[go: up one dir, main page]

WO2025033330A1 - Antitumor therapy by combined use of chymase inhibitor and immunotherapy - Google Patents

Antitumor therapy by combined use of chymase inhibitor and immunotherapy Download PDF

Info

Publication number
WO2025033330A1
WO2025033330A1 PCT/JP2024/027634 JP2024027634W WO2025033330A1 WO 2025033330 A1 WO2025033330 A1 WO 2025033330A1 JP 2024027634 W JP2024027634 W JP 2024027634W WO 2025033330 A1 WO2025033330 A1 WO 2025033330A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
chymase inhibitor
chymase
cells
interleukin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/JP2024/027634
Other languages
French (fr)
Japanese (ja)
Inventor
真司 ▲高▼井
徳男 金
春樹 岡村
文 李
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
International Cooperation For Medical Innovation Co Ltd
Original Assignee
International Cooperation For Medical Innovation Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by International Cooperation For Medical Innovation Co Ltd filed Critical International Cooperation For Medical Innovation Co Ltd
Publication of WO2025033330A1 publication Critical patent/WO2025033330A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a novel antitumor therapy characterized by combining a chymase inhibitor with immunotherapy using interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, interleukin-12 (IL-12), or the like.
  • IL-18 interleukin-18
  • IL-12 interleukin-12
  • Chymase is a neutral serine protease found mainly in mast cells. Human chymase is known to be involved in hypertension and arteriosclerosis by producing angiotensin II using angiotensin I as a substrate. There have been reports showing that chymase correlates with angiogenesis around tumor tissues in lung cancer, colon cancer, and pancreatic cancer (Non-Patent Documents 1-3), but there have been no reports showing the direct effect of chymase on tumors.
  • Interleukin-18 is a cytokine discovered by Okamura et al. as an interferon-gamma (IFN ⁇ ) production inducer.
  • IFN ⁇ interferon-gamma
  • IL-18 binds to the IL-18 receptor expressed on the cell surface of cells in which IFN ⁇ production is induced in the presence of IL-12, enhancing IFN ⁇ production and enhancing Th1-type immune responses.
  • IL-18 enhances Th2-type immune responses.
  • IL-18 is expected to have an antitumor effect
  • cancer treatment drugs that combine IL-18 with rituximab or Herceptin (Patent Document 1) and with a molecular target antibody have been reported (Patent Document 2).
  • Patent Document 4 Recently, it has been reported that a strong antitumor effect was achieved by using CAR-T cells that secrete IL-18 to secrete IL-18 in a tumor-specific manner (Non-Patent Document 4).
  • IL-18 is produced as an IL-18 precursor containing a propeptide, which is cleaved by caspases and released outside the cell as active IL-18.
  • a method for producing IL-18 has been reported in which the IL-18 precursor is cleaved by chymase instead of caspase (Patent Document 3). This report does not show the effect of chymase inhibitors on active (mature) IL-18, but suggests that chymase inhibitors can inhibit IL-18 activity by inhibiting the conversion of the IL-18 precursor to the active form.
  • PD-1 and PD-L1 are representative immune checkpoint molecules, and antibodies targeting these molecules have been shown to have significant anti-cancer effects in a variety of cancer types, and are currently being developed as medicines.
  • the remission rate for solid cancers with anti-PD-1/PD-L1 antibody therapy is low, and the response rate cannot be said to be sufficient.
  • the objective of the present invention is to provide a novel method for treating tumors.
  • a pharmaceutical composition for treating a tumor comprising a chymase inhibitor as an active ingredient, in combination with one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
  • IL-18 interleukin-18
  • IL-12 interleukin-12
  • a pharmaceutical composition for treating a tumor comprising a combination of one or more selected from interleukin-18 (IL-18), PD-1/PD-L1 antibody, and interleukin-12 (IL-12) and a chymase inhibitor.
  • a pharmaceutical composition for treating a tumor comprising as an active ingredient any one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), wherein the pharmaceutical composition is used in combination with a chymase inhibitor.
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenyle
  • [5] The pharmaceutical composition according to any one of [1] to [4], wherein one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
  • [6] The pharmaceutical composition according to any one of [1] to [5], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-am ino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethy
  • the present application provides the following [1] to [6].
  • [1] A method for treating a tumor, comprising administering to a subject a combination of a chymase inhibitor and one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
  • IL-18 interleukin-18
  • IL-12 interleukin-12
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-am ino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethy
  • IL-18 interleukin-18
  • IL-12 interleukin-12
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-amino-2-(4-fluoroph enyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenyleth
  • the present application provides the following [1] to [7].
  • An agent for use in treating a tumor comprising a chymase inhibitor, wherein the agent is used in combination with any one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
  • a drug for use in treating a tumor comprising a combination of one or more selected from interleukin-18 (IL-18), PD-1/PD-L1 antibody, and interleukin-12 (IL-12) and a chymase inhibitor.
  • a drug for use in treating a tumor comprising any one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), characterized in that the drug is used in combination with a chymase inhibitor.
  • IL-18 interleukin-18
  • IL-12 interleukin-12
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenyle
  • [5] The composition for use according to any one of [1] to [4], wherein IL-18, the anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
  • [6] The drug for use according to any one of [1] to [5], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
  • [7] The agent for use according to any of [1] to [6], wherein the chymase inhibitor enhances the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12.
  • the present application provides the following [1] to [7].
  • [1] Use of one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), together with a chymase inhibitor, in the manufacture of a medicine for treating a tumor.
  • [2] Use of a chymase inhibitor in the manufacture of a medicament for treating a tumor, characterized in that the medicament is used in combination with one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
  • Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenyle
  • [5] The use according to any one of [1] to [4], wherein IL-18, the anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
  • [6] The use according to any one of [1] to [5], wherein one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
  • the chymase inhibitor enhances the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12.
  • the present invention provides novel clinical applications of chymase inhibitors and immunotherapeutic agents such as IL-18, anti-PD-1/PD-L1 antibodies, and L-12.
  • FIG. 1 shows survival curves for the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group 100 days after tumor (CT26) cell transplantation.
  • FIG. 2 shows photographs of the tumor transplant site of a mouse that survived 100 days after tumor (CT26) cell transplantation (top: IL-18, bottom: combined use of IL-18 and chymase inhibitor).
  • FIG. 3 shows survival curves for the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group, 110 days after tumor (CT26) cell transplantation.
  • FIG. 1 shows survival curves for the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group 100 days after tumor (CT26) cell transplantation.
  • FIG. 2 shows photographs of the tumor transplant site of a mouse that survived 100 days after tumor (CT26) cell transplantation (top: IL-18, bottom: combined use of IL-18 and chymase inhibitor).
  • FIG. 4 shows the percentage (A), absolute number (B), cell size (C), and size of intracellular granules (D) of mast cells in ascites after tumor (CT26) cell transplantation.
  • FIG. 5 shows the number of CD4 + T cells (A), the number of CD8 + T cells (B), and the ratio of CD4 + T cells/CD8 + T cells (C) after tumor (CT26) cell transplantation.
  • FIG. 6 shows the percentage of NK cells in ascites after tumor (CT26) cell transplantation (A), the absolute number of NK cells (B), and the percentage of DX5 + B220 + NK cells (C).
  • FIG. 7 shows the change in the numbers of immune cells (mast cells, CD8 + T cells, NK cells, and DX5 + B220 + NK cells) in the ascites after tumor (CT26) cell transplantation.
  • Figure 8 shows the results of comparing T cells in ascites in the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group.
  • the ratio of CD4 + T cells/CD8 + T cells A
  • CD4 + T cell count B
  • CD8 + T cell count C
  • D CD44high CD8 + T cell count
  • Figure 9 shows the results of comparing the number of NK cells in ascites in the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combined administration group.
  • Figure 10 shows survival curves for the placebo group, the anti-PD-1 antibody administration group, the chymase inhibitor administration group, and the anti-PD-1 antibody/chymase inhibitor combination group 34 days after tumor (CT26) cell transplantation.
  • a chymase inhibitor means a drug capable of inhibiting the activity of chymase.
  • Chymase is a neutral serine protease
  • human chymase is known to be involved in hypertension and arteriosclerosis by producing angiotensin II from angiotensin I.
  • the chymase inhibitor of the present invention is preferably a human chymase inhibitor.
  • a known chymase inhibitor can be used as the chymase inhibitor.
  • known chymase inhibitors include compounds that have already been reported for clinical application or validation in preclinical models, such as SUN-C8257, SUN13834, BCEAB, Compound 17, NK3201, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), Suc-Val-Pro-Phe P (OPh) 2 , Y-40613, Y-40079, TY-51469, Chymostatin, Fulacimstat (BAY1142524), ASB17061, ONO-8020435, and ONO-2220445 (Heuston and Hyland, Br J Pharmacol. 2012 Oct; 167(4): 732-740.doi: 10.1111/j.1476-5381.2012.02055.x etc.)
  • WO2016/132483, JP 2016-150915, JP 2016-535095, JP 2012-516335, JP 2012-502034, JP 2012-121898, JP 2010-536768, WO2010/147133, JP 2010-515699, JP 2010-505900, WO Chymase inhibitors disclosed in WO 2005/018672, WO03/018061, JP 2004-507539, WO01/062294, WO01/062293, WO01/062292, WO00/005204, WO99/032459, JP 11-001479, and JP 10-087493 can also be used.
  • Interleukin-18 Interleukin-18 (IL-18) is a cytokine that was discovered as an IFN- ⁇ production inducer. IL-18, together with IL-12, acts on CD4 + CD8 + T cells and NK cells to induce IFN- ⁇ production.
  • IL-18 is produced as an inactive precursor (pro-IL-18) containing the propeptide, and is released as active IL-18 following cleavage of the propeptide by caspase 1, caspase 4, etc.
  • the source of IL-18 is not particularly limited, but it is preferable that the IL-18 is derived from the same species as the subject to be administered, or a closely related species. Therefore, when administered to humans, mammalian IL-18, particularly human IL-18, is preferred.
  • Human IL-18 precursor consists of 193 amino acids (24 KDa), and mature (active) human IL-18 without the propeptide consists of 157 amino acids (18 KDa).
  • the amino acid sequence of human IL-18 is known, and an example of the amino acid sequence of human IL-18 is the amino acid sequence shown in SEQ ID NO: 1.
  • Human IL-18 having an amino acid sequence that has 80%, 85%, 90%, 95%, 96%, 98%, or 99% or more sequence identity with this amino acid sequence can also be used as the IL-18 of the present invention as long as it has the activity of IL-18.
  • IL-18 activity means the physiological activity of IL-18, such as IFN ⁇ production-inducing activity.
  • human IL-18 is the following protein (1) or (2): (1) A protein comprising the amino acid sequence shown in SEQ ID NO:1, or (2) A protein having 80% or more, preferably 90% or more, and more preferably 95% or more sequence identity with the amino acid sequence shown in SEQ ID NO:1 and having IL-18 activity (such as IFN ⁇ production-inducing activity).
  • IL-18 exerts its physiological activity by binding to a receptor on the cell membrane, but since there is an excess of IL-18-specific IL-18-binding proteins in the body, much of the secreted IL-18 loses its activity upon binding to the IL-18-binding proteins. Recently, it has been reported that the antitumor effect of IL-18 is enhanced by using modified IL-18 with increased stability and by secreting IL-18 from Car-T cells. Such modified IL-18 and new delivery systems are also within the scope of the present invention, so long as they do not contradict the objectives of the present invention.
  • Anti-PD-1/PD-L1 antibody (anti-PD-1 antibody, anti-PD-L1 antibody)
  • anti-PD-1/PD-L1 antibody refers to an antibody that specifically inhibits PD-1 or PD-L1, an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • anti-PD-1 antibody used in the present invention is not particularly limited as long as it can bind to PD-1 and inhibit its function as an immune checkpoint.
  • Anti-PD-1 antibodies include those that have already been approved and sold as medicines and those that are under development, and these can be used preferably. Examples of such "anti-PD-1 antibodies” include nivolumab (Opdivo (GSK/Ono Pharmaceutical)), pembrolizumab (Merck), which is a humanized IgG4 antibody, and dipirizumab, but are not limited to these.
  • anti-PD-L1 antibody used in the present invention is not particularly limited as long as it can bind to PD-L1 and inhibit its function as an immune checkpoint.
  • PD-L1 antibodies include those that have already been approved (sold) as medicines and those that are under development, and these can be used preferably.
  • Examples of such "anti-PD-L1 antibodies” include, but are not limited to, atezolizumab (Roche/Chugai), durvalumab (AstraZeneca), avelumab (Merck), and MED10680/AMP-514.
  • the antibody may be produced according to a conventional method.
  • the antibody is preferably a chimeric antibody, a humanized antibody, or a fully human antibody in order to reduce heterologous antigenicity to humans.
  • the antibody may be an antibody fragment so long as it has a function as an immune checkpoint inhibitor. Examples of the antibody fragment include F(Ab') 2 , Fab', Fab, Fv, and scFv.
  • Interleukin-12 Interleukin-12 (IL-12) is a cytokine produced mainly by macrophages, polymorphonuclear leukocytes, dendritic cells, etc., and is known to induce the production of IFN- ⁇ in NK cells and T cells, and to enhance the activity of NK cells and cytotoxic T cells.
  • IL-12 preferentially activates Th1 cells and activates cellular immunity by suppressing Th2 cells, and is therefore expected to be used in the treatment of Th2-driven immune disorders and in the treatment of cancer and infectious diseases by activating cellular immune function.
  • IL-12 along with IL-18, acts on CD4 + CD8 + T cells and NK cells to induce the production of IFN- ⁇ . Therefore, IL-12 and IL-18 may be used in combination in the treatment of cancer and infectious diseases.
  • compositions are characterized by effectively treating tumors by combining one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 with a chymase inhibitor that does not exhibit anti-tumor activity alone.
  • the pharmaceutical composition of the present invention is a pharmaceutical composition for treating a tumor, comprising a chymase inhibitor as an active ingredient, and characterized in that it is used in combination with one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.
  • the pharmaceutical composition of the present invention is a pharmaceutical composition for treating a tumor, comprising a combination of a chymase inhibitor and one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.
  • the chymase inhibitor and one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 may be formulated together, or may be formulated separately as a kit formulation.
  • the pharmaceutical composition of the present invention is a pharmaceutical composition for treating tumors, comprising one or more active ingredients selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12, and is characterized in that it is used in combination with a chymase inhibitor.
  • the pharmaceutical composition of the present invention is a pharmaceutical composition for enhancing the action of one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 in tumor treatment, comprising a chymase inhibitor as an active ingredient.
  • the pharmaceutical composition of the present invention may contain pharmacologically acceptable carriers and additives.
  • carriers and additives include excipients, binders, lubricants, solvents, disintegrants, solubilizers, suspending agents, emulsifiers, isotonicity agents, stabilizers, soothing agents, preservatives, antioxidants, flavoring agents, colorants, buffers, and flow promoters, but are not limited to these, and other commonly used carriers and additives can be used as appropriate.
  • excipients examples include organic excipients such as sugars such as lactose, glucose, and D-mannitol, starches, and celluloses such as crystalline cellulose, and inorganic excipients such as calcium carbonate and kaolin.
  • Binders include pregelatinized starch, gelatin, gum arabic, methylcellulose, carboxymethylcellulose, sodium carboxymethylcellulose, crystalline cellulose, D-mannitol, trehalose, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, etc.
  • lubricant examples include stearic acid, fatty acid salts such as stearates, talc, silicates, and the like.
  • solvent examples include purified water, physiological saline, and phosphate buffer.
  • Disintegrants include low-substituted hydroxypropyl cellulose, chemically modified cellulose, and starches.
  • Solubilizing agents include polyethylene glycol, propylene glycol, trehalose, benzyl benzoate, ethanol, sodium carbonate, sodium citrate, sodium salicylate, and sodium acetate.
  • Suspending agents or emulsifying agents include sodium lauryl sulfate, gum arabic, gelatin, lecithin, glycerin monostearate, polyvinyl alcohol, polyvinylpyrrolidone, celluloses such as sodium carboxymethylcellulose, polysorbates, polyoxyethylene hydrogenated castor oil, etc.
  • Isotonicity agents include sodium chloride, potassium chloride, sugars, glycerin, urea, etc.
  • Stabilizers include polyethylene glycol, sodium dextran sulfate, and other amino acids.
  • Examples of the soothing agents include glucose, calcium gluconate, and procaine hydrochloride.
  • Preservatives include paraoxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, and sorbic acid.
  • Antioxidants include the water-soluble antioxidants ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, and sodium sulfite, the fat-soluble antioxidants ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, and ⁇ -tocopherol, and the metal chelating agents citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, and phosphoric acid.
  • BHA butylated hydroxyanisole
  • BHT butylated hydroxytoluene
  • lecithin propyl gallate
  • ⁇ -tocopherol and the metal chelating agents citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, and phosphoric acid.
  • Flavors include sweeteners and fragrances that are commonly used in the pharmaceutical field, and colorants include colorants that are commonly used in the pharmaceutical field.
  • the administration route of the pharmaceutical composition of the present invention is not particularly limited, and may be oral or parenteral.
  • parenteral administration include injection, nasal administration, pulmonary administration, and transdermal administration.
  • parenteral administration include intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, intramedullary injection, intrathecal injection, and intradermal injection.
  • the administration method can be appropriately selected depending on the age and symptoms of the patient.
  • compositions of the present invention are administered to a subject in a therapeutically effective amount.
  • a “therapeutically effective amount” refers to an amount of a therapeutic agent that is useful for alleviating a selected condition.
  • the therapeutically effective amount is determined as appropriate depending on the intended use, route of administration, age and symptoms of the patient, etc.
  • Tumors to which the pharmaceutical composition of the present invention can be applied include, for example, leukemias such as acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, malignant lymphomas such as Hodgkin's lymphoma and non-Hodgkin's lymphoma, multiple myeloma, myelodysplastic syndrome, head and neck cancer, tongue cancer, pharyngeal cancer, oral cancer, esophageal cancer, esophageal adenocarcinoma, gastric cancer, colorectal cancer, colon cancer, rectal cancer, liver cancer, gallbladder/bile duct cancer, biliary tract cancer, and pancreatic cancer.
  • leukemias such as acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia
  • malignant lymphomas such as Hodgkin'
  • lung cancer such as non-small cell lung cancer and small cell lung cancer
  • breast cancer ovarian cancer
  • cervical cancer uterine cancer
  • endometrial cancer vaginal cancer
  • vulvar cancer renal cancer
  • adenocarcinoma urothelial carcinoma
  • prostate cancer testicular tumor
  • bone and soft tissue sarcoma uveal malignant melanoma
  • malignant melanoma skin cancer
  • Merkel cell carcinoma malignant tumors
  • malignant tumors such as neuroblastoma, glioblastoma, brain tumor, and pleural mesothelioma
  • benign tumors such as angiomyolipoma, but are not limited to these.
  • the tumor is a malignant tumor such as colorectal cancer.
  • the pharmaceutical composition of the present invention may be used in combination with other drugs or treatment methods as long as the purpose of the present invention is not impaired.
  • Treatment Method also provides a method for treating a tumor, comprising administering to a subject a chymase inhibitor in combination with one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.
  • the present application also provides a method for enhancing the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibodies, and IL-12, comprising administering a chymase inhibitor to a subject to which any one or more selected from IL-18, anti-PD-1/PD-L1 antibodies, and IL-12 are administered or which has been administered to a subject.
  • the chymase inhibitor and one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 may be administered simultaneously or sequentially. There are no particular limitations on the administration route for each, and the method described in the previous section can be applied.
  • Example 1 Antitumor effect of combined use of peptidic chymase inhibitor and IL-18 Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration.
  • Example 2 Antitumor effect of a combination of a commercially available chymase inhibitor and IL-18 Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration.
  • mice were checked daily for mortality. Analysis method: Significance tests for survival curves of each group were performed using the Kaplan-Meier method, with a P value of 5% or less considered significant.
  • CT26 5 x 104 cells
  • mice were anesthetized with Isoflurane, and chilled PBS was injected intraperitoneally and collected (total 8 ml). The collected ascites was centrifuged and washed twice, and the number of cells was counted using a trypan blue exclusion assay test. The collected cells were applied to a chamber and subjected to HE and TB staining. Cell subsets were also analyzed by flow cytometry.
  • Mast cells showed a tendency to increase from 7 days after cell transplantation, but granular mast cells decreased (Fig. 4). Mast cells also tended to gather around the tumor tissue (data not shown). Tumor cell transplantation increased CD4 + T cells and CD8 + T cells, while the ratio of CD4 + T cells/CD8 + T cells decreased (Fig. 5). This is presumably due to an increase in CD8 + killer cells. NK cells, especially activated killer NK cells (DX5 + B220 + NK cells), also increased after tumor cell transplantation (Fig. 6).
  • NK cells increased early after transplantation up to day 10
  • CD8 + T cells increased from days 10 to 21, and mast cells gradually increased after day 7 (Figure 7).
  • Example 4 Changes in intraperitoneal antitumor lymphocytes by combined use of a peptide chymase inhibitor and IL-18
  • Colon cancer cells C26: 5 x 104 cells
  • the control group received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day.
  • the IL-18 alone administration group received 2 ⁇ g/body of IL-18 (see above mouse IL-18) intraperitoneally once every two days.
  • the chymase inhibitor alone administration group received 3mg/kg (100 ⁇ g/body) of Suc-Val-Pro-Phe p (OPh) 2 intraperitoneally once a day.
  • the IL-18/chymase inhibitor combination group received intraperitoneal administration of 2 ⁇ g/body of IL-18 once every two days and 3 mg/kg of chymase inhibitor once a day. Medication was terminated 28 days after cell transplantation.
  • ascites was collected from the mice 21 to 28 days after cell transplantation, and the cell subsets in the ascites were analyzed.
  • composition of immune cell subsets in the ascites also suggested that the combined use of IL-18 and chymase enhanced the antitumor effect compared to IL-18 alone.
  • Example 5 Antitumor effect of combination of chymase inhibitor and anti-PD-1 antibody
  • Colon cancer cells C26: 5 x 104 cells
  • the placebo group received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day.
  • the present invention is useful in treating tumors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to: a pharmaceutical composition which is for treating a tumor, and which includes a chymase inhibitor as an active ingredient, said pharmaceutical composition being characterized by being used in combination with at least one selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12); a pharmaceutical composition which is for treating a tumor, and which includes, as an active ingredient, at least one selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), said pharmaceutical composition being characterized by being used in combination with a chymase inhibitor; and the like.

Description

キマーゼ阻害剤と免疫療法の併用による抗腫瘍療法Antitumor therapy using chymase inhibitors in combination with immunotherapy

関連出願
 本出願は、日本特許出願2023-127528(2023年8月4日出願)に基づく優先権を主張しており、この内容は本明細書に参照として取り込まれる。
技術分野
 本発明は、キマーゼ阻害剤とインターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、インターロイキン-12(IL-12)などによる免疫療法を併用することを特徴とする、新規な抗腫瘍療法に関する。
RELATED APPLICATIONS This application claims priority based on Japanese Patent Application No. 2023-127528 (filed August 4, 2023), the contents of which are incorporated herein by reference.
TECHNICAL FIELD The present invention relates to a novel antitumor therapy characterized by combining a chymase inhibitor with immunotherapy using interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, interleukin-12 (IL-12), or the like.

 キマーゼは、主として肥満細胞に存在する中性のセリンプロテアーゼであり、ヒトキマーゼは、アンジオテンシンIを基質として、アンジオテンシンIIを産生することで、高血圧や動脈硬化症に関与することが知られている。これまで、キマーゼが、肺がん、大腸がん、膵臓がんの腫瘍組織周囲の血管新生と相関することを示した報告はあるが(非特許文献1~3)、キマーゼの腫瘍に対する直接効果を示した報告はない。 Chymase is a neutral serine protease found mainly in mast cells. Human chymase is known to be involved in hypertension and arteriosclerosis by producing angiotensin II using angiotensin I as a substrate. There have been reports showing that chymase correlates with angiogenesis around tumor tissues in lung cancer, colon cancer, and pancreatic cancer (Non-Patent Documents 1-3), but there have been no reports showing the direct effect of chymase on tumors.

 インターロイキン-18(IL-18)は、インターフェロンγ(IFNγ)産生誘導因子として岡村らにより発見されたサイトカインである。IL-18は、IL-12の存在下でIFNγの産生が誘導されている細胞の細胞表面上に発現しているIL-18受容体に結合し、IFNγの産生を亢進し、Th1型の免疫応答を増強する。一方、IL-12の不存在下では、IL-18はTh2型の免疫応答を増強する。 Interleukin-18 (IL-18) is a cytokine discovered by Okamura et al. as an interferon-gamma (IFNγ) production inducer. IL-18 binds to the IL-18 receptor expressed on the cell surface of cells in which IFNγ production is induced in the presence of IL-12, enhancing IFNγ production and enhancing Th1-type immune responses. On the other hand, in the absence of IL-12, IL-18 enhances Th2-type immune responses.

 IL-18には抗腫瘍効果が期待されており、IL-18と、リツキシマブ又はハーセプチンとの併用や(特許文献1)、IL-18と分子標的抗体との併用によるがん治療薬が報告されている(特許文献2)。最近では、IL-18を分泌するCAR-T細胞を用いて、腫瘍特異的にIL-18を分泌させることで強力な抗腫瘍効果が得られたことが報告されている(非特許文献4)。 IL-18 is expected to have an antitumor effect, and cancer treatment drugs that combine IL-18 with rituximab or Herceptin (Patent Document 1) and with a molecular target antibody have been reported (Patent Document 2). Recently, it has been reported that a strong antitumor effect was achieved by using CAR-T cells that secrete IL-18 to secrete IL-18 in a tumor-specific manner (Non-Patent Document 4).

 IL-18は、プロペプチドを含むIL-18前駆体として産生され、カスパーゼなどによる切断を受けて活性型IL-18として細胞外に放出される。カスパーゼに代えて、IL-18前駆体をキマーゼで切断することを特徴とするIL-18の生産方法が報告されている(特許文献3)。この報告は、活性型(成熟型)IL-18に対するキマーゼ阻害剤の作用を示すものではなく、またIL-18前駆体に対しては、キマーゼ阻害剤は活性型への変換を阻害することで、IL-18活性を阻害し得ることを示唆するものである。 IL-18 is produced as an IL-18 precursor containing a propeptide, which is cleaved by caspases and released outside the cell as active IL-18. A method for producing IL-18 has been reported in which the IL-18 precursor is cleaved by chymase instead of caspase (Patent Document 3). This report does not show the effect of chymase inhibitors on active (mature) IL-18, but suggests that chymase inhibitors can inhibit IL-18 activity by inhibiting the conversion of the IL-18 precursor to the active form.

 PD-1やPD-L1は、代表的な免疫チェックポイント分子であり、この分子を標的とする抗体は、様々ながん種において、顕著な抗がん作用を示すことが確認され、現在医薬として開発されている。しかし、抗PD-1/PD-L1抗体療法については、固形がんでの寛解率は低く、その奏効率は十分なものとは言えない。 PD-1 and PD-L1 are representative immune checkpoint molecules, and antibodies targeting these molecules have been shown to have significant anti-cancer effects in a variety of cancer types, and are currently being developed as medicines. However, the remission rate for solid cancers with anti-PD-1/PD-L1 antibody therapy is low, and the response rate cannot be said to be sufficient.

WO2006/116423WO2006/116423 WO2016/021720WO2016/021720 特開2007-151414JP2007-151414

Ibaraki et al., The relationship of tryptase- and chymase-positive mast cells to angiogenesis in stage I non-small cell lung cancer. Eur J Cardiothorac Surg. 2005;28(4):617-621.Ibaraki et al., The relationship of tryptase- and chymase-positive mast cells to angiogenesis in stage I non-small cell lung cancer. Eur J Cardiothorac Surg. 2005;28(4):617-621. Kondo et al., Expression of chymase-positive cells in gastric cancer and its correlation with the angiogenesis. J Surg Oncol. 2006;93(1):36-43.Kondo et al., Expression of chymase-positive cells in gastric cancer and its correlation with the angiogenesis. J Surg Oncol. 2006;93(1):36-43. Laface et al., Chymase-positive mast cells correlate with tumor angiogenesis: first report in pancreatic cancer patients. Eur Rev Med Pharmacol Sci. 2021;25(22):6862-6873.Laface et al., Chymase-positive mast cells correlate with tumor angiogenesis: first report in pancreatic cancer patients. Eur Rev Med Pharmacol Sci. 2021;25(22):6862-6873. Svoboda et al., Interleukin-18 Secreting Autologous Anti-CD19 CAR T-Cells (huCART19-IL18) in Patients with Non-Hodgkin Lymphomas Relapsed or Refractory to Prior CAR T-Cell Therapy. Blood (2022) 140 (Supplement 1): 4612-4614Svoboda et al., Interleukin-18 Secreting Autologous Anti-CD19 CAR T-Cells (huCART19-IL18) in Patients with Non- Hodgkin Lymphomas Relapsed or Refractory to Prior CAR T-Cell Therapy. Blood (2022) 140 (Supplement 1): 4612-4614

 本発明の課題は、新規な腫瘍の治療方法を提供することにある。 The objective of the present invention is to provide a novel method for treating tumors.

 発明者らは、IL-18や抗PD-1/PD-L1抗体(抗PD-1抗体又は抗PD-L1抗体)等による免疫療法に、単独では抗腫瘍効果を有しないキマーゼ阻害剤を併用することにより、前記免疫療法による抗腫瘍効果が増強されることを見出した。本願は、前記知見に基づくものであり、第1の態様として、以下の[1]~[8]を提供する。
[1] キマーゼ阻害剤を有効成分として含む、腫瘍を治療するための医薬組成物であって、インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と併用されることを特徴とする、前記医薬組成物。
[2]インターロイキン-18(IL-18)、PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と、キマーゼ阻害剤とを組み合わせて含む、腫瘍を治療するための医薬組成物。
[3]インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上を有効成分として含む、腫瘍を治療するための医薬組成物であって、キマーゼ阻害剤と併用されることを特徴とする、前記医薬組成物。
[4]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、及び(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide)、SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、Y-40079、ASB17061、ONO-8020435、及びONO-2220445から選択されるいずれかである、[1]~[3]のいずれかに記載の医薬組成物。
[5]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上がヒト由来である、[1]~[4]のいずれかに記載の医薬組成物。
[6]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とキマーゼ阻害剤同時に又は順次に投与される、[1]~[5]のいずれかに記載の医薬組成物。
[7]キマーゼ阻害剤を有効成分として含む、腫瘍の治療においてインターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上の作用を増強するための医薬組成物。
[8]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide) SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、ASB17061、ONO-8020435、及びONO-2220445から選択されるいずれかである、[7]に記載の医薬組成物。
The inventors have found that the antitumor effect of immunotherapy using IL-18, anti-PD-1/PD-L1 antibody (anti-PD-1 antibody or anti-PD-L1 antibody), or the like can be enhanced by combining the immunotherapy with a chymase inhibitor that does not have an antitumor effect by itself. The present application is based on the above findings, and provides the following [1] to [8] as a first aspect.
[1] A pharmaceutical composition for treating a tumor, comprising a chymase inhibitor as an active ingredient, in combination with one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
[2] A pharmaceutical composition for treating a tumor, comprising a combination of one or more selected from interleukin-18 (IL-18), PD-1/PD-L1 antibody, and interleukin-12 (IL-12) and a chymase inhibitor.
[3] A pharmaceutical composition for treating a tumor, comprising as an active ingredient any one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), wherein the pharmaceutical composition is used in combination with a chymase inhibitor.
[4] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide), SUN-C8257, SUN13834, BCEAB, Compound The pharmaceutical composition according to any one of [1] to [3], which is selected from the group consisting of 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), Y-40079, ASB17061, ONO-8020435, and ONO-2220445.
[5] The pharmaceutical composition according to any one of [1] to [4], wherein one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
[6] The pharmaceutical composition according to any one of [1] to [5], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
[7] A pharmaceutical composition for enhancing the effect of any one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and IL-12 in tumor treatment, comprising a chymase inhibitor as an active ingredient.
[8] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-am ino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide) The pharmaceutical composition according to [7], which is any one selected from SUN-C8257, SUN13834, BCEAB, Compound 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), ASB17061, ONO-8020435, and ONO-2220445.

 本願は、第2の態様として、以下の[1]~[6]を提供する。
[1]キマーゼ阻害剤とインターロイキン-18(IL-18)、抗PD-1/PD-L1抗体及びインターロイキン-12(IL-12)から選ばれるいずれか1以上を組み合わせて対象に投与することを特徴とする、腫瘍を治療する方法。
[2]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide) SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、ASB17061、ONO-8020435、ONO-2220445から選択されるいずれかである、[1]に記載の方法。
[3]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とキマーゼ阻害剤が同時に投与される、[1]に記載の方法。
[4]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とキマーゼ阻害剤が順次に投与される、[1]に記載の方法。
[5]インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体及びインターロイキン-12(IL-12)から選ばれるいずれか1以上の抗腫瘍効果を増強するための方法であって、IL-18、抗PD-1/PD-L1抗体、及びIL-12を投与する又は投与された対象に、キマーゼ阻害剤を投与することを含む、方法。
[6]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide)、SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、ASB17061、ONO-8020435、及びONO-2220445から選択されるいずれかである、[5]に記載の方法。
As a second aspect, the present application provides the following [1] to [6].
[1] A method for treating a tumor, comprising administering to a subject a combination of a chymase inhibitor and one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
[2] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-am ino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide) The method according to [1], wherein the compound is any one selected from SUN-C8257, SUN13834, BCEAB, Compound 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), ASB17061, ONO-8020435, and ONO-2220445.
[3] The method according to [1], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 are administered simultaneously with a chymase inhibitor.
[4] The method according to [1], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered sequentially.
[5] A method for enhancing the anti-tumor effect of any one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), comprising administering a chymase inhibitor to a subject administered or who has been administered IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.
[6] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, (2-[5-amino-2-(4-fluoroph enyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide), SUN-C8257, SUN13834, BCEAB, Compound 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), ASB17061, ONO-8020435, and ONO-2220445.

 本願は、第3の態様として、以下の[1]~[7]を提供する。
[1]キマーゼ阻害剤を含む、腫瘍の治療における使用のための薬剤(agent)であって、インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と併用されることを特徴とする、前記薬剤。
[2]インターロイキン-18(IL-18)、PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と、キマーゼ阻害剤とを組み合わせて含む、腫瘍の治療における使用のための薬剤。
[3]インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上を含む、腫瘍の治療における使用のための薬剤であって、キマーゼ阻害剤と併用されることを特徴とする、前記薬剤。
[4]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、及び(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide)、SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、Y-40079、ASB17061、ONO-8020435、及びONO-2220445から選択されるいずれかである、[1]~[3]のいずれかに記載の使用のための薬剤。
[5]IL-18、抗PD-1/PD-L1抗体、及びIL-12がヒト由来である、[1]~[4]のいずれかに記載の使用のための組成物。
[6]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とキマーゼ阻害剤が同時に又は順次に投与される、[1]~[5]のいずれかに記載の使用のための薬剤。
[7]キマーゼ阻害剤が、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上の抗腫瘍作用を増強する、[1]~[6]のいずれかに記載の使用のための薬剤。
As a third aspect, the present application provides the following [1] to [7].
[1] An agent for use in treating a tumor, comprising a chymase inhibitor, wherein the agent is used in combination with any one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
[2] A drug for use in treating a tumor, comprising a combination of one or more selected from interleukin-18 (IL-18), PD-1/PD-L1 antibody, and interleukin-12 (IL-12) and a chymase inhibitor.
[3] A drug for use in treating a tumor, comprising any one or more selected from interleukin-18 (IL-18), an anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), characterized in that the drug is used in combination with a chymase inhibitor.
[4] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide), SUN-C8257, SUN13834, BCEAB, Compound 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), Y-40079, ASB17061, ONO-8020435, and ONO-2220445.
[5] The composition for use according to any one of [1] to [4], wherein IL-18, the anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
[6] The drug for use according to any one of [1] to [5], wherein one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
[7] The agent for use according to any of [1] to [6], wherein the chymase inhibitor enhances the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12.

 本願は、第4の態様として、以下の[1]~[7]を提供する。
[1]腫瘍を治療するための医薬の製造における、インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と、キマーゼ阻害剤の使用。
[2]腫瘍を治療するための医薬の製造における、キマーゼ阻害剤の使用であって、前記医薬がインターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と併用されることを特徴とする、前記使用。
[3]腫瘍を治療するための医薬の製造における、インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上の使用であって、前記医薬がキマーゼ阻害剤と併用されることを特徴とする、前記使用。
[4]キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、及び(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide)、SUN-C8257、SUN13834、BCEAB、Compound 17、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、Y-40079、ASB17061、ONO-8020435、及びONO-2220445から選択されるいずれかである、[1]~[3]のいずれかに記載の使用。
[5]IL-18、抗PD-1/PD-L1抗体、及びIL-12がヒト由来である、[1]~[4]のいずれかに記載の使用。
[6]IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とキマーゼ阻害剤が同時に又は順次に投与される、[1]~[5]のいずれかに記載の使用。
[7]キマーゼ阻害剤が、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上の抗腫瘍作用を増強する、[1]~[6]のいずれかに記載の使用。
As a fourth aspect, the present application provides the following [1] to [7].
[1] Use of one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), together with a chymase inhibitor, in the manufacture of a medicine for treating a tumor.
[2] Use of a chymase inhibitor in the manufacture of a medicament for treating a tumor, characterized in that the medicament is used in combination with one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).
[3] Use of one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12) in the manufacture of a medicament for treating a tumor, characterized in that the medicament is used in combination with a chymase inhibitor.
[4] Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid, N-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide, and (2-[5-amino-2-(4-fluorop henyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide), SUN-C8257, SUN13834, BCEAB, Compound The use according to any one of [1] to [3], wherein the compound is selected from the group consisting of 17, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), Y-40079, ASB17061, ONO-8020435, and ONO-2220445.
[5] The use according to any one of [1] to [4], wherein IL-18, the anti-PD-1/PD-L1 antibody, and IL-12 are of human origin.
[6] The use according to any one of [1] to [5], wherein one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 and a chymase inhibitor are administered simultaneously or sequentially.
[7] The use according to any one of [1] to [6], wherein the chymase inhibitor enhances the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12.

 本発明により、キマーゼ阻害剤と、IL-18、抗PD-1/PD-L1抗体、L-12等の免疫療法剤の新規な臨床用途が提供される。 The present invention provides novel clinical applications of chymase inhibitors and immunotherapeutic agents such as IL-18, anti-PD-1/PD-L1 antibodies, and L-12.

図1は、腫瘍(CT26)細胞移植後100日時点での、プラセボ群、IL-18投与群、キマーゼ阻害剤投与群、IL-18・キマーゼ阻害剤併用群の生存曲線を示す。FIG. 1 shows survival curves for the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group 100 days after tumor (CT26) cell transplantation. 図2は、腫瘍(CT26)細胞移植後100日時点で生存していたマウスの腫瘍移植部分の写真である(上:IL-18、下:IL-18及びキマーゼ阻害剤併用)。FIG. 2 shows photographs of the tumor transplant site of a mouse that survived 100 days after tumor (CT26) cell transplantation (top: IL-18, bottom: combined use of IL-18 and chymase inhibitor). 図3は、腫瘍(CT26)細胞移植後110日時点での、プラセボ群、IL-18投与群、キマーゼ阻害剤投与群、IL-18・キマーゼ阻害剤併用群の生存曲線を示す。FIG. 3 shows survival curves for the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group, 110 days after tumor (CT26) cell transplantation. 図4は、腫瘍(CT26)細胞移植後の腹水中のマスト細胞の割合(A)、絶対数(B)、細胞サイズ(C)、細胞内顆粒の大きさ(D)を示す。FIG. 4 shows the percentage (A), absolute number (B), cell size (C), and size of intracellular granules (D) of mast cells in ascites after tumor (CT26) cell transplantation. 図5は、腫瘍(CT26)細胞移植後のCD4+T細胞数(A)、CD8+T細胞数(B)、CD4+T細胞/CD8+T細胞の割合(C)を示す。FIG. 5 shows the number of CD4 + T cells (A), the number of CD8 + T cells (B), and the ratio of CD4 + T cells/CD8 + T cells (C) after tumor (CT26) cell transplantation. 図6は、腫瘍(CT26)細胞移植後の腹水中のNK細胞の割合(A)、NK細胞の絶対数(B)、DX5+B220+NK細胞の割合(C)を示す。FIG. 6 shows the percentage of NK cells in ascites after tumor (CT26) cell transplantation (A), the absolute number of NK cells (B), and the percentage of DX5 + B220 + NK cells (C). 図7は、腫瘍(CT26)細胞移植後の腹水中の免疫細胞(マスト細胞、CD8+T細胞、NK細胞、DX5+B220+NK細胞の数の推移を示す。FIG. 7 shows the change in the numbers of immune cells (mast cells, CD8 + T cells, NK cells, and DX5 + B220 + NK cells) in the ascites after tumor (CT26) cell transplantation. 図8は、プラセボ群、IL-18投与群、キマーゼ阻害剤投与群、IL-18・キマーゼ阻害剤併用群において腹水中のT細胞を比較した結果を示す。CD4+T細胞/CD8+T細胞の割合(A)、CD4+T細胞数(B)、CD8+T細胞数(C)、CD44high CD8+T細胞数(D)Figure 8 shows the results of comparing T cells in ascites in the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combination group. The ratio of CD4 + T cells/CD8 + T cells (A), CD4 + T cell count (B), CD8 + T cell count (C), and CD44high CD8 + T cell count (D). 図9はプラセボ群、IL-18投与群、キマーゼ阻害剤投与群、IL-18・キマーゼ阻害剤併用群において、腹水中のNK細胞数を比較した結果を示す。DX5+NK細胞数(A)、DX5+B220+NK細胞数(B)Figure 9 shows the results of comparing the number of NK cells in ascites in the placebo group, IL-18 administration group, chymase inhibitor administration group, and IL-18/chymase inhibitor combined administration group. DX5 + NK cell count (A), DX5 + B220 + NK cell count (B) 図10は、腫瘍(CT26)細胞移植後34日時点での、プラセボ群、抗PD-1抗体投与群、キマーゼ阻害剤投与群、抗PD-1抗体・キマーゼ阻害剤併用群の生存曲線を示す。Figure 10 shows survival curves for the placebo group, the anti-PD-1 antibody administration group, the chymase inhibitor administration group, and the anti-PD-1 antibody/chymase inhibitor combination group 34 days after tumor (CT26) cell transplantation.

1. キマーゼ阻害剤
 本明細書において、キマーゼ阻害剤とは、キマーゼの活性を阻害できる薬剤を意味する。キマーゼとは、中性のセリンプロテアーゼであり、ヒトキマーゼは、アンジオテンシンIからアンジオテンシンIIを産生することで、高血圧や動脈硬化症に関与することが知られている。本発明のキマーゼ阻害剤は、好ましくはヒトキマーゼ阻害剤である。
1. Chymase Inhibitor In this specification, a chymase inhibitor means a drug capable of inhibiting the activity of chymase. Chymase is a neutral serine protease, and human chymase is known to be involved in hypertension and arteriosclerosis by producing angiotensin II from angiotensin I. The chymase inhibitor of the present invention is preferably a human chymase inhibitor.

 キマーゼ阻害剤としては、公知のキマーゼ阻害剤を使用することができる。公知のキマーゼ阻害剤としては、例えば、既に臨床適用や前臨床モデルでの検証が報告されている化合物:SUN-C8257、SUN13834、BCEAB、Compound 17、NK3201、TEI-E548、RO5066852、JNJ-10311795(RWJ-355871)、Suc-Val-Pro-PheP(OPh)2、Y-40613、Y-40079、TY-51469、Chymostatin、Fulacimstat(BAY1142524)、ASB17061、ONO-8020435、ONO-2220445等を挙げることができる(Heuston and Hyland, Br J Pharmacol. 2012 Oct; 167(4): 732-740.doi: 10.1111/j.1476-5381.2012.02055.x等参照)。 As the chymase inhibitor, a known chymase inhibitor can be used. Examples of known chymase inhibitors include compounds that have already been reported for clinical application or validation in preclinical models, such as SUN-C8257, SUN13834, BCEAB, Compound 17, NK3201, TEI-E548, RO5066852, JNJ-10311795 (RWJ-355871), Suc-Val-Pro-Phe P (OPh) 2 , Y-40613, Y-40079, TY-51469, Chymostatin, Fulacimstat (BAY1142524), ASB17061, ONO-8020435, and ONO-2220445 (Heuston and Hyland, Br J Pharmacol. 2012 Oct; 167(4): 732-740.doi: 10.1111/j.1476-5381.2012.02055.x etc.)

 あるいは、WO2016/132483、特開2016-150915、特表2016-535095、特表2012-516335、特表2012-502034、特開2012-121898、特表2010-536768、WO2010/147133、特表2010-515699、特表2010-505900、WO2005/018672、WO03/018061、特表2004-507539、WO01/062294、WO01/062293、WO01/062292、WO00/005204、WO99/032459、特開平11-001479、特開平10-087493に開示されているキマーゼ阻害剤を使用することもできる。 Or, WO2016/132483, JP 2016-150915, JP 2016-535095, JP 2012-516335, JP 2012-502034, JP 2012-121898, JP 2010-536768, WO2010/147133, JP 2010-515699, JP 2010-505900, WO Chymase inhibitors disclosed in WO 2005/018672, WO03/018061, JP 2004-507539, WO01/062294, WO01/062293, WO01/062292, WO00/005204, WO99/032459, JP 11-001479, and JP 10-087493 can also be used.

Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-I000002
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-I000002

2.インターロイキン-18(IL-18)
 インターロイキン-18(IL-18)は、IFN-γ産生誘導因子として発見されたサイトカインである。IL-18は、IL-18はIL-12とともにCD4CD8T細胞及びNK細胞に作用してIFN-γの産生を誘導する。
2. Interleukin-18 (IL-18)
Interleukin-18 (IL-18) is a cytokine that was discovered as an IFN-γ production inducer. IL-18, together with IL-12, acts on CD4 + CD8 + T cells and NK cells to induce IFN-γ production.

 IL-18は、プロペプチドを含む不活性な前駆体(pro-IL-18)として産生され、カスパーゼ1やカスパーゼ4などによるプロペプチドの切断を受けて、活性型IL-18として放出される。本発明において、IL-18の起源は特に限定されないが、投与される対象と同種又は近縁種由来のIL-18であることが好ましい。したがって、ヒトに投与される場合には、哺乳類、とくにヒトIL-18が好ましい。 IL-18 is produced as an inactive precursor (pro-IL-18) containing the propeptide, and is released as active IL-18 following cleavage of the propeptide by caspase 1, caspase 4, etc. In the present invention, the source of IL-18 is not particularly limited, but it is preferable that the IL-18 is derived from the same species as the subject to be administered, or a closely related species. Therefore, when administered to humans, mammalian IL-18, particularly human IL-18, is preferred.

 ヒトIL-18前駆体は193アミノ酸(24KDa)からなり、プロペプチドを含まない成熟型(活性型)のヒトIL-18は157アミノ酸(18KDa)からなる。ヒトIL-18のアミノ酸配列は公知であり、例えば、ヒトIL-18のアミノ酸配列としては、配列番号1に示されるアミノ酸配列を挙げることができる。このアミノ酸配列と80%、85%、90%、95%、96%、98%、又は99%以上の配列同一性を有するアミノ酸配列を有するヒトIL-18もまた、IL-18の活性を有する限り、本発明のIL-18として使用することができる。IL-18活性とは、IFNγ産生誘導活性などIL-18が有する生理活性を意味する。 Human IL-18 precursor consists of 193 amino acids (24 KDa), and mature (active) human IL-18 without the propeptide consists of 157 amino acids (18 KDa). The amino acid sequence of human IL-18 is known, and an example of the amino acid sequence of human IL-18 is the amino acid sequence shown in SEQ ID NO: 1. Human IL-18 having an amino acid sequence that has 80%, 85%, 90%, 95%, 96%, 98%, or 99% or more sequence identity with this amino acid sequence can also be used as the IL-18 of the present invention as long as it has the activity of IL-18. IL-18 activity means the physiological activity of IL-18, such as IFNγ production-inducing activity.

 ヒトIL-18の一例としては、例えば、以下の(1)又は(2)のタンパク質を挙げることができる:
(1) 配列番号1に示されるアミノ酸配列を含むタンパク質、又は
(2) 配列番号1に示されるアミノ酸配列と80%以上、好ましくは90%以上、より好ましくは95%以上の配列同一性を有し、IL-18活性(IFNγ産生誘導活性など)を有するタンパク質を挙げることができる。
An example of human IL-18 is the following protein (1) or (2):
(1) A protein comprising the amino acid sequence shown in SEQ ID NO:1, or
(2) A protein having 80% or more, preferably 90% or more, and more preferably 95% or more sequence identity with the amino acid sequence shown in SEQ ID NO:1 and having IL-18 activity (such as IFNγ production-inducing activity).

 IL-18は細胞膜上の受容体に結合することで、その生理活性を発揮するが、生体内にはIL-18に特異的なIL-18結合タンパク質が過剰に存在するため、分泌されたIL-18の多くはIL-18結合タンパク質と結合して活性を失う。最近では、IL-18の安定性を高めた改変体や、Car-T細胞でIL-18を分泌させることで、IL-18の抗腫瘍効果が増強されることが報告されている。こうしたIL-18の改変体や新たな送達システムも、本発明の目的に反しない限り、本発明の範囲内にある。 IL-18 exerts its physiological activity by binding to a receptor on the cell membrane, but since there is an excess of IL-18-specific IL-18-binding proteins in the body, much of the secreted IL-18 loses its activity upon binding to the IL-18-binding proteins. Recently, it has been reported that the antitumor effect of IL-18 is enhanced by using modified IL-18 with increased stability and by secreting IL-18 from Car-T cells. Such modified IL-18 and new delivery systems are also within the scope of the present invention, so long as they do not contradict the objectives of the present invention.

3.抗PD-1/PD-L1抗体(抗PD-1抗体、抗PD-L1抗体)
 本発明において、「抗PD-1/PD-L1抗体」とは、PD-1又はPD-L1を特異的に阻害する抗体、抗PD-1抗体又は抗PD-L1抗体を意味する。
3. Anti-PD-1/PD-L1 antibody (anti-PD-1 antibody, anti-PD-L1 antibody)
In the present invention, the term "anti-PD-1/PD-L1 antibody" refers to an antibody that specifically inhibits PD-1 or PD-L1, an anti-PD-1 antibody or an anti-PD-L1 antibody.

 本発明で使用される「抗PD-1抗体」は、PD-1に結合し、その免疫チェックポイントとしての機能を阻害できるものであれば特に限定されない。抗PD-1抗体としては、既に医薬として承認販売されているものや、開発中のものがあり、これらを好適に利用することができる。そのような「抗PD-1抗体」としては、ニボルマブ(オプジーボ(GSK/小野薬品))、ヒト化IgG4型抗体であるペムブロリズマブ(Merck)、ジピリズマブが挙げられるが、これらに限定されない。 The "anti-PD-1 antibody" used in the present invention is not particularly limited as long as it can bind to PD-1 and inhibit its function as an immune checkpoint. Anti-PD-1 antibodies include those that have already been approved and sold as medicines and those that are under development, and these can be used preferably. Examples of such "anti-PD-1 antibodies" include nivolumab (Opdivo (GSK/Ono Pharmaceutical)), pembrolizumab (Merck), which is a humanized IgG4 antibody, and dipirizumab, but are not limited to these.

 本発明で使用される「抗PD-L1抗体」は、PD-L1に結合し、その免疫チェックポイントとしての機能を阻害できるものであれば特に限定されない。PD-L1抗体としては、既に医薬として承認(販売)されているものや、開発中のものがあり、これらを好適に利用することができる。そのような「抗PD-L1抗体」としては、アテゾリズマブ(Roche/中外)、デュルバルマブ(アストラゼネカ)、アベルマブ(Merck)、MED10680/AMP-514が挙げられるが、これらに限定されない。 The "anti-PD-L1 antibody" used in the present invention is not particularly limited as long as it can bind to PD-L1 and inhibit its function as an immune checkpoint. PD-L1 antibodies include those that have already been approved (sold) as medicines and those that are under development, and these can be used preferably. Examples of such "anti-PD-L1 antibodies" include, but are not limited to, atezolizumab (Roche/Chugai), durvalumab (AstraZeneca), avelumab (Merck), and MED10680/AMP-514.

 抗体は、常法に基づいて作製してもよい。その場合、抗体は、ヒトに対する異種抗原性を低下させるために、キメラ(Chimeric)抗体、ヒト化(Humanized)抗体であるか、あるいは完全ヒト抗体であることが好ましい。抗体は免疫チェックポイント阻害剤としての機能を有する限り、抗体断片でもよい。抗体断片としては、F(Ab')2、Fab'、Fab、Fv、scFvを挙げることができる。 The antibody may be produced according to a conventional method. In this case, the antibody is preferably a chimeric antibody, a humanized antibody, or a fully human antibody in order to reduce heterologous antigenicity to humans. The antibody may be an antibody fragment so long as it has a function as an immune checkpoint inhibitor. Examples of the antibody fragment include F(Ab') 2 , Fab', Fab, Fv, and scFv.

4.インターロイキン-12(IL-12)
 インターロイキン-12(IL-12)は、主としてマクロファージ、多核白血球、樹状細胞などで産生されるサイトカインで、NK細胞やT細胞においてIFNγの産生を誘導することや、NK細胞や細胞障害性T細胞の活性を増強することが知られている。
4. Interleukin-12 (IL-12)
Interleukin-12 (IL-12) is a cytokine produced mainly by macrophages, polymorphonuclear leukocytes, dendritic cells, etc., and is known to induce the production of IFN-γ in NK cells and T cells, and to enhance the activity of NK cells and cytotoxic T cells.

 IL-12は、Th1細胞を優先的に活性化させ、Th2細胞の抑制による細胞性免疫の活性化作用があることが報告されており、そのため、Th2主導型免疫異常の治療や、細胞性免疫機能の賦活によるがんや感染症治療への応用が期待されている。前述のように、IL-12は、IL-18ともにCD4CD8T細胞及びNK細胞に作用してIFN-γの産生を誘導する。したがって、がんや感染症の治療において、IL-12とIL-18を併用してもよい。 It has been reported that IL-12 preferentially activates Th1 cells and activates cellular immunity by suppressing Th2 cells, and is therefore expected to be used in the treatment of Th2-driven immune disorders and in the treatment of cancer and infectious diseases by activating cellular immune function. As mentioned above, IL-12, along with IL-18, acts on CD4 + CD8 + T cells and NK cells to induce the production of IFN-γ. Therefore, IL-12 and IL-18 may be used in combination in the treatment of cancer and infectious diseases.

5.医薬組成物(医薬、薬剤)
 本発明の医薬組成物は、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上に、単独では抗腫瘍活性を示さないキマーゼ阻害剤を組合せることで、腫瘍を効果的に治療することを特徴とする。
5. Pharmaceutical compositions (medicines, drugs)
The pharmaceutical composition of the present invention is characterized by effectively treating tumors by combining one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 with a chymase inhibitor that does not exhibit anti-tumor activity alone.

 第1の実施形態において、本発明の医薬組成物は、キマーゼ阻害剤を有効成分として含む、腫瘍を治療するための医薬組成物であって、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上と併用されることを特徴とする医薬組成物である。 In a first embodiment, the pharmaceutical composition of the present invention is a pharmaceutical composition for treating a tumor, comprising a chymase inhibitor as an active ingredient, and characterized in that it is used in combination with one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.

 第2の実施形態において、本発明の医薬組成物は、キマーゼ阻害剤と、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上とを組み合わせて含む、腫瘍を治療するための医薬組成物である。この場合、キマーゼ阻害剤とIL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上はいっしょに製剤化されていてもよいし、別々に製剤化されたキット製剤であってもよい。 In a second embodiment, the pharmaceutical composition of the present invention is a pharmaceutical composition for treating a tumor, comprising a combination of a chymase inhibitor and one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12. In this case, the chymase inhibitor and one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12 may be formulated together, or may be formulated separately as a kit formulation.

 第3の実施形態において、本発明の医薬組成物は、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上を有効成分として含む、腫瘍を治療するための医薬組成物であって、キマーゼ阻害剤と併用されることを特徴とする医薬組成物である。 In a third embodiment, the pharmaceutical composition of the present invention is a pharmaceutical composition for treating tumors, comprising one or more active ingredients selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12, and is characterized in that it is used in combination with a chymase inhibitor.

 第4の実施形態において、本発明の医薬組成物は、キマーゼ阻害剤を有効成分として含む、腫瘍の治療においてIL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上の作用を増強するための医薬組成物である。 In a fourth embodiment, the pharmaceutical composition of the present invention is a pharmaceutical composition for enhancing the action of one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 in tumor treatment, comprising a chymase inhibitor as an active ingredient.

 本発明の医薬組成物は、薬理学上許容される担体や添加物を含んでいてもよい。そのような担体や添加物としては、例えば、賦形剤、結合剤、滑沢剤、溶剤、崩壊剤、溶解補助剤、懸濁化剤、乳化剤、等張化剤、安定化剤、無痛化剤、防腐剤、抗酸化剤、矯味剤、着色剤、緩衝剤、流動性促進剤等が挙げられるが、これらに制限されず、その他常用の担体や添加物が適宜使用できる。 The pharmaceutical composition of the present invention may contain pharmacologically acceptable carriers and additives. Examples of such carriers and additives include excipients, binders, lubricants, solvents, disintegrants, solubilizers, suspending agents, emulsifiers, isotonicity agents, stabilizers, soothing agents, preservatives, antioxidants, flavoring agents, colorants, buffers, and flow promoters, but are not limited to these, and other commonly used carriers and additives can be used as appropriate.

 賦形剤としては、乳糖、ブドウ糖、D-マンニトールなどの糖類、でんぷん類、結晶セルロースなどのセルロース類などの有機系賦形剤、炭酸カルシウム、カオリンなどの無機系賦形剤などが挙げられる。 Examples of excipients include organic excipients such as sugars such as lactose, glucose, and D-mannitol, starches, and celluloses such as crystalline cellulose, and inorganic excipients such as calcium carbonate and kaolin.

 結合剤としては、α化デンプン、ゼラチン、アラビアゴム、メチルセルロース、カルボキシメチルセルロース、カルボキシメチルセルロースナトリウム、結晶セルロース、D-マンニトール、トレハロース、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルピロリドン、ポリビニルアルコールなどが挙げられる。 Binders include pregelatinized starch, gelatin, gum arabic, methylcellulose, carboxymethylcellulose, sodium carboxymethylcellulose, crystalline cellulose, D-mannitol, trehalose, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylpyrrolidone, polyvinyl alcohol, etc.

 滑沢剤としては、ステアリン酸、ステアリン酸塩などの脂肪酸塩、タルク、珪酸塩類などが挙げられる。
 溶剤としては、精製水、生理的食塩水、リン酸緩衝液などが挙げられる。
Examples of the lubricant include stearic acid, fatty acid salts such as stearates, talc, silicates, and the like.
Examples of the solvent include purified water, physiological saline, and phosphate buffer.

 崩壊剤としては、低置換度ヒドロキシプロピルセルロース、化学修飾されたセルロースやデンプン類などが挙げられる。 Disintegrants include low-substituted hydroxypropyl cellulose, chemically modified cellulose, and starches.

 溶解補助剤としては、ポリエチレングリコール、プロピレングリコール、トレハロース、安息香酸ベンジル、エタノール、炭酸ナトリウム、クエン酸ナトリウム、サリチル酸ナトリウム、酢酸ナトリウムなどが挙げられる。 Solubilizing agents include polyethylene glycol, propylene glycol, trehalose, benzyl benzoate, ethanol, sodium carbonate, sodium citrate, sodium salicylate, and sodium acetate.

 懸濁化剤あるいは乳化剤としては、ラウリル硫酸ナトリウム、アラビアゴム、ゼラチン、レシチン、モノステアリン酸グリセリン、ポリビニルアルコール、ポリビニルピロリドン、カルボキシメチルセルロースナトリウムなどのセルロース類、ポリソルベート類、ポリオキシエチレン硬化ヒマシ油などが挙げられる。 Suspending agents or emulsifying agents include sodium lauryl sulfate, gum arabic, gelatin, lecithin, glycerin monostearate, polyvinyl alcohol, polyvinylpyrrolidone, celluloses such as sodium carboxymethylcellulose, polysorbates, polyoxyethylene hydrogenated castor oil, etc.

 等張化剤としては、塩化ナトリウム、塩化カリウム、糖類、グリセリン、尿素などが挙げられる。 Isotonicity agents include sodium chloride, potassium chloride, sugars, glycerin, urea, etc.

 安定化剤としては、ポリエチレングリコール、デキストラン硫酸ナトリウム、その他のアミノ酸類などが挙げられる。
 無痛化剤としては、ブドウ糖、グルコン酸カルシウム、塩酸プロカインなどが挙げられる。
Stabilizers include polyethylene glycol, sodium dextran sulfate, and other amino acids.
Examples of the soothing agents include glucose, calcium gluconate, and procaine hydrochloride.

 防腐剤としては、パラオキシ安息香酸エステル類、クロロブタノール、ベンジルアルコール、フェネチルアルコール、デヒドロ酢酸、ソルビン酸などが挙げられる。 Preservatives include paraoxybenzoic acid esters, chlorobutanol, benzyl alcohol, phenethyl alcohol, dehydroacetic acid, and sorbic acid.

 抗酸化剤としては、水溶性抗酸化剤であるアスコルビン酸、システインハイドロクロライド、重硫酸ナトリウム、メタ重亜硫酸ナトリウム、亜硫酸ナトリウム、脂溶性抗酸化剤であるアスコルビルパルミテート、ブチル化ハイドロキシアニソール(BHA)、ブチル化ハイドロキシトルエン(BHT)、レシチン、プロピルガレート、α-トコフェロール、及び金属キレート剤であるクエン酸、エチレンジアミン四酢酸(EDTA)、ソルビトール、酒石酸、リン酸が挙げられる。 Antioxidants include the water-soluble antioxidants ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, and sodium sulfite, the fat-soluble antioxidants ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, and α-tocopherol, and the metal chelating agents citric acid, ethylenediaminetetraacetic acid (EDTA), sorbitol, tartaric acid, and phosphoric acid.

 矯味矯臭剤としては、医薬分野において通常に使用される甘味料、香料などが、着色剤としては、医薬分野において通常に使用される着色料が挙げられる。 Flavors include sweeteners and fragrances that are commonly used in the pharmaceutical field, and colorants include colorants that are commonly used in the pharmaceutical field.

 本発明の医薬組成物の投与経路は特に限定されず、経口でも非経口投与でもよい。非経口投与としては、具体的には、注射投与、経鼻投与、経肺投与、経皮投与、などが挙げられる。注射投与としては、静脈内注射、筋肉内注射、腹腔内注射、皮下注射、骨髄内注射、髄腔内注射、皮内注射が例示できる。投与方法は、患者の年齢、症状により適宜選択することができる。 The administration route of the pharmaceutical composition of the present invention is not particularly limited, and may be oral or parenteral. Specific examples of parenteral administration include injection, nasal administration, pulmonary administration, and transdermal administration. Examples of injection include intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, intramedullary injection, intrathecal injection, and intradermal injection. The administration method can be appropriately selected depending on the age and symptoms of the patient.

 本発明の医薬組成物は、治療的有効量で対象に投与される。「治療的有効量」とは、選択された状態を緩和するのに有用である治療作用物質の量を指す。治療的有効量は、使用目的、投与経路、患者の年齢や症状等によって適宜決定される。 The pharmaceutical compositions of the present invention are administered to a subject in a therapeutically effective amount. A "therapeutically effective amount" refers to an amount of a therapeutic agent that is useful for alleviating a selected condition. The therapeutically effective amount is determined as appropriate depending on the intended use, route of administration, age and symptoms of the patient, etc.

 本発明の医薬組成物の適用対象となる腫瘍としては、例えば、急性骨髄性白血病、慢性骨髄性白血病、急性リンパ性白血病、及び慢性リンパ性白血病などの白血病、ホジキンリンパ腫、及び非ホジキンリンパ腫などの悪性リンパ腫、多発性骨髄腫、骨髄異形成症候群、頭頸部癌、舌癌、咽頭癌、口腔癌、食道癌、食道腺癌、胃癌、大腸癌、結腸癌、直腸癌、肝臓癌、胆嚢・胆管癌、胆道癌、膵臓癌、甲状腺癌、非小細胞肺癌及び小細胞肺癌などの肺癌、乳癌、卵巣癌、子宮頚癌、子宮体癌、子宮内膜癌、膣癌、外陰部癌、腎癌、腺癌、尿路上皮癌、前立腺癌、精巣腫瘍、骨・軟部肉腫、ブドウ膜悪性黒色腫、悪性黒色腫、メルケル細胞癌などの皮膚癌、神経芽腫、神経膠芽腫、脳腫瘍、及び胸膜中皮腫などの悪性腫瘍、ならびに血管筋脂肪腫などの良性腫瘍が挙げられるが、これらに限定されない。好ましくは、腫瘍は大腸がんなどの悪性腫瘍である。 Tumors to which the pharmaceutical composition of the present invention can be applied include, for example, leukemias such as acute myeloid leukemia, chronic myeloid leukemia, acute lymphocytic leukemia, and chronic lymphocytic leukemia, malignant lymphomas such as Hodgkin's lymphoma and non-Hodgkin's lymphoma, multiple myeloma, myelodysplastic syndrome, head and neck cancer, tongue cancer, pharyngeal cancer, oral cancer, esophageal cancer, esophageal adenocarcinoma, gastric cancer, colorectal cancer, colon cancer, rectal cancer, liver cancer, gallbladder/bile duct cancer, biliary tract cancer, and pancreatic cancer. , thyroid cancer, lung cancer such as non-small cell lung cancer and small cell lung cancer, breast cancer, ovarian cancer, cervical cancer, uterine cancer, endometrial cancer, vaginal cancer, vulvar cancer, renal cancer, adenocarcinoma, urothelial carcinoma, prostate cancer, testicular tumor, bone and soft tissue sarcoma, uveal malignant melanoma, malignant melanoma, skin cancer such as Merkel cell carcinoma, malignant tumors such as neuroblastoma, glioblastoma, brain tumor, and pleural mesothelioma, as well as benign tumors such as angiomyolipoma, but are not limited to these. Preferably, the tumor is a malignant tumor such as colorectal cancer.

 本発明の医薬組成物は、本発明の目的を損なわない限り、他の薬剤や治療方法と併用してもよい。 The pharmaceutical composition of the present invention may be used in combination with other drugs or treatment methods as long as the purpose of the present invention is not impaired.

6.治療方法
 本出願は、キマーゼ阻害剤とIL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上を組み合わせて対象に投与することを特徴とする、腫瘍を治療する方法も提供する。
6. Treatment Method The present application also provides a method for treating a tumor, comprising administering to a subject a chymase inhibitor in combination with one or more selected from IL-18, an anti-PD-1/PD-L1 antibody, and IL-12.

 本出願は、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上を投与する又は投与された対象に、キマーゼ阻害剤を投与することを含む、IL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上の抗腫瘍効果を増強するための方法も提供する。 The present application also provides a method for enhancing the antitumor effect of any one or more selected from IL-18, anti-PD-1/PD-L1 antibodies, and IL-12, comprising administering a chymase inhibitor to a subject to which any one or more selected from IL-18, anti-PD-1/PD-L1 antibodies, and IL-12 are administered or which has been administered to a subject.

 上記方法において、キマーゼ阻害剤とIL-18、抗PD-1/PD-L1抗体、及びIL-12から選ばれるいずれか1以上は、同時に投与してもよいし、順次に投与してもよい。投与経路は各々特に限定されず、前項に記載した方法を適用できる。 In the above method, the chymase inhibitor and one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and IL-12 may be administered simultaneously or sequentially. There are no particular limitations on the administration route for each, and the method described in the previous section can be applied.

 以下、本発明について実施例を用いて具体的に説明するが、本発明はこれらの実施例に限定されるものではない。 The present invention will be specifically explained below using examples, but the present invention is not limited to these examples.

実施例1:ペプチド性キマーゼ阻害剤とIL-18併用の抗腫瘍効果
 方法:7週齢の雄性BALB-cマウス(日本SLCより購入)の腹腔内に大腸がん細胞(CT26:5 x 104 cells)を投与し、投与後3日目より投薬を開始した。プラセボ群(n=8)は、0.17%ジメチルスルホキシド(薬物の溶媒液:プラセボ)を1日1回腹腔内投与した。IL-18投与群(n=8)は、2 μg/bodyのIL-18(mouse IL-18:GSK社から入手)を2日に1回腹腔内投与した。キマーゼ阻害剤投与群(n=8)は、3 mg/kgのSuc-Val-Pro-Phep(OPh)2(ペプチド性キマーゼ阻害剤:ペプチド研究所で合成)を1日1回腹腔内投与した。IL-18・キマーゼ阻害剤併用群(n=8)は、2 μg/mouseのIL-18を2日に1回及び3 mg/kgのキマーゼ阻害剤を1日1回腹腔内投与した。投薬は細胞移植後30日で終了し、細胞移植後31日と100日の時点で解析を行った。
Example 1: Antitumor effect of combined use of peptidic chymase inhibitor and IL-18 Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration. The placebo group (n=8) received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day. The IL-18 administration group (n=8) received 2 μg/body of IL-18 (mouse IL-18: obtained from GSK) intraperitoneally once every two days. The chymase inhibitor administration group (n=8) received 3 mg/kg of Suc-Val-Pro-Phe p (OPh) 2 (peptidic chymase inhibitor: synthesized at Peptide Institute) intraperitoneally once a day. The IL-18/chymase inhibitor combination group (n=8) received 2 μg/mouse IL-18 once every two days and 3 mg/kg chymase inhibitor once a day intraperitoneally. Medication was terminated 30 days after cell transplantation, and analysis was performed 31 and 100 days after cell transplantation.

 観察:朝、夕2回、マウスの生死について観察した。
 解析方法:各群生存曲線の有意差検定は、カプランマイヤー法にて実施し、P値が5%以下を有意とした。
Observation: Mice were observed twice a day, morning and evening, for survival.
Analysis method: Significance tests for survival curves of each group were performed using the Kaplan-Meier method, with a P value of 5% or less considered significant.

 結果:細胞移植後31日の時点で、プラセボ群の生存曲線とIL-18投与群(P = 0.5024)及びキマーゼ阻害剤群(P = 0.2068)の生存曲線の間には有意な生存期間の差を認めなかった。一方、プラセボ群とIL-18・キマーゼ阻害剤併用群の生存曲線の間には有意差が認められ(P = 0.0125)、IL-18・キマーゼ阻害剤併用群は、プラセボ群に比して有意に生存期間を延長することが認められた(図1)。細胞移植後100日の時点で、キマーゼ阻害剤群の生存したマウスはなく、IL-18投与群では1匹生存していたが、プラセボ群の生存曲線とIL-18投与群(P = 0.2916)及びキマーゼ阻害剤群(P = 0.2507)の生存曲線の間には有意な生存期間の差を認めなかった(図1)。一方、プラセボ群とIL-18・キマーゼ阻害剤併用群の生存曲線の間には有意差が認められた(P = 0.0126、図1)。また、生存していたマウスは、肉眼的にはがんの存在が消失していた(図2)。 Results: At 31 days after cell transplantation, there was no significant difference in survival time between the placebo group and the IL-18 group (P = 0.5024) or the chymase inhibitor group (P = 0.2068). On the other hand, there was a significant difference between the placebo group and the IL-18/chymase inhibitor combination group (P = 0.0125), indicating that the IL-18/chymase inhibitor combination group had a significantly longer survival time than the placebo group (Figure 1). At 100 days after cell transplantation, there were no surviving mice in the chymase inhibitor group, and one mouse in the IL-18 group was surviving, but there was no significant difference in survival time between the placebo group and the IL-18 group (P = 0.2916) or the chymase inhibitor group (P = 0.2507) (Figure 1). On the other hand, a significant difference was observed between the survival curves of the placebo group and the IL-18/chymase inhibitor combination group (P = 0.0126, Figure 1). Furthermore, the tumors in the surviving mice were macroscopically clear (Figure 2).

 今回の実験において、キマーゼ阻害剤の単独投与では、がん移植後の生存期間の延長を認めなかったが、IL-18とキマーゼ阻害剤を併用することにより、IL-18単独に比較して生存期間の延長を認めた。 In this experiment, administration of a chymase inhibitor alone did not result in an extension of survival time after cancer transplantation, but administration of IL-18 in combination with a chymase inhibitor extended survival time compared to administration of IL-18 alone.

実施例2:市販キマーゼ阻害剤とIL-18併用の抗腫瘍効果
 方法:7週齢の雄性BALB-cマウス(日本SLCより購入)の腹腔内に大腸がん細胞(CT26:5 x 104 cells)を投与し、投与後3日目より投薬を開始した。プラセボ群(n=10)は、0.17%ジメチルスルホキシド(薬物の溶媒液:プラセボ)を1日1回腹腔内投与した。IL-18投与群(n=10)は、2μg/bodyのIL-18(前掲mouse IL-18)を2日に1回腹腔内投与した。キマーゼ阻害剤投与群(n=10)は、5 mg/kg(100μg/body)のFulacimstat(キマーゼ阻害剤:MedChemExpress社)を1日1回腹腔内投与した。IL-18・キマーゼ阻害剤併用群(n=10)は、2 μg/bodyのIL-18を2日に1回及び5 mg/kgのキマーゼ阻害剤を1日1回腹腔内投与した。投薬は細胞移植後30日で終了し、その後110日まで解析を行った。
Example 2: Antitumor effect of a combination of a commercially available chymase inhibitor and IL-18 Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration. The placebo group (n=10) received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day. The IL-18 administration group (n=10) received 2μg/body of IL-18 (mouse IL-18 as above) intraperitoneally once every two days. The chymase inhibitor administration group (n=10) received 5mg/kg (100μg/body) of Fulacimstat (chymase inhibitor: MedChemExpress) intraperitoneally once a day. The IL-18 + chymase inhibitor combination group (n=10) received 2 μg/body IL-18 once every two days and 5 mg/kg chymase inhibitor once a day intraperitoneally. Medication was discontinued 30 days after cell transplantation, and analysis was continued up to 110 days after.

 投薬終了後、マウスの生死を毎日確認した。
 解析方法:各群生存曲線の有意差検定は、カプランマイヤー法にて実施し、P値が5%以下を有意とした。
After the administration, the mice were checked daily for mortality.
Analysis method: Significance tests for survival curves of each group were performed using the Kaplan-Meier method, with a P value of 5% or less considered significant.

 結果:プラセボ群とキマーゼ阻害剤投与群では、生存率に相違はなく、いずれも40日以前にすべてのマウスは死亡した。一方、IL-18投与群とIL-18・キマーゼ阻害剤併用群では、プラセボに比較して明らかな生存率の向上がみられ、細胞移植後110日の時点で、IL-18・キマーゼ阻害剤併用群はIL-18投与群よりも高い生存率を示した(図3)。 Results: There was no difference in survival rate between the placebo group and the chymase inhibitor group, and all mice died before 40 days in both groups. On the other hand, the IL-18 group and the IL-18/chymase inhibitor combination group showed a clear improvement in survival rate compared to the placebo group, and at 110 days after cell transplantation, the IL-18/chymase inhibitor combination group showed a higher survival rate than the IL-18 group (Figure 3).

 異なるキマーゼ阻害剤を用いた場合においても、全体として、ペプチド性キマーゼ阻害剤と同様の傾向がみられた。すなわち、キマーゼ阻害剤の単独投与では、がん移植後の生存期間の延長を認めなかったが、IL-18とキマーゼ阻害剤を併用することにより、IL-18単独に比較して生存期間の延長を認めた。  Even when different chymase inhibitors were used, the overall trend was similar to that of peptidic chymase inhibitors. In other words, administration of a chymase inhibitor alone did not result in an extension of survival time after cancer transplantation, but the combination of IL-18 and a chymase inhibitor extended survival time compared to IL-18 alone.

実施例3:腫瘍抗原刺激による腹腔内リンパ球細胞の変化
 方法:7週齢の雄性BALB-cマウス(日本SLCより購入)の腹腔内に大腸がん細胞(CT26:5 x 104 cells)を投与し、細胞移植後3日、5日、7日、11日、14日、17日、21日に腹水を回収(毎回n=4)した。
Example 3: Changes in intraperitoneal lymphocyte cells due to tumor antigen stimulation Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and ascites was collected 3, 5, 7, 11, 14, 17, and 21 days after cell transplantation (n=4 each time).

 腹水の回収は、マウスをIsofluraneで麻酔し、腹腔内に冷却したPBSを注射し、回収した(全8ml)。回収した腹水を遠心し、2回洗浄した後、trypan blue exclusion assay testを用いて細胞数を数えた。回収した細胞をチャンバーに塗布し、HE染色及びTB染色に供した。また、フローサイトメトリーで細胞のサブセットを解析した。 To collect ascites, mice were anesthetized with Isoflurane, and chilled PBS was injected intraperitoneally and collected (total 8 ml). The collected ascites was centrifuged and washed twice, and the number of cells was counted using a trypan blue exclusion assay test. The collected cells were applied to a chamber and subjected to HE and TB staining. Cell subsets were also analyzed by flow cytometry.

 結果:マスト細胞は、細胞移植後7日以降増加傾向を示したが、顆粒を有するマスト細胞は減少した(図4)。腫瘍組織の周囲にマスト細胞が集まる傾向もみられた(データ示さず)。腫瘍細胞の移植により、CD4+T細胞、CD8+T細胞は増加し、CD4+T細胞/CD8+T細胞の割合は減少した(図5)。これはCD8+Killer細胞の増加によると推測される。NK細胞、特に活性されたkiller NK cell(DX5+B220+ NK cell)も腫瘍細胞の移植により増加した(図6)。 Results: Mast cells showed a tendency to increase from 7 days after cell transplantation, but granular mast cells decreased (Fig. 4). Mast cells also tended to gather around the tumor tissue (data not shown). Tumor cell transplantation increased CD4 + T cells and CD8 + T cells, while the ratio of CD4 + T cells/CD8 + T cells decreased (Fig. 5). This is presumably due to an increase in CD8 + killer cells. NK cells, especially activated killer NK cells (DX5 + B220 + NK cells), also increased after tumor cell transplantation (Fig. 6).

 腹腔内の免疫細胞サブセットの解析では、10日までの移植後早期にはNK細胞が増加し、10日~21日まではCD8+T細胞が増加し、マスト細胞は7日以後に徐々に増加した(図7)。 Analysis of immune cell subsets in the peritoneal cavity showed that NK cells increased early after transplantation up to day 10, CD8 + T cells increased from days 10 to 21, and mast cells gradually increased after day 7 (Figure 7).

実施例4:ペプチド性キマーゼ阻害剤とIL-18併用による、腹腔内抗腫瘍リンパ球細胞の変化
 方法:7週齢の雄性BALB-cマウス(日本SLCより購入)の腹腔内に大腸がん細胞(CT26:5 x 104 cells)を投与し、投与後3日目より投薬を開始した。対照群は、0.17%ジメチルスルホキシド(薬物の溶媒液:プラセボ)を1日1回腹腔内投与した。IL-18単独投与群は、2μg/bodyのIL-18(前掲mouse IL-18)を2日に1回腹腔内投与した。キマーゼ阻害剤単独投与群は、3 mg/kg(100μg/body)のSuc-Val-Pro-Phep(OPh)2を1日1回腹腔内投与した。IL-18・キマーゼ阻害剤併用群は、2μg/bodyのIL-18を2日に1回及び3 mg/kgのキマーゼ阻害剤を1日1回腹腔内投与した。投薬は細胞移植後28日で終了した。
Example 4: Changes in intraperitoneal antitumor lymphocytes by combined use of a peptide chymase inhibitor and IL-18 Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration. The control group received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day. The IL-18 alone administration group received 2μg/body of IL-18 (see above mouse IL-18) intraperitoneally once every two days. The chymase inhibitor alone administration group received 3mg/kg (100μg/body) of Suc-Val-Pro-Phe p (OPh) 2 intraperitoneally once a day. The IL-18/chymase inhibitor combination group received intraperitoneal administration of 2 μg/body of IL-18 once every two days and 3 mg/kg of chymase inhibitor once a day. Medication was terminated 28 days after cell transplantation.

 実施例3にしたがい、細胞移植後21日~28日にマウスの腹水を採取し、腹水内の細胞のサブセットを解析した。 Following Example 3, ascites was collected from the mice 21 to 28 days after cell transplantation, and the cell subsets in the ascites were analyzed.

 結果:IL-18投与群とIL-18・キマーゼ阻害剤併用群では、いずれも腹腔内滲出細胞(PEC)におけるCD4+T細胞/CD8+T細胞の割合がプラセボより優位に低く、CD8+T細胞、特にCD44highCD8+T細胞の数が増加した(図8)。IL-18投与群とIL-18・キマーゼ阻害剤併用群では、NK細胞、特にB220+killing NK細胞の数がプラセボより優位に高かった(図9)。 Results: In both the IL-18 group and the IL-18/chymase inhibitor combination group, the ratio of CD4 + T cells/CD8 + T cells in peritoneal exudate cells (PEC) was significantly lower than that in the placebo group, and the number of CD8 + T cells, especially CD44highCD8 + T cells, increased (Fig. 8). In the IL-18 group and the IL-18/chymase inhibitor combination group, the number of NK cells, especially B220 + killing NK cells, was significantly higher than that in the placebo group (Fig. 9).

 腹水中の免疫細胞サブセットの構成からも、IL-18とキマーゼを併用することで、IL-18単独に比較して、抗腫瘍効果が高まることが示唆された。 The composition of immune cell subsets in the ascites also suggested that the combined use of IL-18 and chymase enhanced the antitumor effect compared to IL-18 alone.

実施例5:キマーゼ阻害剤と抗PD-1抗体併用の抗腫瘍効果
 方法:7週齢の雄性BALB-cマウス(日本SLCより購入)の腹腔内に大腸がん細胞(CT26:5 x 104 cells)を投与し、投与後3日目より投薬を開始した。プラセボ群(n=10)は、0.17%ジメチルスルホキシド(薬物の溶媒液:プラセボ)を1日1回腹腔内投与した。抗PD-1抗体投与群(n=10)は、200μg/bodyの抗PD-1抗体(BioXCell製)を1週間に3回(計12回)腹腔内投与した。キマーゼ阻害剤投与群(n=10)は、100μg/bodyのFulacimstat(キマーゼ阻害剤:MedChemExpress社)を1日1回(計30回)腹腔内投与した。抗PD-1抗体・キマーゼ阻害剤併用群(n=10)は、200μg/bodyの抗PD-1抗体を1週間に3回(計12回)及び100μg/bodyのキマーゼ阻害剤を1日1回(計30回)腹腔内投与した。投薬は細胞移植後30日で終了し、その後34日まで解析を行った。
Example 5: Antitumor effect of combination of chymase inhibitor and anti-PD-1 antibody Method: Colon cancer cells (CT26: 5 x 104 cells) were administered intraperitoneally to 7-week-old male BALB-c mice (purchased from Japan SLC), and administration was started on the third day after administration. The placebo group (n=10) received 0.17% dimethyl sulfoxide (drug solvent: placebo) intraperitoneally once a day. The anti-PD-1 antibody administration group (n=10) received 200μg/body of anti-PD-1 antibody (manufactured by BioXCell) intraperitoneally three times a week (total 12 times). The chymase inhibitor administration group (n=10) received 100μg/body of Fulacimstat (chymase inhibitor: MedChemExpress) intraperitoneally once a day (total 30 times). The anti-PD-1 antibody/chymase inhibitor combination group (n=10) received intraperitoneal administration of 200μg/body of anti-PD-1 antibody three times a week (total 12 times) and 100μg/body of chymase inhibitor once a day (total 30 times). Medication was completed 30 days after cell transplantation, and analysis was continued up to 34 days after that.

 観察:朝、夕2回、マウスの生死について観察した。
 解析方法:各群生存曲線の有意差検定は、カプランマイヤー法にて実施し、P値が5%以下を有意とした。
Observation: Mice were observed twice a day, morning and evening, for survival.
Analysis method: Significance tests for survival curves of each group were performed using the Kaplan-Meier method, with a P value of 5% or less considered significant.

 結果:がん細胞移植後34日の時点で、抗PD-1抗体とキマーゼ阻害剤併用群は、プラセボ群(20%)、キマーゼ阻害剤単独投与群(20%)のみならず、抗PD-1抗体単独投与群(10%)比べて、大きな生存率(50%)を示し、抗PD-1抗体にキマーゼ阻害剤を加えることによる併用効果が認められた(図10)。 Results: 34 days after cancer cell transplantation, the group receiving the combination of anti-PD-1 antibody and chymase inhibitor showed a higher survival rate (50%) than not only the placebo group (20%) and the group receiving chymase inhibitor alone (20%), but also the group receiving anti-PD-1 antibody alone (10%), demonstrating the combined effect of adding a chymase inhibitor to an anti-PD-1 antibody (Figure 10).

 今回の実験において、キマーゼ阻害剤の単独投与では、がん移植後の生存期間の延長を認めなかったが、抗PD-1抗体とキマーゼ阻害剤を併用することにより、抗PD-1抗体単独に比較して生存期間の延長を認めた。 In this experiment, administration of a chymase inhibitor alone did not result in an extension of survival time after cancer transplantation, but the combination of an anti-PD-1 antibody and a chymase inhibitor extended survival time compared to anti-PD-1 antibody alone.

 本発明は腫瘍の治療において有用である。 The present invention is useful in treating tumors.

 本明細書中で引用した全ての刊行物、特許及び特許出願をそのまま参考として本明細書中にとり入れるものとする。 All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety.

Claims (6)

 キマーゼ阻害剤を有効成分として含む、腫瘍を治療するための医薬組成物であって、インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と併用されることを特徴とする、前記医薬組成物。 A pharmaceutical composition for treating a tumor, comprising a chymase inhibitor as an active ingredient, the pharmaceutical composition being used in combination with one or more of interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12).  インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上と、キマーゼ阻害剤とを組み合わせて含む、腫瘍を治療するための医薬組成物。 A pharmaceutical composition for treating a tumor comprising a combination of one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12) with a chymase inhibitor.  インターロイキン-18(IL-18)、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上を有効成分として含む、腫瘍を治療するための医薬組成物であって、キマーゼ阻害剤と併用されることを特徴とする、前記医薬組成物。 A pharmaceutical composition for treating tumors, comprising as an active ingredient one or more selected from interleukin-18 (IL-18), anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12), which is used in combination with a chymase inhibitor.  キマーゼ阻害剤が、Suc-Val-Pro-PheP(OPh)2、Chymostatin、Fulacimstat、2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic acid、及びN-[1-Benzyl-2,3-dioxo-6-(2-pyridyloxy)hexyl]-2-[2-phenyl-5-(formylamino)-6-oxo-1,6-dihydropyrimidine-1-yl]acetamide、(2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide)から選択されるいずれかである、請求項1~3のいずれか1項に記載の医薬組成物。 Chymase inhibitors include Suc-Val-Pro-PheP(OPh) 2 , Chymostatin, Fulacimstat, 2-[4-[[(5-Fluoro-3-methylbenzo[b]thien-2-yl)sulfonyl]amino]-3-(methylsulfonyl)phenyl]-4-thiazolecarboxylic The pharmaceutical composition according to any one of claims 1 to 3, wherein the compound is any one selected from the group consisting of (2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide), (2-[5-amino-2-(4-fluorophenyl)-1,6-dihydro-6-oxo-1-pyrimidinyl]-N-[1-[(5-methoxycarbonyl-2-benzoxazolyl)carbonyl]-2-phenylethyl]acetamide).  IL-18、抗PD-1/PD-L1抗体、及びIL-12が、ヒト由来である、請求項1~3のいずれか1項に記載の医薬組成物。 The pharmaceutical composition according to any one of claims 1 to 3, wherein IL-18, anti-PD-1/PD-L1 antibody, and IL-12 are derived from humans.  IL-18、抗PD-1/PD-L1抗体、及びインターロイキン-12(IL-12)から選ばれるいずれか1以上とキマーゼ阻害剤が同時に又は順次に投与される、請求項1~3のいずれか1項に記載の医薬組成物。
 
The pharmaceutical composition according to any one of claims 1 to 3, wherein the chymase inhibitor and any one or more selected from IL-18, anti-PD-1/PD-L1 antibody, and interleukin-12 (IL-12) are administered simultaneously or sequentially.
PCT/JP2024/027634 2023-08-04 2024-08-02 Antitumor therapy by combined use of chymase inhibitor and immunotherapy Pending WO2025033330A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2023127528 2023-08-04
JP2023-127528 2023-08-04

Publications (1)

Publication Number Publication Date
WO2025033330A1 true WO2025033330A1 (en) 2025-02-13

Family

ID=94534372

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2024/027634 Pending WO2025033330A1 (en) 2023-08-04 2024-08-02 Antitumor therapy by combined use of chymase inhibitor and immunotherapy

Country Status (1)

Country Link
WO (1) WO2025033330A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001521906A (en) * 1997-11-03 2001-11-13 ザ・ウイスター・インステイテユート・オブ・アナトミー・アンド・バイオロジー Methods and compositions for inhibiting angiogenesis and treating cancer
JP2010522200A (en) * 2007-03-23 2010-07-01 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Combination cancer treatment of human IL-18 and anti-CD20 antibody
JP2015505813A (en) * 2011-10-11 2015-02-26 ウニヴェルズィテート チューリッヒ Pharmaceutical composition comprising IL-12 and a T cell inhibitory molecular blocker for tumor therapy
WO2016021720A1 (en) * 2014-08-07 2016-02-11 学校法人兵庫医科大学 Therapeutic agent for cancer which comprises combination of il-18 and molecule-targeting antibody

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001521906A (en) * 1997-11-03 2001-11-13 ザ・ウイスター・インステイテユート・オブ・アナトミー・アンド・バイオロジー Methods and compositions for inhibiting angiogenesis and treating cancer
JP2010522200A (en) * 2007-03-23 2010-07-01 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Combination cancer treatment of human IL-18 and anti-CD20 antibody
JP2015505813A (en) * 2011-10-11 2015-02-26 ウニヴェルズィテート チューリッヒ Pharmaceutical composition comprising IL-12 and a T cell inhibitory molecular blocker for tumor therapy
WO2016021720A1 (en) * 2014-08-07 2016-02-11 学校法人兵庫医科大学 Therapeutic agent for cancer which comprises combination of il-18 and molecule-targeting antibody

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HAMANISHI JUNZO; MANDAI MASAKI; MATSUMURA NORIOMI; ABIKO KAORU; BABA TSUKASA; KONISHI IKUO: "PD-1/PD-L1 blockade in cancer treatment: perspectives and issues", INTERNATIONAL JOURNAL OF CLINICAL ONCOLOGY, SPRINGER SINGAPORE, SINGAPORE, vol. 21, no. 3, 22 February 2016 (2016-02-22), Singapore, pages 462 - 473, XP035974610, ISSN: 1341-9625, DOI: 10.1007/s10147-016-0959-z *
YI LUO; HITOSHI OHMORI; TAKASUMI SHIMOMOTO; KIYOMU FUJII; TOMONORI SASAHIRA; YOSHITOMO CHIHARA; HIROKI KUNIYASU: "Anti-angiotensin and hypoglycemic treatments suppress liver metastasis of colon cancer cells. Pathobiology. ", PATHOBIOLOGY, S. KARGER, vol. 78, no. 5, 1 January 2011 (2011-01-01), pages 285 - 290, XP009560758, ISSN: 1015-2008 *

Similar Documents

Publication Publication Date Title
US11622961B2 (en) Combination therapies for treating cancer
RU2723021C2 (en) Use of the plinabulin in combination with the immune response control point inhibitors
KR20230006568A (en) Triple combination therapy to enhance cancer cell death in cancers with low immunogenicity
CN105873440B (en) Use of the composition inhibiting TIE2 kinase in the preparation of medicines for treating cancer
US20200085839A1 (en) Combination of immunotherapeutics and bisfluoroalkyl-1,4-benzodiazepinone compounds for treating lymphomas
Ströhlein et al. Immunotherapy of peritoneal carcinomatosis with the antibody catumaxomab in colon, gastric, or pancreatic cancer: an open-label, multicenter, phase I/II trial
WO2019124500A1 (en) Combination drug including tlr7 agonist
TW201922793A (en) Uses of PD-1 antibody combined with VEGFR inhibitor for treating small cell lung cancer
JPWO2016204193A1 (en) Anti-cancer agent
JP6240083B2 (en) Small molecule enhancer for dendritic cell carcinoma vaccine
JP2024513505A (en) Compositions and methods for treating tumors
JP2021502423A (en) Methods and Compositions for Treating Cancer by Modifying Multiple Arms of the Immune System
JP2020506945A (en) Methods, compositions and kits for treating cancer
US20200399377A1 (en) Combination cancer therapy with anti-cancer agents and antibodies targeting a complex comprising non-classical hla-i and neoantigen
KR20160050026A (en) Antitumor agent and antitumor effect enhancer
KR20070108402A (en) Treatment of lymphoma with a combination of chemotherapeutic agents and IL-2 and optionally anti-CD20 antibodies
TW202200191A (en) Pharmaceutical composition for treating cancer, comprising fusion protein comprising il-2 protein and cd80 protein and anticancer drug
US20220025061A1 (en) Combination therapies for treating disease using an innate immunity modifier and an ox40 agonist
KR20190051983A (en) Combinations containing ABX196 for the treatment of cancer
WO2025033330A1 (en) Antitumor therapy by combined use of chymase inhibitor and immunotherapy
KR102032933B1 (en) Pharmaceutical composition for treatment or palliation of elderly or end-stage cancer patient
KR20210131316A (en) Administration of SUMO-activating enzyme inhibitors and checkpoint inhibitors
JP2016104703A (en) Antitumor agent
US20230390312A1 (en) Novel therapeutic combinations comprising derivatives of oxazaphosphorines for the treatment of cancer
TW202315631A (en) Trimodality therapy to enhance cancer cell killing in poorly-immunogenic cancers can induce immunogens and inhibit immune checkpoint

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: 24851755

Country of ref document: EP

Kind code of ref document: A1