerbation due to administration of the thrombolytic drug. is there.
  • the administration method of the therapeutic pharmaceutical composition is not particularly limited and can be appropriately selected according to the type and content of the thrombolytic drug or the inhibitor in the therapeutic pharmaceutical composition, for example, Oral administration method, injection method, inhalation method and the like.
  • the dose of the therapeutic pharmaceutical composition is not particularly limited, and various factors such as the age, weight, constitution, symptom of the administration subject and the presence or absence of administration of a drug containing other ingredients as active ingredients are considered. Can be selected as appropriate.
  • the animal species to be administered is not particularly limited and can be appropriately selected according to the purpose. For example, human, monkey, pig, cow, sheep, goat, dog, cat, mouse, rat, bird, etc. Among these, it is preferably used for humans.
  • the thrombolytic agent and the inhibitor in the therapeutic pharmaceutical composition may be administered in combination at the same time or may be administered separately. Further, the same composition may be used, the inhibitor may be administered prior to administration of the thrombolytic agent, or the thrombolytic agent may be administered within 30 minutes after administration of the inhibitor. Good.
  • the thrombolytic agent is t-PA
  • plasmin activated by the t-PA is involved in the processing of the VEGF, so that the inhibitor is administered in the brain prior to the administration of the t-PA.
  • the VEGF or the VEGF receptor binds to the inhibitor, and the site of the ischemic event such as the cerebral circulatory system is bound to the VEGF or the VEGF receptor. Therefore, the signal transduction of VEGF is more strongly inhibited. Therefore, the t-PA may be administered after the inhibitor is administered, and the t-PA may be administered within 30 minutes after the inhibitor is administered.
  • the dosage and administration method of the thrombolytic drug are not particularly limited and may be appropriately selected depending on the intended purpose. However, the dosage and administration method according to the instructions of each pharmaceutical manufacturer are preferable. For example, when the thrombolytic drug is alteplase, which is one of the t-PA preparations, the dosage and administration method are not particularly limited and can be appropriately selected according to the purpose.
  • the upper limit is a method of intravenously administering 60 mg to 90 mg per individual. Specifically, a method of intravenously injecting 10% of the total dose by bolus administration for 1 to 2 minutes and the remaining 90% by infusion administration for 1 hour can be mentioned.
  • indication are preferable.
  • the inhibitor is the anti-VEGF-A neutralizing antibody or a derivative thereof
  • a method of intravenously administering 5 mg / kg to 10 mg / kg is preferable.
  • the anti-VEGF-A neutralizing antibody is bevacizumab
  • the dosage and administration method include a method of orally administering 10 mg to 45 mg per individual per day.
  • the dosage and administration method include a method of orally administering 25 mg to 75 mg per individual once a day.
  • the dosage and administration method thereof are not particularly limited and can be appropriately selected according to the purpose.
  • 400 mg to 800 mg per individual Examples include a method of oral administration once a day.
  • the dosage and administration method thereof are not particularly limited and can be appropriately selected depending on the purpose.
  • 500 mg to 1,500 mg per individual is used.
  • a method of oral administration once a day for example, when the inhibitor is pazopanib, the dosage and administration method thereof are not particularly limited and can be appropriately selected according to the purpose.
  • 400 mg to 1,200 mg per individual is used.
  • a method of oral administration once a day is 400 mg to 1,200 mg per individual is used.
  • the dosage and administration method of the therapeutic pharmaceutical composition are not particularly limited, depending on the purpose. It can select suitably, According to the kind of said thrombolytic agent and the said inhibitor in the said composition, content, etc., it can select suitably.
  • the therapeutic pharmaceutical composition can also be administered to patients after an acute phase of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism, and can improve complications such as cerebral hemorrhage and prognostic deterioration Therefore, it can be suitably used for treating severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • an inhibitor that inhibits signal transduction mediated by the VEGF receptor at the same time as the step of administering a thrombolytic agent and the step of administering the thrombolytic agent or in advance. It is preferable to use a treatment method including the step of administering.
  • a kit containing the thrombolytic drug and the inhibitor is also included in the present invention.
  • the inhibitor in the kit may be an inhibitor that inhibits signal transduction caused by the binding of VEGF to the VEGF receptor, or an antibody that inhibits the binding between VEGF and the VEGF receptor, Other binding factors may be used.
  • Example 1 Preparation of rat cerebral infarction model
  • Example 2 Preparation of rat cerebral infarction model
  • Sprague-Dawley rats male, 8 weeks old, obtained from Charles River, Japan
  • FIG. 1A and FIG. 1B the preparation method of the rat cerebral infarction model of this invention is demonstrated.
  • a nylon thread is applied to the bifurcation of the external carotid artery (ECA) 1 and the common carotid artery (CCA) 3, or from the external carotid artery (ECA) 1 to the origin of the middle cerebral artery (MCA) 2.
  • the middle cerebral artery was blocked by invading (FIG. 1A).
  • the thrombus was obtained by coagulating rat autologous blood and thrombin as a gel in a polyethylene tube catheter (PE-50, manufactured by Becton Dickinson) with a diameter of 0.35 mm. After being allowed to stand overnight, it was cut into a length of 1 mm. It was. The thrombus was injected from the rat external carotid artery (ECA) 1 into the rat middle cerebral artery (MCA) 2 under 1% to 1.5% by weight halothane anesthesia using the catheter.
  • ECA rat external carotid artery
  • MCA rat middle cerebral artery
  • brain surface blood flow values were measured using a laser Doppler blood flow meter (AFL21, Advance Co., Ltd., Tokyo).
  • AFL21 Advance Co., Ltd., Tokyo
  • An animal having a cerebral blood flow value of less than 50% compared to that before infusion of thrombus was used as a rat cerebral infarction model animal in the following experiment.
  • t-PA (alteplase, manufactured by Mitsubishi Tanabe Pharma Corporation), a thrombolytic drug, was intravenously injected into the femoral vein for 1 hour or 4 hours after thrombus injection (30 minutes). 10 mg / kg, 10% bolus administration and 90% infusion administration).
  • FIG. 2A to 2C are photographs of coronal sections showing the cerebral infarction reducing effect and cerebral hemorrhage-inducing effect of t-PA administration.
  • a black part shows a healthy tissue, and a white part shows a cerebral infarction part.
  • Extensive cerebral infarction was observed in the operative cerebrum after 24 hours without administration of t-PA after thrombus injection (FIG. 2A).
  • t-PA was administered 1 hour after thrombus injection, a reduction in the cerebral infarction portion was observed as compared to animals not treated with t-PA (FIG. 2B).
  • Example 2 Inhibition of VEGF expression using anti-VEGF antibody
  • 100 ⁇ g of rabbit anti-rat VEGF antibody IgG (RB-222, Lab Vision- Neomarkers (hereinafter sometimes referred to as “anti-VEGF antibody”) was administered as a bolus with t-PA.
  • 100 ⁇ g of rabbit anti-human IgG (R5G10-048, manufactured by OEM Concepts, hereinafter sometimes referred to as “control antibody”) was administered as a bolus with t-PA.
  • an anti-VEGF antibody (SC-152, manufactured by Santa Cruz Biotechnologies, dilution ratio 1: 200) is used as a primary antibody, and a peroxidase-conjugated anti-rabbit IgG antibody (dilution ratio 1:10) is used as a secondary antibody. , 000) was used.
  • the anti- ⁇ -actin antibody (SC-1616, SC-1616, (Santa Cruz Biotechnologies, dilution ratio 1: 2,000) and the secondary antibody were reacted to detect ⁇ -actin.
  • FIG. 3 is a Western blot diagram showing that VEGF expression was suppressed after co-administration of t-PA and anti-VEGF antibody.
  • Lane 1 shows a sample of an animal that did not develop cerebral infarction due to thrombus injection
  • lane 2 shows a sample of an animal that did not develop cerebral infarction due to thrombus injection and administered t-PA and a control antibody.
  • 3 shows a sample of an animal to which only a control antibody was administered 1 hour after the onset of cerebral infarction due to thrombus injection
  • lane 4 represents an animal to which t-PA and a control antibody were administered 1 hour after the onset of cerebral infarction due to thrombus injection.
  • lane 5 shows a sample of an animal administered with t-PA and an anti-VEGF antibody in combination 1 hour after the onset of cerebral infarction due to thrombus injection
  • lane 6 shows a sample of t -Shows samples of animals administered PA and control antibody
  • lane 7 is an animal administered together with t-PA and anti-VEGF antibody 4 hours after cerebral infarction due to thrombus injection It illustrates a sample.
  • VEGF vascular endothelial cell damage and subsequent cerebral blood barrier dysfunction are related to cerebral hemorrhage after t-PA administration.
  • VEGF activates MMP-9
  • activated MMP-9 is known to degrade proteins involved in the brain blood barrier, such as Zona oculusens-1 and basement membrane type IV collagen. Therefore, without being bound by theory, the mechanism of action of the combined administration of t-PA and anti-VEGF antibody is to suppress the increase in VEGF by t-PA administration after acute cerebral infarction. It may be explained by preventing cerebral hemorrhage by preventing cerebral blood barrier dysfunction such as -9 activation.
  • Example 3 Evaluation of influence of combined administration of t-PA and anti-VEGF antibody
  • the co-administration of t-PA and anti-VEGF antibody was performed as described in Example 2.
  • the effect of the combined administration of t-PA and anti-VEGF antibody 4 hours after the onset of cerebral infarction due to thrombus injection is the effect of cerebral infarct volume, edema on the TTC-stained coronal section 24 hours after the onset of cerebral infarction due to thrombus injection. Volume, cerebral hemorrhage, and motor function scales were measured and evaluated.
  • the volume of cerebral infarction and edema of the TTC-stained coronal section is described in Swanson, R. et al. A. (J. Cereb.
  • 4A to 4C are bar graphs showing cerebral infarction volume, edema volume, and cerebral hemorrhage volume, respectively, of a TTC-stained coronal section 24 hours after the onset of cerebral infarction due to thrombus injection.
  • the white bar is the group that received only control antibody 4 hours after the onset of cerebral infarction due to thrombus injection, and the black bar is the group that received t-PA and control antibody 4 hours after the onset of cerebral infarction due to thrombus injection.
  • the gray bar is a group to which t-PA and anti-VEGF antibody were administered 4 hours after the onset of cerebral infarction due to thrombus injection.
  • FIG. 4D is a band graph showing the motor function scale 24 hours after the onset of cerebral infarction due to thrombus injection.
  • the different colored parts of the band represent the number of individuals in each of the five stages.
  • the left band shows a group (number of individuals 23) administered only with control antibody 4 hours after the onset of cerebral infarction due to thrombus injection, and the middle band shows t-PA and control 4 hours after the onset of cerebral infarction due to thrombus injection.
  • the group to which the antibody was administered (20 individuals) was shown, and the right band represents the group to which t-PA and anti-VEGF antibody were administered 4 hours after the onset of cerebral infarction due to thrombus injection (12 individuals).
  • the combined administration of t-PA and anti-VEGF antibody can prolong the time until administration of t-PA in patients who have developed cerebral infarction, and is associated with cerebral hemorrhage complications. It was shown that the motor function and the survival rate can be improved while preventing the above.
  • Example 4 Combined administration of t-PA and SU14978
  • a VEGF receptor kinase inhibitor included in the VEGF receptor.
  • Specific inhibitors for the VEGF receptor include SU1498 ((E) -3- (3,5-diisopropyl-4-hydroxyphenyl) -2-((3-phenyl-n-propyl) aminocarbonyl) acrylonitrile, Calbiochem. Catalog No. 572888) manufactured by the company was used.
  • SU1498 was dissolved in 1 mL of DMSO (dimethyl sulfoxide) per kg of the patient's body weight to give 20 mg / kg, and was administered as a single bolus with t-PA 4 hours after cerebral infarction.
  • DMSO dimethyl sulfoxide
  • 5A to 5C show the volume of cerebral infarction and the volume of edema, respectively, of TTC-stained coronal sections 24 hours after the onset of rats administered with t-PA and SU1498 at 4 hours after the onset of cerebral infarction due to thrombus injection.
  • a bar graph showing the amount of cerebral hemorrhage.
  • the black bar is a group administered with t-PA and DMSO 4 hours after the onset of cerebral infarction due to thrombus injection
  • the gray bar is a group administered with t-PA and SU1498 4 hours after the onset of cerebral infarction due to thrombus injection It is.
  • FIG. 5D is a band graph showing a motor function scale 24 hours after the onset of rats administered with t-PA and SU1498 in combination 4 hours after the onset of cerebral infarction due to thrombus injection.
  • Different colored bands represent each of the five levels.
  • the left band shows the group administered t-PA and DMSO 4 hours after the onset of cerebral infarction due to thrombus injection
  • the right band shows the group administered t-PA and SU1498 4 hours after the onset of cerebral infarction due to thrombus injection Indicates. The number of individuals was 10 in both cases. From the comparison of the left and right bands, the group administered t-PA and SU1498 4 hours after the onset of cerebral infarction tended to improve the prognosis more than the group administered t-PA and DMSO.
  • the combined administration of t-PA and SU1498 is similar to the combined administration of t-PA and anti-VEGF antibody in the time until administration of t-PA in patients with cerebral infarction. It has been shown that the motor function and survival rate can be improved while preventing cerebral hemorrhage complications.
  • the therapeutic pharmaceutical composition can also be administered to patients after an acute phase of severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism, and can improve complications such as cerebral hemorrhage and prognostic deterioration Therefore, it can be suitably used for treating severe ischemic events including cerebral infarction, myocardial infarction, and pulmonary embolism.
  • ECA External carotid artery
  • MCA Middle cerebral artery
  • CCA Common carotid artery

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Zoology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne une composition pharmaceutique thérapeutique comprenant un agent thrombolytique et un inhibiteur capable d'inhiber une signalisation à médiation par un récepteur d'un facteur de croissance de l'endothélium vasculaire (VEGF). Ladite composition pharmaceutique thérapeutique peut être utilisée pour le traitement d'événements ischémiques graves, dont l'infarctus cérébral, l'infarctus du myocarde et l'embolie pulmonaire.
PCT/JP2010/062631 2009-07-27 2010-07-27 Composition pharmaceutique utilisable dans le cadre du traitement d'événements ischémiques WO2011013668A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/359,281 US8652476B2 (en) 2009-07-27 2012-01-26 Pharmaceutical composition for treating ischemic events
US14/151,507 US9439961B2 (en) 2009-07-27 2014-01-09 Pharmaceutical composition for treating ischemic events

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
JP2009-174099 2009-07-27
JP2009174099 2009-07-27
JP2009-174098 2009-07-27
JP2009174098 2009-07-27
JP2010-124374 2010-05-31
JP2010124374A JP5823671B2 (ja) 2009-07-27 2010-05-31 免疫製剤を含む脳梗塞治療用医薬品組成物
JP2010-124382 2010-05-31
JP2010124382A JP5823672B2 (ja) 2009-07-27 2010-05-31 受容体シグナル伝達阻害剤を含む脳梗塞治療用医薬品組成物

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/359,281 Continuation-In-Part US8652476B2 (en) 2009-07-27 2012-01-26 Pharmaceutical composition for treating ischemic events

Publications (1)

Publication Number Publication Date
WO2011013668A1 true WO2011013668A1 (fr) 2011-02-03

Family

ID=43529322

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2010/062631 WO2011013668A1 (fr) 2009-07-27 2010-07-27 Composition pharmaceutique utilisable dans le cadre du traitement d'événements ischémiques

Country Status (1)

Country Link
WO (1) WO2011013668A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9913899B2 (en) 2014-03-13 2018-03-13 Shimojani, LLC Diagnostic marker for treatment of cerebral ischemia
US10617756B2 (en) 2017-01-05 2020-04-14 Shimojani, LLC Drug regimen for treatment of cerebral ischemia

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6322026A (ja) * 1986-05-12 1988-01-29 ザ ウエルカム フアウンデ−シヨン リミテツド 医薬組成物
JPH11515010A (ja) * 1995-10-25 1999-12-21 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ 神経保護物質および血栓溶解剤の注射剤
JP2002534359A (ja) * 1998-12-22 2002-10-15 ジェネンテック・インコーポレーテッド 血管内皮細胞増殖因子アンタゴニストとその用途
WO2007128526A2 (fr) * 2006-05-04 2007-11-15 Fovea Pharmaceuticals Sa Méthodes de traitement des maladies néovasculaires

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6322026A (ja) * 1986-05-12 1988-01-29 ザ ウエルカム フアウンデ−シヨン リミテツド 医薬組成物
JPH11515010A (ja) * 1995-10-25 1999-12-21 ジヤンセン・フアーマシユーチカ・ナームローゼ・フエンノートシヤツプ 神経保護物質および血栓溶解剤の注射剤
JP2002534359A (ja) * 1998-12-22 2002-10-15 ジェネンテック・インコーポレーテッド 血管内皮細胞増殖因子アンタゴニストとその用途
WO2007128526A2 (fr) * 2006-05-04 2007-11-15 Fovea Pharmaceuticals Sa Méthodes de traitement des maladies néovasculaires

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Annual Meeting of the Japan Neurosurgical Society Abstract, 2007", vol. 66TH, article YOSHIYUKI CHIBA: "Rat No Kyoketsu Model ni Okeru VEGF Receptor Kikkozai no Koka", pages: 1K - 01-1-2 *
MASATO KANAZAWA ET AL.: "Rat Chu Daino Domyaku Sokusen Model ni Okeru Kekkan Naihi Saibo no VEGF Hatsugen", PROGRAM AND ABSTRACTS OF THE MEETING OF THE JAPANESE SOCIETY OF NEUROLOGY, vol. 49TH, 2008, pages 267 *
RYOTA KIMURA ET AL.: "VEGF Chuwa Kotai no No Jomyaku Kosoku ni Taisuru No Hogo Koka", JOMYAKU, vol. 15, no. 2, 2004, pages 155 *
TAKAYOSHI SHIMOHATA ET AL.: "Post t-PA Jidai no Shinkei-Kekkan Hogo Ryoho", NIIGATA MEDICAL ASSOCIATION KAIHO, January 2009 (2009-01-01), pages 2 - 6 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9913899B2 (en) 2014-03-13 2018-03-13 Shimojani, LLC Diagnostic marker for treatment of cerebral ischemia
US10617756B2 (en) 2017-01-05 2020-04-14 Shimojani, LLC Drug regimen for treatment of cerebral ischemia
US11077188B2 (en) 2017-01-05 2021-08-03 Shimojani, LLC Drug regimen for treatment of cerebral ischemia
EP4289430A2 (fr) 2017-01-05 2023-12-13 Shimojani LLC Agent thrombolytique et bevacizumab pour le traitement de l'ischémie cérébrale

Similar Documents

Publication Publication Date Title
JP6941148B2 (ja) 補体成分c5抗体
CN108178789B (zh) 内皮糖蛋白多肽及其用途
AU2009291747B2 (en) Methods for inhibiting ocular angiogenesis
JP6643244B2 (ja) 補体因子Bb抗体
EP2170366B1 (fr) Composition et procédé pour le traitement d'une lésion de reperfusion et d'un dommage tissulaire
AU2003215337B2 (en) Methods of extending corneal graft survival
JP2023025005A (ja) 疾患の治療または予防に用いるためのファクター1およびファクター2タンパク質、およびその阻害剤
JP2003520826A (ja) 血管新生を抑制するための治療方法
CA2625992A1 (fr) Inhibition de la metastase et/ou de l'angiogenese induites par le facteur de croissance du placenta (p1gf)
Guyot et al. VEGF splicing and the role of VEGF splice variants: from physiological-pathological conditions to specific pre-mRNA splicing
JP2002539076A (ja) 新脈管形成抑制のための方法および組成物
JP2018158948A (ja) 免疫製剤を含む脳梗塞治療用医薬品組成物
US9439961B2 (en) Pharmaceutical composition for treating ischemic events
JP2014221769A (ja) Egfl8アンタゴニストを使用する血管新生阻害方法
WO2022192445A1 (fr) Modulation de la signalisation wnt dans des troubles gastro-intestinaux
JPWO2005018671A1 (ja) 癌転移阻害剤
WO2011013668A1 (fr) Composition pharmaceutique utilisable dans le cadre du traitement d'événements ischémiques
JP2003508352A (ja) MMP−9およびβ1インテグリンに基づくアンタゴニストを用いた血管形成の抑制のための新規な方法および組成物
Jin et al. FP3: a novel VEGF blocker with antiangiogenic effects in vitro and antitumour effects in vivo
JP5823672B2 (ja) 受容体シグナル伝達阻害剤を含む脳梗塞治療用医薬品組成物
Arrigo et al. Towards the Development of Longer and More Efficacious Therapies for Wet and Dry Age-related Macular Degeneration
CN114980929A (zh) 糖尿病性自主神经障碍的预防或治疗剂

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: 10804414

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10804414

Country of ref document: EP

Kind code of ref document: A1