[go: up one dir, main page]

WO2020136879A1 - Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor - Google Patents

Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor Download PDF

Info

Publication number
WO2020136879A1
WO2020136879A1 PCT/JP2018/048494 JP2018048494W WO2020136879A1 WO 2020136879 A1 WO2020136879 A1 WO 2020136879A1 JP 2018048494 W JP2018048494 W JP 2018048494W WO 2020136879 A1 WO2020136879 A1 WO 2020136879A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
immunotherapeutic agent
patient
cells
injured
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/JP2018/048494
Other languages
French (fr)
Japanese (ja)
Inventor
幸太 岩堀
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.)
University of Osaka NUC
Original Assignee
Osaka University NUC
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 Osaka University NUC filed Critical Osaka University NUC
Priority to PCT/JP2018/048494 priority Critical patent/WO2020136879A1/en
Publication of WO2020136879A1 publication Critical patent/WO2020136879A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins

Definitions

  • the present disclosure provides a method of presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor, a method for assisting in determining the usefulness of an immunotherapeutic agent for treating a malignant tumor, an immunotherapy for treating a malignant tumor.
  • Tumor immunotherapy is a tumor treatment method that uses the immune response of tumor patients to tumor cells.
  • anti-CTLA-4 antibody, anti-PD-1 antibody, etc. that bind to a protein on T cells are used to activate cytotoxic T cells of the tumor patient's own, and cancer cell is activated by the function.
  • Are killed Non-Patent Document 1 and Non-Patent Document 2.
  • Non-Patent Document 2 and 3 A bispecific molecule (bispecific antibody) that specifically recognizes a cell surface marker of a tumor cell and a surface marker of a T cell with one molecule is currently used as the engager.
  • the tumor immunotherapeutic drug pembrolizumab (trade name KEYTRUDA), which is covered by insurance for non-small cell lung cancer, has a response rate of 19% and is currently ineffective for many patients. For this reason, companion diagnosis is performed in order to select patient groups in which the therapeutic drug for tumor immunotherapy is effective.
  • pembrolizumab PD-L1 staining using a biopsy sample is used as a companion diagnostic agent, but the response rate is 45% even in a patient group positive for PD-L1 staining by 50% or more. Therefore, development of companion diagnostics superior to PD-L1 staining has been earnestly desired.
  • tumor immunotherapeutic drugs may cause adverse events such as interstitial lung disease and encephalitis, which are treatment discontinuation. No serious adverse events can be predicted.
  • an object of the present invention is to present information necessary for companion diagnosis for predicting the usefulness of immunotherapeutic agents for individual patients. Another object is to provide an immunotherapeutic agent to be administered to a patient who is suggested to be useful.
  • the inventors of the present invention have conducted extensive studies and found that immunotherapy can be performed in vitro by evaluating whether tumor cells are injured by using mononuclear cells in peripheral blood collected from individual patients. It was found that the usefulness of the agent can be presented for each patient.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • the usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and cannot resume administration, When the step 2 shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent for the patient needs to be interrupted and the administration cannot be restarted. If the step of presenting that no adverse event occurs, and/or the step 2 is shown in the evaluation that the tumor cells are not injured, the patient is treated with the immunotherapeutic agent.
  • a step showing that the tumor cells are not injured The presentation method according to any one of (I-1) to (I-3), including: (I-5) The presentation according to (I-4), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells.
  • Method. (I-6) In (I-4), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measurement value of interferon- ⁇ in the culture medium. How to present the description.
  • the in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, from (I-1) to (I) -The presentation method according to any one of 6).
  • the bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells.
  • (I-10) The method according to any one of (I-1) to (I-9), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
  • the method for evaluating the usefulness of the immunotherapeutic agent in a patient having the malignant tumor Including gadgets, Testing reagent.
  • Step i a step of evaluating whether or not tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured
  • step ii the tumor in the evaluation
  • step ii shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent is harmful to the patient because the treatment needs to be interrupted and the administration cannot be restarted. If the step of presenting no event and/or step ii is shown in the assessment that the tumor cells are not injured, the immunotherapeutic agent is directed to the patient for treatment interruption.
  • the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measurement value of interferon- ⁇ in the culture medium. Described assistance method.
  • the in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, (I-12) to (I-12) -The auxiliary method according to any one of 17).
  • the bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells.
  • (I-21) The assisting method according to any one of (I-12) to (I-20), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
  • (I-22) The adjuvant method according to any one of (I-18) to (I-21), wherein the immunotherapeutic agent is used to evaluate the usefulness in a patient having the malignant tumor. Including a gadget, Testing reagent.
  • (II) Immunotherapeutic agent for treating malignant tumor (II-1) It is shown that the immunotherapeutic agent is useful in the presenting method according to any one of (I-1) to (I-10). An immunotherapeutic agent for treating a malignant tumor, which is administered to a patient having an established malignant tumor.
  • (II-2) First treatment for a patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for.
  • (II-3) Preoperative Chemistry for Patients with Malignant Tumors Presented as Useful for Immunotherapeutic Agents in the Presenting Method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for therapy.
  • (II-4) Postoperative assistance for a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for chemotherapy.
  • (II-5) A patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and radiation treatment An immunotherapeutic agent to be subsequently administered to the patient.
  • (II-6) A patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and the actinic radiation An immunotherapeutic agent administered to the patient after treatment.
  • Agent. (II-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (II-1) to (II-8) The immunotherapeutic agent according to any one of 1) above.
  • (II-10) The immunotherapy agent according to (II-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapy agent.
  • (II-11) Administered to a patient having a malignant tumor for whom an immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) , An immune checkpoint inhibitor, which is administered in combination with one or more other immune checkpoint inhibitors.
  • (II-12) Administered to a patient having a malignant tumor for whom the immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) , An immune checkpoint inhibitor, which is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (II-11) or (II-12) Immune checkpoint inhibitor described in (4).
  • the immune checkpoint inhibitor includes an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody (II-11) to (II-13) The immune checkpoint inhibitor according to any one of 1.
  • (III) Immunotherapy for treating malignant tumor (III-1) It has been proposed that the immunotherapeutic agent is useful in the presentation method according to any one of (I-1) to (I-10). Immunotherapy of a malignant tumor, wherein an immunotherapeutic agent is administered to a patient with a malignant tumor.
  • (III-3) An immunotherapeutic agent for a patient having a malignant tumor, which is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Immunotherapy of malignant tumors given as preoperative chemotherapy.
  • (III-4) An immunotherapeutic agent for a patient having a malignant tumor, which is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Is a postoperative adjuvant chemotherapy for malignant tumors.
  • III-5) A patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and radiation treatment Immunotherapy of a malignant tumor, wherein an immunotherapeutic agent is subsequently administered to the patient.
  • (III-6) A patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), wherein the actinic radiation is Immunotherapy of malignant tumors, wherein the patient is treated with an immunotherapeutic agent.
  • the immune checkpoint inhibitor is an immunotherapeutic agent containing an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody Therapy.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (III-1) to (III-8)
  • the immunotherapy of the malignant tumor as described in any one of 1) above.
  • III-11 Two types for patients with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Immunotherapy for malignant tumors, which is administered in combination with the above immune checkpoint inhibitors.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (III-11) or (III-12) ) Immunotherapy for malignant tumors described in (1) above.
  • (III-14) The (III-11) to (III-14), wherein the immune checkpoint inhibitor includes an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody.
  • the immunotherapy of the malignant tumor as described in any one of 1) above.
  • (III-15) Administered to a patient having a malignant tumor for whom the immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) Immunotherapy of malignant tumors, which comprises administering an immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent in combination.
  • (III-16) The malignant tumor according to (III-15), wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. Immunotherapy.
  • III-17 The immunotherapy for malignant tumor according to (III-15) or (III-16), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
  • (IV) Immunotherapeutic agent for treating malignant tumor (IV-1) It is shown that the immunotherapeutic agent is useful in the presentation method according to any one of (I-1) to (I-10).
  • An immunotherapeutic agent for treating a malignant tumor which is administered to a patient having an established malignant tumor.
  • IV-2 The immunotherapeutic agent according to (IV-1), which is administered for the primary treatment of malignant tumor.
  • IV-3 The immunotherapy agent according to (IV-1), which is administered for preoperative chemotherapy.
  • IV-4 The immunotherapy agent according to (IV-1), which is administered for postoperative adjuvant chemotherapy.
  • IV-5 The immunotherapy agent according to (IV-1), which is administered to the patient after radiation treatment.
  • (IV-6) The immunotherapy agent according to (IV-1), which is administered to the patient after chemoradiotherapy.
  • IV-7 The immunotherapy agent according to any one of (IV-1) to (IV-6), wherein the immunotherapy agent is an immune checkpoint inhibitor.
  • IV-8) The immunotherapy according to (IV-7), wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. Agent.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (IV-1) to (IV-8) An immunotherapy agent according to any one of (1) to (4).
  • IV-11 Any one of (IV-11) to (IV-10), wherein the immunotherapeutic agent is an immune checkpoint inhibitor, and is administered in combination with one or more other immune checkpoint inhibitors.
  • the immunotherapeutic agent according to one item.
  • the immunotherapeutic agent is an immune checkpoint inhibitor, and is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor, (IV-11) to (IV-10) The immunotherapeutic agent according to any one of 1. (IV-13) The immunotherapeutic agent according to any one of (IV-11) to (IV-10), which is administered in combination with one or more anticancer agents other than the immunotherapeutic agent.
  • Step 1 An immunotherapeutic agent for treating a malignant tumor, comprising the following Step 1 and Step 2, wherein: Of an active ingredient for the manufacture of an immunotherapeutic agent for treating a malignant tumor, which is administered to said patient for whom the immunotherapeutic agent has been shown to be useful by a method of presenting utility in a patient having use: Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • Step 2 Regarding the immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by the method of presenting the usefulness in the patient having the malignant tumor.
  • Step 3 Regarding the immunotherapeutic agent for treating a malignant tumor, comprising the following Step 1 and Step 2, the immunotherapeutic agent being useful by the method of presenting its usefulness in a patient having the malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • Step 5 Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • Step 6 Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor.
  • V-7) Use of an active ingredient for producing the immunotherapeutic agent according to any one of (V-1) to (V-6), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
  • the active ingredient is derived from anti-PD1 antibody, anti-PD-L1 antibody, anti-CTLA-4 antibody, anti-Tim3 antibody, or anti-LAG3 antibody, (V-1) to (V-7) Use of an active ingredient for the manufacture of the immunotherapeutic agent according to any one of claims.
  • V-9 The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (V-1) to (V-7)
  • V-10 Use of the active ingredient for the manufacture of the immunotherapeutic agent according to any one of (1) to (4).
  • V-10) The use of the active ingredient for producing the immunotherapeutic agent according to (V-9), wherein the lung cancer is complicated by interstitial pneumonia at the start of administration of the immunotherapeutic agent.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • an immunotherapeutic agent for treating a malignant tumor which comprises the following Step 1 and Step 2
  • the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor.
  • an immune checkpoint inhibitor which is administered to said patient presented with, wherein the immune checkpoint inhibitor is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (V-11) or (V-12) Use of an active ingredient for the manufacture of an immune checkpoint inhibitor as described in 1.).
  • the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody, (V-11) to (V-13) Use of an active ingredient for the manufacture of the immune checkpoint inhibitor according to any one of (1) to (4).
  • V-15 Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • V-15 The immunotherapy according to (V-14), wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma.
  • the active ingredient for the manufacture of a drug.
  • V-16 Use of the active ingredient for producing the immunotherapeutic agent according to (V-14) or (V-15), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
  • VI Method of obtaining information for presenting usefulness of immunotherapeutic agent for treating malignant tumor, and use for producing test reagent (VI-1) Treating malignant tumor, comprising the following steps Regarding the immunotherapeutic agent for achieving the above, a method for obtaining information for presenting usefulness in the patient having the malignant tumor: A step of obtaining information indicating whether or not the tumor cells, which are directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro, are injured. (VI-2) The above-mentioned usefulness is the presence or absence of an adverse event resulting from the immunotherapeutic agent that requires discontinuation of treatment and is incapable of resuming administration. Acquisition method.
  • (VI-3) The acquisition method according to (VI-1) or (VI-2), wherein the usefulness is the presence or absence of the effect of the immunotherapeutic agent.
  • (VI-4) The step of acquiring information reflecting whether or not the tumor cells are injured, In the step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, the value reflecting the tumor cytotoxic activity is the number of living cells or dead cells of the tumor cells.
  • (VI-5) The step of acquiring information that reflects whether tumor cells are injured, A step of obtaining a value that reflects the tumor cytotoxic activity of peripheral blood mononuclear cells that have come into contact with the tumor cells, wherein the in vitro contact is contact in a culture medium and reflects the tumor cytotoxic activity.
  • (VI-6) The in vitro contact is indirect contact of peripheral blood mononuclear cells collected from the patient with tumor cells via an engager, (VI-1) to (VI) -The acquisition method according to any one of 5).
  • (VI-7) The acquisition method according to (VI-6), wherein the engager is a bispecific molecule.
  • the bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells.
  • (VI-7) The method according to any one of (VI-1) to (VI-8), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
  • the information for presenting the usefulness of the immunotherapeutic agent in the patient having the malignant tumor is acquired. Use of an engager to produce a test reagent that
  • VI Device for presenting usefulness of immunotherapeutic agent for treating malignant tumor
  • the apparatus includes a processing unit,
  • the processing unit is Peripheral blood mononuclear cells collected from the patient, directly or indirectly, to obtain an evaluation result of evaluating whether tumor cells contacted in vitro are injured, If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is useful to the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is not useful to the patient, if not indicated, Presentation device.
  • the usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires discontinuation of treatment and cannot resume administration,
  • the processing unit is When the evaluation result shows that the tumor cells are injured, the immunotherapeutic agent does not cause an adverse event for the patient, which requires discontinuation of treatment and resumption of administration is impossible. And/or the evaluation result indicates that the tumor cells are not injured, the immunotherapeutic agent requires treatment interruption to the patient and resumption of administration is not possible. Suggesting that a possible adverse event will occur, The presentation device according to (VII-1).
  • the usefulness is presence or absence of the effect of the immunotherapeutic agent
  • the processing unit is If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is effective for the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is ineffective to the patient, The presentation device according to (VII-1) or (VII-2).
  • (VII-4) Whether the tumor cells contacted with the peripheral blood mononuclear cells are damaged or not, Obtain a value that reflects the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, compare the value that reflects the tumor cytotoxic activity with the corresponding reference value, the value from the corresponding reference value
  • the presentation device according to any one of (VII-1) to (VII-3), which is evaluated by whether or not it is high.
  • (VII-5) Evaluation of whether or not the value is higher than the corresponding reference value is Indicating that the tumor cells are injured when said value is higher than the corresponding reference value, and/or The presentation device according to (VII-6), comprising indicating that the tumor cells are not injured when the value is lower than a corresponding reference value.
  • (VII-6) The presentation according to (VII-5), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. apparatus.
  • the in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, (VII-1) to (VII) -The presentation device according to any one of 7).
  • the presentation device according to (VII-9) The presentation device according to (VII-8), wherein the engager is a bispecific molecule.
  • the bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (VII-9).
  • (VII-11) The presentation device according to any one of (VII-1) to (VII-10), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
  • the apparatus includes a processing unit, The processing unit is Peripheral blood mononuclear cells collected from the patient, directly or indirectly, to obtain an evaluation result of evaluating whether tumor cells contacted in vitro are injured, If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is useful to the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is not useful to the patient, if not indicated, Decision aid.
  • VIII-2 The usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and cannot resume administration,
  • the processing unit is When the evaluation result shows that the tumor cells are injured, the immunotherapeutic agent does not cause an adverse event for the patient, which requires discontinuation of treatment and resumption of administration is impossible. And/or the evaluation result indicates that the tumor cells are not injured, the immunotherapeutic agent requires treatment interruption to the patient and resumption of administration is not possible. Suggesting that a possible adverse event will occur, The determination assisting device according to (VIII-1).
  • VIII-3 The usefulness is whether or not the immunotherapeutic agent has an effect, The processing unit is If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is effective for the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is ineffective to the patient, The determination assisting device according to (VIII-1) or (VIII-2).
  • VIII-4 Whether the tumor cells contacted with the peripheral blood mononuclear cells are damaged or not, Obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, comparing the value reflecting the tumor cytotoxic activity with the corresponding reference value, the value from the corresponding reference value
  • the determination assisting device according to any one of (VII-1) to (VIII-3), which is evaluated according to whether or not it is high.
  • VIII-5) Evaluation of whether or not the value is higher than the corresponding reference value is Indicating that the tumor cells are injured when said value is higher than the corresponding reference value, and/or The decision aid of (VIII-4), comprising indicating that the tumor cells are not injured when the value is below the corresponding reference value.
  • VIII-6 The determination according to (VIII-5), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. Auxiliary device.
  • VIII-7) In (VIII-5), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measured value of interferon- ⁇ in the culture medium. The described decision aid device.
  • VIII-8) The in vitro contact is indirect contact of peripheral blood mononuclear cells collected from the patient with tumor cells via an engager, (VIII-1) to (VIII) -The determination assisting device according to any one of 7).
  • VIII-9 The determination aid device according to (VIII-8), wherein the engager is a bispecific molecule.
  • the bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells.
  • VIII-9 The determination assisting device according to any one of (VIII-1) to (VIII-10), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation
  • An immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2.
  • Use of immunotherapeutic agents given to the presented patient for postoperative adjuvant chemotherapy Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • (IX-7) Use of the immunotherapeutic agent according to any one of (IX-1) to (IX-6), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
  • the immunotherapy according to (IX-7), wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, or renal cell carcinoma, urothelial cancer, gastric cancer, malignant pleural mesothelioma, (IX-1) to (IX-8) Use of the immunotherapeutic agent according to any one of (1) to (4).
  • (IX-10) Use of the immunotherapeutic agent according to (IX-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapeutic agent.
  • an immunotherapeutic agent for treating a malignant tumor which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful in a method of presenting usefulness in a patient having the malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation
  • IX-12 Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following step 1 and step 2, the immunotherapeutic agent is useful in a method of presenting usefulness in a patient having the malignant tumor.
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (IX-11) or (IX-12) Use of the immune checkpoint inhibitor described in 1.).
  • the immune checkpoint inhibitor contains an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody (IX-11) to (IX-13) Use of the immune checkpoint inhibitor described in 1.).
  • Step 1 Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured
  • Step 2 the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment.
  • the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (IX-14) or (IX-15) Use of the immunotherapeutic agent described in (1).
  • PBMC mononuclear cells
  • FIG. 1 shows an outline of the present invention.
  • FIG. 2 shows an example of the outline of the presentation device and the determination assisting device.
  • FIG. 3 is an example of a block diagram showing a hardware configuration of a presentation device and a decision assisting device.
  • FIG. 4 is an example of a flowchart showing processing performed by the processing unit of the presentation device.
  • FIG. 5 is an example of a flowchart showing processing performed by the processing unit of the presentation device.
  • FIG. 6 is an example of a flowchart showing a process performed by the processing unit of the determination assisting device.
  • FIG. 7 is an example of a flowchart showing a process performed by the processing unit of the determination assisting device.
  • FIG. 8 shows an example of the configuration of a test reagent and a test kit.
  • FIG. 9 shows the tumor cytotoxic activity of lung cancer patient PBMC.
  • FIG. 10 shows the results of repertoire analysis of peripheral blood CD8-positive T cells and lung cancer tissue CD8-positive T cells.
  • FIG. 11 shows the correlation between the tumor cytotoxic activity of T cells in tumor (lung cancer) tissue and the amount of IFN ⁇ production by PBMC via the engager (FIG. 11A), or the tumor cytotoxic activity of peripheral blood T cells (FIG. 11B). It is a graph which shows a relationship.
  • FIG. 12 shows the correlation between the effect of nivolumab on T cells in tumor (lung cancer) tissue and the tumor cytotoxic activity of peripheral blood T cells (FIG. 12A), or the cutoff value of the tumor cytotoxic activity of peripheral blood T cells.
  • FIG. 12 shows the correlation between the effect of nivolumab on T cells in tumor (lung cancer) tissue and the tumor cytotoxic activity of peripheral blood T cells (FIG. 12A), or the cutoff value of the tumor cytotoxic activity of
  • FIG. 12B is a graph showing the effect of nivolumab on T cells in tumor (lung cancer) tissues of each group when the percentage is 28% (FIG. 12B).
  • A shows a ROC curve of percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC.
  • B shows an ROC curve of the positive rate of PD-L1 expressing cells. The comparison result of the tumor cell cytotoxic activity percentage of the peripheral blood T cell derived from PBMC and the positive rate of the PD-L1 expression cell in a tumor tissue is shown.
  • A Progression-free survival time of a patient group with a cut-off value of percent tumor cytotoxicity of 18.98% or more and a patient group of less than 18.98% in patients treated with pembrolizumab or nivolumab.
  • B Shows progression-free survival of patients with pembrolizumab or nivolumab who had PD-L1-expressing cells in the tumor tissue with a cutoff value of 55.00% or more and less than 55.00%.
  • C Shows progression-free survival in patients with pembrolizumab or nivolumab administered, with a cut-off value of PD-L1 expressing cells in the tumor tissue of 50.00% or more and less than 50.00%.
  • (A) Shows the progression-free survival of patients with pembrolizumab-treated patients with a cut-off value of percent tumor cytotoxicity of 18.98% or more and patients with a cutoff value of less than 18.98%.
  • (B) Shows progression-free survival in a group of patients with a positive cutoff value of PD-L1 expressing cells in tumor tissues of pembrolizumab-treated patients of 55.00% or more and a patient group of less than 55.00%.
  • A Progression-free survival of a patient group with a cutoff value of 18.98% or more and a patient group of less than 18.98% for the percent tumor cytotoxic activity in patients treated with nivolumab.
  • B Shows progression-free survival in a group of patients with a positive cutoff value of PD-L1 expressing cells in tumor tissues of nivolumab-treated patients of 55.00% or more and a patient group of less than 55.00%.
  • C The progression-free survival time of the patient group with a positive cutoff value of PD-L1 expressing cells in the tumor tissue of nivolumab-treated patients of 50.00% or more and the patient group of less than 50.00%.
  • (B) shows the ROC curve of the positive rate of PD-L1 expressing cells regarding the occurrence of adverse events. The correlation between the percentage of tumor cytotoxic activity of peripheral blood T cells and the concentration of interferon- ⁇ in the culture supernatant is shown.
  • FIG. 1 shows an example in which T cells contained in PBMCs are brought into contact with tumor cells that have been seeded and cultured in a petri dish in advance via an engager, and a value that reflects tumor cytotoxic activity is obtained.
  • the present embodiment specifically, it is evaluated whether or not the tumor cells contacted with the peripheral blood mononuclear cells collected from the patient directly or indirectly in vitro are injured.
  • the step of contacting the peripheral blood mononuclear cells collected from the patient with the tumor cells directly or indirectly in vitro (step 1), and the contact with the peripheral blood mononuclear cells in step 1 above
  • the step of evaluating whether the tumor cells are injured (step 2), it is evaluated whether the tumor cells are injured.
  • the present embodiment may be carried out by a human or may be carried out by the presentation device 10 or the auxiliary device 20 described later.
  • “Usefulness” means that the immunotherapeutic agent brings favorable results to the patient when treating a malignant tumor existing in the patient with the immunotherapeutic agent.
  • the usefulness can include, for example, the presence or absence of an adverse event resulting from the immunotherapeutic agent that requires treatment interruption and that makes it impossible to resume administration, and/or the therapeutic effect of the immunotherapeutic agent. ..
  • Adverse events are adverse events for patients that result from the administration of immunotherapeutic agents.
  • Adverse events include, for example, interstitial lung disease (preferably, if the patient has already developed interstitial pneumonia at the start of immunotherapeutic agent administration, the following adverse events including exacerbation of interstitial pneumonia).
  • intestinal disorders such as colitis, diarrhea, intestinal obstruction; epithelial tissue disorders such as mucocutaneous ocular syndrome; erythema multiforme; pemphigoid; neuropathy (peripheral neuropathy, Guillain-Barre syndrome, etc.) ); Hepatobiliary disorders such as liver dysfunction, hepatitis, and sclerosing cholangitis; thyroid dysfunction (symptomatic hypothyroidism, hyperthyroidism, thyroiditis, etc.), pituitary dysfunction (symptomatic pituitary gland) Endocrine disorders such as inflammation, symptomatic hypopituitarism) and adrenal dysfunction (symptomatic adrenal insufficiency, adrenal crisis, etc.); abnormal blood glucose levels such as type 1 diabetes (including fulminant type 1 diabetes) Renal disorders (renal insufficiency, renal interstitial nephritis, etc.); pancreatitis; myopathy such as myositis, rhabdomyolysis, myasthenia gravis, myos
  • An adverse event that requires treatment interruption (or treatment discontinuation) is an adverse event that requires treatment interruption (or treatment discontinuation) with an immunotherapeutic agent, such as interstitial lung disease (CTCAE v4.0 lung).
  • CTCAE v4.0 lung Pulmonary disorders such as Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5) of inflammation; colitis, diarrhea, bowel obstruction (CTCAE v4.0 colitis and diarrhea Grade 2, Grade 3, Grade 4), or Grade 5, bowel obstruction Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5 and other intestinal disorders; cutaneous mucosal-eye syndrome (Stevens-Johnson syndrome) and other epithelial tissue disorders (CTCAE v4.0 Grade 3) , Grade 4, or Grade 5); erythema multiforme (Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5 in CTCAE v4.0); pemphigoid; neuropathy (peripheral neuropathy, Guillain-Barre syndrome) Etc.) (Grade 2, Grade 3, Grade 4, or Grade 5 of neuropathy in CTCAE v4.0); liver
  • Adverse events that require discontinuation of treatment and incapable of resuming administration include treatment interruption (or discontinuation of treatment) with immunotherapeutic agents and resumption of administration of immunotherapeutic agents. It is an adverse event that makes it impossible to, for example, interstitial lung disease (Grade 1, Grade 2, Grade 3, or Grade 4 of pneumonitis in CTCAE v4.0 appears, and immunotherapeutic agent for malignant tumor Disorders such as discontinuation of administration of the drug or discontinuation of administration and coping therapy does not improve symptoms); colitis, diarrhea, (CTCAE v4.0 colitis/diarrhea Grade 2, Grade) 3 or Grade 4 symptoms appear, and even if the administration of the immunotherapeutic agent for malignant tumor is interrupted, or the symptoms do not improve to Grade 1 even if the administration is discontinued and coping therapy is performed), bowel obstruction, etc.
  • interstitial lung disease Grade 1, Grade 2, Grade 3, or Grade 4 of pneumonitis in CTCAE v4.0 appears
  • immunotherapeutic agent for malignant tumor Disorders such as discontinuation of administration of the drug
  • Cutaneous mucosal-eye syndrome (Stevens-Johnson syndrome) and other epithelial tissue disorders (Grade 3 or Grade 4 of rash in CTCAE v4.0 appear, and administration of immunotherapeutic agent for malignant tumor is discontinued. Or, if the symptoms do not improve to Grade 1 even after discontinuation of administration and coping therapy); erythema multiforme; pemphigus vulgaris; neuropathy (peripheral neuropathy, Guillain-Barre syndrome, etc.) (CTCAE v4.0 Symptoms of Grade 2, Grade 3, or Grade 4 of neuropathy appear, and even if the administration of the immunotherapeutic agent for malignant tumor is discontinued, or the treatment is discontinued and coping therapy is applied, the symptoms improve to the baseline state.
  • hepatic dysfunction, hepatitis, sclerosing cholangitis appear, and immunotherapeutic agent for malignant tumor is administered.
  • Hepatobiliary disorders such as hepatic function not improving to baseline even after discontinuation or discontinuation of administration and coping therapy; thyroid dysfunction (symptomatic hypothyroidism, symptomatic thyroid function) Symptoms such as hyperactivity and symptomatic thyroiditis appear, and the symptoms do not improve even if the administration of immunotherapeutic agents for malignant tumors is stopped, or if the treatment is stopped and coping therapy is performed), pituitary function
  • Endocrine disorders such as disorders (symptomatic pituitaryitis, symptomatic hypopituitarism, etc.), adrenal dysfunction (symptomatic adrenal insufficiency, adrenal crisis, etc.) appear, and immunotherapeutic agents for malignant tumors appear.
  • Symptoms do not improve after discontinuation of administration or discontinuation of administration and coping therapy Case: Abnormal blood glucose level of type 1 diabetes (including fulminant type 1 diabetes); renal disorder (renal failure, tubulointerstitial nephritis, etc.) (Grade 2, Grade 3, or acute renal disorder in CTCAE v4.0) Grade 4 symptoms appear, and if the administration of the immunotherapeutic agent for malignant tumor is discontinued, or if the treatment is discontinued and coping therapy is used, the symptoms do not improve to Grade 1); pancreatitis; myositis, striated muscle Myopathy such as lysis, myasthenia gravis, myocarditis; heart disorders; central nervous system disorders such as encephalitis, meningitis, encephalopathy, leukoencephalopathy; immune thrombocytopenic purpura, hemolytic anemia, erythroblasts Hematopoietic disorders such as lichen; toxic epidermal necrolysis (TEN); hypertension (Grade 2, Grade 3, Grade 4 or
  • the immunotherapeutic agent is not limited as long as it can treat a malignant tumor by increasing the damaging activity of T cells on tumor cells.
  • treating may include ameliorating, curing and/or preventing a malignant tumor.
  • treating is ameliorating and/or curing a malignant tumor.
  • treating includes ameliorating or curing a malignant tumor. Improving here includes suppressing the size reduction or expansion of malignant tumors, and/or suppressing the reduction or expansion of metastases.
  • Curing includes the disappearance of malignant tumor cells, preferably the absence of recurrence for more than 1 year, 2 years, 3 years, 5 years, or 7 years.
  • Preventing includes preventing recurrence of malignant tumors.
  • the recurrence may be a recurrence in the primary lesion of the malignant tumor or a recurrence in the metastatic lesion.
  • the immunotherapy agent in the present disclosure is not limited as long as it can be used for immunotherapy of malignant tumor.
  • examples of the immunotherapeutic agent include the immunotherapeutic agents described in "8. Immunotherapeutic agents" described later.
  • malignant tumors include epithelial malignant tumors and non-epithelial malignant tumors, preferably epithelial malignant tumors.
  • malignant tumors that a patient has include respiratory malignant tumors that develop from the trachea, bronchi, lungs, etc.; nasopharynx, esophagus, stomach, duodenum, jejunum, ileum, cecum, appendix, ascending colon, transverse colon, sigmoid Gastrointestinal malignant tumors arising from the colon, rectum or anus; liver cancer; pancreatic cancer; urinary system malignant tumors arising from the bladder, ureter or kidney; female reproductive system arising from ovaries, fallopian tubes, uterus, etc.
  • respiratory epithelial malignant tumor such as lung cancer (squamous cell cancer, small cell cancer, large cell cancer, adenocarcinoma); gastric cancer, duodenal cancer, colon cancer (S colon cancer, rectal cancer, etc.)
  • Gastrointestinal epithelial malignancies liver cancer; pancreatic cancer; bladder cancer; thyroid cancer; ovarian cancer; breast cancer: prostate cancer; head and neck cancer; malignant melanoma; renal cell carcinoma, urothelial cancer, classic Hodgkin lymphoma, Mention may be made of Merkel cell carcinoma or malignant pleural mesothelioma.
  • lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapeutic agent.
  • lung cancer includes squamous cell carcinoma, small cell carcinoma, large cell carcinoma or adenocarcinoma. Preferred is non-small cell lung cancer.
  • the “patient” is not limited as long as it is a person to whom an immunotherapy agent can be applied.
  • a patient a patient who has any of the malignant tumors described above and is untreated, a patient who has already been treated for some malignant tumor, a patient who is undergoing treatment for a malignant tumor, or a malignant tumor has recurred. Or a patient with metastasis can be mentioned.
  • the above-mentioned treatment of malignant tumor preferably includes surgical tumor resection, chemotherapy (more preferably, chemotherapy by administration of an anti-cancer agent other than immunotherapeutic agent), radiation therapy and the like.
  • the patient is also preferably dependent on the application of the administered immunotherapeutic agent, eg, non-small cell carcinoma malignancies; unresectable, advanced non-small cell carcinoma malignancies, and/or recurrent Non-small cell malignancies; unresectable malignant melanoma; unresectable or metastatic renal cell carcinoma); relapsed or refractory classic Hodgkin lymphoma; head and neck cancer with recurrent or distant metastases; cancer chemistry Unresectable advanced or recurrent gastric cancer that worsened after therapy; unresectable advanced or recurrent malignant pleural splenomegaly that worsened after cancer chemotherapy; unresectable urothelial cancer that worsened after cancer chemotherapy; unresectable Merkel cell carcinoma).
  • immunotherapeutic agent eg, non-small cell carcinoma malignancies; unresectable, advanced non-small cell carcinoma malignancies, and/or recurrent Non-small cell malignancies; unresectable malignant melanoma
  • peripheral blood is not limited as long as mononuclear cells including T cells can be obtained.
  • the peripheral blood may be arterial blood or venous blood, but is preferably venous blood.
  • Peripheral blood can be collected, for example, from the blood vessels of the extremities.
  • blood may be collected from blood vessels other than the extremities at the time of surgery or biopsy.
  • the method of collecting peripheral blood is not limited as long as mononuclear cells can be obtained, but it is preferable to collect blood using an anticoagulant.
  • the anticoagulant include ethylenediaminetetraacetate, citrate, heparin salt and the like. Heparin is preferred.
  • -PBMC can be obtained according to a known method. For example, it can be obtained by centrifugation using a specific gravity liquid for human lymphocyte separation (having a density of about 1.077 ⁇ 0.001 g/ml).
  • the red blood cells in the peripheral blood may be hemolyzed using a hemolytic agent, and the resulting nucleated cells may be used as PBMC.
  • desired cells may be obtained using a cell sorter or magnetic cell separation method. Examples of the desired cells include T cells, preferably CD3 positive T cells or CD8 positive T cells.
  • the obtained PBMC is preferably brought into contact with tumor cells without undergoing a step such as culturing after the acquisition. For example, within 24 hours, within 18 hours, within 12 hours, within 6 hours, within 4 hours, within 3 hours, within 2 hours, within 1 hour, within 30 minutes, or within 15 minutes after obtaining PBMC. Contacting with tumor cells is preferred.
  • the tumor cells that come into contact with PBMC are not limited as long as they are cells derived from the above tumor.
  • a preferable example of the tumor cell is a cultured cell line.
  • the cultured cell line is not particularly limited, and examples thereof include cultured cell lines derived from malignant tumors such as U251 cell line, RERF cell line, and A549 cell line.
  • the tumor cells do not have to be cells of the same species as the malignant tumor of the patient, but may be cells of the same species.
  • the cells can grow under the same culture conditions as PBMC.
  • the contact method between PBMC and tumor cells in vitro is not limited as long as PBMC and tumor cells directly or indirectly contact each other.
  • the tumor cells When contacting PBMCs with tumor cells in vitro, it is preferable to seed the tumor cells in vitro on a plate or culture bottle in advance.
  • the tumor cells are adhesive cells, it is more preferable that the tumor cells are in a viable state in vitro and adhere to a petri dish or a culture bottle.
  • the tumor cells can grow for 12 hours to 48 hours under conditions (eg, 35° C. to 37° C. in the presence of about 5% carbon dioxide gas). It is preferably in a state of being cultured to some extent.
  • the culture medium for culturing the tumor cells is not limited as long as the tumor cells can survive or develop. For example, RPMI1640 medium (including modified medium), ⁇ -MEM medium (including modified medium), Dulbecco MEM medium (including modified medium), etc. containing about 8% to 20% fetal bovine serum can be mentioned.
  • the number of PBMC cells when contacting PBMCs with tumor cells is, for example, about 1 ⁇ 10 3 to 1 ⁇ 10 5 , preferably about 5 ⁇ 10 3 to 5 ⁇ 10 4 cells per well of a 96-well plate. It is preferable that the tumor cells are brought into contact with PBMC at about 1 ⁇ 10 3 to 1 ⁇ 10 5 , preferably about 5 ⁇ 10 3 to 5 ⁇ 10 4 .
  • a method of directly contacting PBMC with tumor cells a method of seeding PBMC on a plate or culture bottle seeded with tumor cells can be mentioned.
  • the seeded PBMC are floating, then 1,000 to 2,000 r.p.m. p. m. It can be centrifuged for about 5 to 10 minutes.
  • engager As a method for indirectly contacting PBMC with tumor cells, a method for contacting them via an engager can be mentioned.
  • the term "engager” is limited as long as it is a molecule capable of engaging PBMC, preferably lymphocytes, more preferably T cells, and even more preferably CD8-positive T cells (cytotoxic T cells) with tumor cells.
  • Engage means cross-linking at least cells (for example, T cells and tumor cells), preferably binding T cells and tumor cells via an engager, and preferably cytotoxic T cells Means a state capable of exhibiting cytotoxic activity against cross-linked tumor cells.
  • a bispecific molecule, a trispecific molecule and the like can be mentioned.
  • a bispecific molecule is more preferable as an engager.
  • the bispecific molecule and the trispecific molecule are molecules containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. preferable.
  • the antigen-binding region is not limited as long as it binds to the target antigen.
  • the antigen-binding region is, for example, at least one of the group consisting of complementary chain determining region 1 (CDR1), complementary chain determining region 2 (CDR2) and complementary chain determining region 3 (CDR3) of immunoglobulin that binds to the antigen. Includes CDR.
  • the antigen-binding region preferably comprises at least two CDRs consisting of CDR1, CDR2 and CDR3, more preferably CDR1, CDR2 and CDR3.
  • the CDR may be derived from an immunoglobulin heavy chain or an immunoglobulin light chain. More preferably, in the antigen-binding region, each CDR may be adjacent to the corresponding framework region.
  • the antigen-binding region may be an immunoglobulin variable region (V region), sc-Fv, an immunoglobulin Fab region, or the like.
  • the number of antigen-binding regions that bind to at least one surface antigen may be one or two or more for one antigen.
  • an antigen-binding region that binds to at least one surface antigen is a combination of Fab regions of the same species, a combination of variable regions of the same species, a combination of sc-Fv of the same species, a heavy chain Fab region derived from one antibody, and a light chain Fab region.
  • Combination of chain Fab regions Combination of heavy chain variable region and light chain variable region derived from one type of antibody may be a combination of heavy chain sc-Fv derived from one type of antibody.
  • the surface antigen of T cell is not limited as long as it exists on the surface of T cell and can bind to at least one antigen-binding region contained in the bispecific molecule or trispecific molecule.
  • the T cell surface antigen include cytotoxic T cell surface antigens.
  • T cell surface antigens include CD3, CD8, TCR, CTLA-4, PD1, Tim3, CD27, CD28, CD40, CD134 (OX40), CD137 (4-1BB), CD278 (ICOS). .. It is preferably CD3.
  • the tumor cell surface antigen is not limited as long as it exists on the surface of the tumor cell and can bind to at least one antigen-binding region contained in the bispecific molecule or the trispecific molecule.
  • EphA1 ephrin type-A receptor 1
  • EphA2 FolR1 (folate receptor 1)
  • EpCAM Epidermal cellulae berbereion molecule
  • CD19 Herb 1-herb (2).
  • HLA-A1+NY-ESO1 HLA-A1 resty-NY-HLA-A1+HN-ESO1+HLA-A2+HLA-A2+HLA-A2+HLA-A2+HLA-A2).
  • HLA-A3+NY-ESO1 HLA-A3 restricted NY-ESO1 and the like.
  • Bispecific molecules include bispecific T cell engagers, bispecific antibodies, etc.
  • Examples of bispecific T cell engagers include those that target CD19 and CD3, and those that target EphA2 and CD3.
  • Examples of bispecific antibodies include antibodies targeting EpCAM and CD3.
  • the number of cells when contacting PBMCs with tumor cells is, for example, about 1 ⁇ 10 3 to 1 ⁇ 10 5 cells, preferably about 5 ⁇ 10 3 to 5 ⁇ 10 4 tumors per well of a 96-well plate.
  • the cells are preferably contacted with about 1 ⁇ 10 3 to 1 ⁇ 10 5 PBMCs, preferably about 5 ⁇ 10 3 to 5 ⁇ 10 4 PBMCs.
  • the bispecific molecule When the bispecific molecule is added, it is preferable to add it at a final concentration of 50 to 200 ng/ml per well of a 96-well plate.
  • Direct or indirect contact between PBMC and tumor cells is preferably performed under conditions in which PBMC and tumor cells can normally develop. For example, it can be performed for 16 hours to 72 hours, preferably for 36 hours to 50 hours under conditions that allow growth (eg, 35° C. to 37° C. in the presence of 5% carbon dioxide gas).
  • the culture medium for contacting the PBMC with the tumor cells is not limited as long as the culture medium allows the PBMC and the tumor cells to survive or develop.
  • RPMI1640 medium including modified medium
  • fetal bovine serum can be mentioned.
  • Assessment of whether or not the tumor cells are injured includes assessing that the tumor cells are injured and/or that the tumor cells are not injured.
  • assessing whether the tumor cells are injured comprises determining whether the tumor cells are injured. Determining whether the tumor cells are injured includes determining that the tumor cells are injured and/or determining that the tumor cells are not injured.
  • the evaluation result of whether the tumor cells are injured as information indicating whether the tumor cells are injured, a value reflecting the cytotoxic activity against the tumor cells described below (tumor cytotoxic activity), Information indicating that the tumor cells are injured based on the reference value corresponding to the above value (for example, “injury present”, “injury positive”, “+ (plus)”, etc.), or tumor cells are injured It includes information indicating that it has not been done (for example, "no injury”, “negative injury”, "-(minus)", etc.).
  • the tumor cells are injured, obtaining information indicating whether the tumor cells are injured, and based on the obtained information, the tumor cells are injured And/or assessing that the tumor cells are not injured.
  • the peripheral blood mononuclear cells collected from the patient are directly or indirectly contacted with the tumor cells in vitro, and then the tumor cells are A value that reflects the tumor cytotoxic activity of contacted peripheral blood mononuclear cells is obtained.
  • the value that reflects the acquired tumor cytotoxic activity is compared with its corresponding reference value. As a result of the comparison, when the value reflecting the tumor cytotoxic activity is higher than the corresponding reference value, it indicates that the tumor cells are injured. As a result of the comparison, when the value reflecting the tumor cell cytotoxicity is lower than the corresponding reference value, it indicates that the tumor cells are not injured.
  • the above-mentioned reference value is not limited as long as it can determine whether or not tumor cells are injured.
  • the reference value is obtained from a value that reflects the tumor cytotoxic activity of the negative control PBMC in which tumor immunity is not activated, or a value that reflects the tumor cytotoxic activity of the positive control PBMC in which tumor immunity is activated. be able to.
  • the method for measuring the value reflecting the tumor cytotoxic activity of negative control PBMC and the value reflecting the tumor cytotoxic activity of positive control PBMC is performed by measuring the value reflecting the tumor cytotoxic activity of PBMC of the patient to be treated. The method is preferably the same as the method described above.
  • the reference value is the upper limit of the value that reflects the tumor cytotoxic activity of the negative control PBMC, the value that reflects the tumor cytotoxic activity of the positive control PBMC, and the value that reflects the tumor cytotoxic activity of the negative control PBMC and is positive.
  • the median, mean, mode, etc. of the values reflecting the tumor cytotoxic activity of the control PBMC can be used.
  • the reference value may be calculated by a ROC curve (Receiver Operating Characteristic curve, receiver operating characteristic curve), discriminant analysis method, mode method, Kittler method, 3 ⁇ method, p-tile method, or the like.
  • the reference value is a value that reflects the tumor cytotoxic activity, and for example, when the value of the tumor cytotoxic activity described later is used, the reference value is based on the number of living cells or the number of dead cells.
  • the percent activity may be selected from 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50%.
  • the reference value may or may not be the same in the case of deciding the presence or absence of an adverse event that requires discontinuation of treatment and in which resumption of administration is impossible, and the case of deciding the presence or absence of a therapeutic effect. Good.
  • the reference value for determining the presence or absence of the therapeutic effect may be selected from 10%, 15%, 20%, 25%, 30%, 35% or 40%.
  • the reference value is a value that reflects the tumor cytotoxicity.
  • the reference value is a value that reflects the tumor cytotoxicity.
  • the reference value may or may not be the same in the case of deciding the presence or absence of an adverse event that requires discontinuation of treatment and in which resumption of administration is impossible, and the case of deciding the presence or absence of a therapeutic effect. Good. If the reference values are different, the reference values for determining the presence or absence of adverse events that require discontinuation of treatment and incapable of resuming administration are 3,500 pg/ml, 4,000 pg/ml, 4,500 pg/ml. It may be selected from ml, 5,000 pg/ml, 5,500 pg/ml, 6,000 pg/ml, or 6,500 pg/ml.
  • the reference value for determining the presence or absence of the therapeutic effect is 2,000 pg/ml, 2,500 pg/ml, 3,000 pg/ml, 3,500 pg/ml, 4,000 pg/ml, 4 , 500 pg/ml, 5,000 pg/ml, 5,500 pg/ml, 6,000 pg/ml, or 6,500 pg/ml.
  • the value that reflects the cytotoxic activity on tumor cells may include, for example, the value of tumor cytotoxic activity or the measured value of interferon- ⁇ .
  • the value of the tumor cytotoxic activity can be determined by measuring the number of viable tumor cells after contact with the PBMC and calculating the tumor cytotoxic activity from the measurement result.
  • the method for measuring the viable cell number of tumor cells is not limited as long as it can evaluate how much PBMC can injure the tumor cells. For example, in the case of directly contacting PBMC with tumor cells, the viable cell number of the tumor cells immediately after contacting PBMC with the tumor cells and the tumor cell 48 hours after contacting PBMC with the tumor cells It can be determined from the value of the number of viable cells.
  • a well to which the engager is added when contacting PBMC and tumor cells
  • PBMC and tumor cells To create a well (negative control well) to which no engager is added when contacting, and obtain the value of tumor cytotoxic activity based on the number of viable cells in the measurement well and the number of viable cells in the negative control well
  • the value of the tumor cytotoxic activity can be determined by the following formula as a percentage (%) of the tumor cytotoxic activity based on living cells.
  • a viable cell staining reagent such as CellTiter96 (registered trademark) AQueousOne Solution Reagent (MTS reagent: Promega)
  • change the viable cell number to The percentage of tumor cell cytotoxic activity based on live cells may be calculated based on the absorbance.
  • the percentage of tumor cytotoxicity may be calculated based on dead cells in place of the above-mentioned number of viable cells, and this value may be used as the value of tumor cytotoxicity.
  • the tumor cytotoxic activity percentage % based on dead cells, for example, the method described below The percent tumor cytotoxic activity can be calculated.
  • a well to which an engager is added when making contact with PBMC and tumor cells (measurement well), and a well to which no engager is added when making contact with PBMC and tumor cells (negative control well)
  • a well (positive control well) to which a substance having a high cell-killing ability such as hydrogen peroxide was added when PBMCs were brought into contact with tumor cells was prepared, and the number of dead cells in the measurement well and the number of dead cells in the negative control well were measured.
  • the value of tumor cytotoxic activity can be obtained based on the number of dead cells in the positive control wells. In this case, the value of the tumor cytotoxic activity can be calculated by the following formula as a percentage (%) of the tumor cytotoxic activity based on dead cells.
  • the measurement value of interferon- ⁇ can be obtained by, for example, collecting a culture medium from the well after contacting PBMC with tumor cells, and measuring the protein concentration (or amount) of interferon- ⁇ in the culture medium by a known ELISA method or the like. Can be obtained by measuring using.
  • an anti-interferon- ⁇ antibody for capturing an antigen is previously immobilized on a solid phase such as a microplate to form a complex of the immobilized anti-interferon- ⁇ antibody and interferon- ⁇ in a sample.
  • a solid phase such as a microplate
  • the method of immobilizing the anti-interferon- ⁇ antibody for capturing the antigen on the solid phase is not particularly limited. It can be done directly using known methods or indirectly through another substance. Examples of the direct bond include physical adsorption. Preferably, the anti-interferon- ⁇ antibody can be physically bound directly to the microplate using an immunoplate or the like.
  • the solid phase material is not particularly limited, and examples thereof include polystyrene and polypropylene.
  • the shape of the solid phase is not particularly limited, and examples thereof include a microplate, a microtube, a test tube, and beads.
  • the method may include an operation of washing the solid phase subsequent to the formation of the complex.
  • PBS containing a surfactant or the like can be used.
  • the detection of the complex is carried out using an anti-interferon- ⁇ antibody for detection labeled with a labeling substance, or an unlabeled anti-interferon- ⁇ antibody and the unlabeled anti-interferon- ⁇ antibody are used. It can be carried out using an anti-immunoglobulin antibody labeled with a labeling substance capable of binding, but it is preferable to use a labeled anti-interferon- ⁇ antibody for detection. Further, it is preferable that the epitope of interferon- ⁇ of the anti-interferon- ⁇ antibody for detection is different from the epitope of interferon- ⁇ of the anti-interferon- ⁇ antibody for antigen capture.
  • the labeling substance used for the anti-interferon- ⁇ antibody for detection or the labeled anti-immunoglobulin antibody is not particularly limited as long as it produces a detectable signal.
  • fluorescent substances, radioisotopes, enzymes and the like can be mentioned.
  • the enzyme include alkaline phosphatase and peroxidase.
  • the fluorescent substance include fluorescent dyes such as fluorescein isothiocyanate (FITC), rhodamine, and Alexa Fluor (registered trademark), and fluorescent proteins such as GFP.
  • FITC fluorescein isothiocyanate
  • Alexa Fluor registered trademark
  • fluorescent proteins such as GFP.
  • radioisotope include 125 I, 14 C, 32 P and the like. Among them, alkaline phosphatase or peroxidase is preferable as the labeling substance.
  • the anti-interferon- ⁇ antibody for detection can be obtained by labeling the anti-interferon- ⁇ antibody with the above-mentioned labeling substance by a labeling method known in the art. Alternatively, labeling may be performed using a commercially available labeling kit or the like.
  • the labeled immunoglobulin antibody may be the same as that used for labeling the anti-interferon- ⁇ antibody, or may be a commercially available one.
  • the measured value of interferon- ⁇ contained in the sample can be obtained by detecting the signal generated by the labeled substance of the labeled anti-interferon- ⁇ antibody contained in the complex.
  • detecting a signal includes qualitatively detecting the presence or absence of a signal, quantifying the signal intensity, and semi-quantitatively detecting the signal intensity.
  • Semi-quantitative detection means indicating the intensity of a signal stepwise such as “no signal is generated", “weak”, “medium”, “strong". In this step, it is preferable to detect the signal intensity quantitatively or semi-quantitatively.
  • a known method can be used as a method for detecting a signal.
  • a measuring method can be appropriately selected according to the type of signal derived from the above-mentioned labeling substance.
  • the labeling substance is an enzyme
  • light generated by reacting a substrate for the enzyme a signal such as color
  • a known device such as a luminometer and a spectrophotometer. it can.
  • the substrate for the enzyme can be appropriately selected from known substrates according to the type of the enzyme.
  • the substrate is CDP-Star (registered trademark) (4-chloro-3-(methoxyspiro[1,2-dioxetane-3,2'-(5'-chloro)tricloxyl
  • a chemiluminescent substrate such as [3.3.1.13,7]decane]-4-yl)phenyl sodium phosphate), 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 5- Color forming substrates such as disodium bromo-6-chloro-indolyl phosphate and p-nitrophenyl phosphate can be mentioned.
  • TMB tetramethylbenzidine
  • the labeled substance is a radioisotope
  • radiation as a signal can be measured using a known device such as a scintillation counter.
  • the labeling substance is a fluorescent substance
  • the fluorescence as a signal can be measured using a known device such as a fluorescence microplate reader.
  • the excitation wavelength and the fluorescence wavelength can be appropriately determined according to the type of fluorescent substance used.
  • the signal detection result can be used as a measurement value for interferon- ⁇ .
  • the signal intensity measurement value itself or a value calculated from the signal intensity measurement value can be used as the interferon- ⁇ protein measurement value.
  • the presenting method comprises a step (step 1) of assessing whether tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured (step 1); In the step of presenting that the immunotherapeutic agent is useful to the patient when it is shown that the tumor cells are injured, and/or the tumor cells are not injured in the evaluation. If indicated, the step of presenting to the patient that the immunotherapeutic agent is not useful may be included (step 2).
  • Step 1 is the above 1. Is as described in.
  • step 2 “present” is intended to be shown so that a person can understand it.
  • the step 2 more specifically includes the evaluation.
  • the immunotherapeutic agent does not cause an adverse event in the patient, which requires treatment interruption and makes it impossible to resume administration. If the step of presenting and/or the evaluation shows that the tumor cells are not injured, the immunotherapeutic agent is required to be interrupted to the patient and the administration is not restarted. Presenting that a possible adverse event will occur. To present that the treatment needs to be interrupted and does not cause an adverse event that makes it impossible to resume the administration, for example, on a paper medium or a display screen, the treatment must be interrupted and the administration can be restarted.
  • the step 2 comprises the step of immunizing the patient when the tumor cells are shown to be injured in the evaluation. Indicating that the immunotherapeutic agent is ineffective to the patient when presenting that the therapeutic agent is effective and/or when the evaluation indicates that the tumor cells are not injured. And presenting.
  • information indicating that the immunotherapeutic agent is effective is displayed on a paper medium or a display screen (for example, “effective”, “ ⁇ ”, “+( Plus)” and the like).
  • the immunotherapeutic agent is invalid, for example, information indicating that the immunotherapeutic agent is invalid is displayed on a paper medium or a display screen (for example, "invalid", "x", "-( Minus)” etc.) is included or displayed.
  • Aiding utility includes assisting a physician in determining utility.
  • the present embodiment may be carried out by a human or may be carried out in the auxiliary device 10 described later.
  • the assisting method comprises a step (step i) of assessing whether tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured, and the assessment.
  • step ii the step of presenting that the immunotherapeutic agent is useful to the patient when it is shown that the tumor cells are injured, and/or the tumor cells are not injured in the evaluation. If indicated, the step of presenting to the patient that the immunotherapeutic agent is not useful may be included (step ii).
  • Step i is 1. Is as described in.
  • step ii “presenting” is intended to be shown so that a person can understand it.
  • the step ii more specifically, the evaluation.
  • the immunotherapeutic agent does not cause an adverse event in the patient, which requires treatment interruption and makes it impossible to resume administration. If the step of presenting and/or the evaluation shows that the tumor cells are not injured, the immunotherapeutic agent is required to be interrupted to the patient and the administration is not restarted. Presenting that a possible adverse event will occur.
  • the treatment needs to be interrupted and does not cause an adverse event that makes it impossible to resume the administration, for example, on a paper medium or a display screen
  • the treatment must be interrupted and the administration can be restarted.
  • This includes describing or displaying information (for example, "none”, “negative”, “-(minus)”, etc.) indicating that no adverse adverse events will occur.
  • an adverse event that requires the treatment interruption and makes it impossible to resume the administration for example, on a paper medium or a display screen
  • the treatment interruption is required and the administration restart is not possible.
  • the step ii includes the step of immunizing the patient when the tumor cells are shown to be injured. Indicating that the immunotherapeutic agent is ineffective to the patient when presenting that the therapeutic agent is effective and/or when the evaluation indicates that the tumor cells are not injured. And presenting.
  • information indicating that the immunotherapeutic agent is effective is displayed on a paper medium or a display screen (for example, “effective”, “ ⁇ ”, “+( Plus)” and the like).
  • information indicating that the immunotherapeutic agent is invalid is displayed on a paper medium or a display screen (for example, "invalid", "x", "-( Minus)” etc.) is included or displayed.
  • a presenting device 10 (hereinafter, also referred to as a presenting device 10).
  • the presentation device 10 may be connected to the input unit 111, the output unit 112, and the storage medium 113. Further, it may be connected to the measurement unit 30 configured by a microplate reader or the like. That is, the presentation device 10 includes a presentation system 50 that is connected to the measurement unit 30 directly or via a network or the like, and presents the utility of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor. It may be configured.
  • a processing unit (CPU) 101, a main storage unit 102, a ROM (read only memory) 103, an auxiliary storage unit 104, a communication interface (I/F) 105, and an input interface (I/F). ) 106, an output interface (I/F) 107, and a media interface (I/F) 108 are connected to each other by a bus 109 so that data communication is possible.
  • the main storage unit 102 and the auxiliary storage unit 104 may be collectively referred to as a storage unit.
  • the CPU 101 is the processing unit 101 of the presentation device 10.
  • the CPU 101 executes the computer program stored in the auxiliary storage unit 104 or the ROM 103 and processes the acquired data, so that the presentation device 10 functions.
  • the ROM 103 is composed of a mask ROM, a PROM, an EPROM, an EEPROM, etc., and stores a computer program executed by the CPU 101 and data used for the computer program.
  • the CPU 101 may be the MPU 101.
  • the ROM 103 stores a boot program executed by the CPU 101 when the presentation device 10 is activated and programs and settings related to the operation of the hardware of the presentation device 10.
  • the main storage unit 102 is configured by a RAM (Random access memory) such as SRAM or DRAM.
  • the main storage unit 102 is used to read the computer programs recorded in the ROM 103 and the auxiliary storage unit 104. Further, the main storage unit 102 is used as a work area when the CPU 101 executes these computer programs. Furthermore, the main storage unit 102 may temporarily store the evaluation result obtained by the processing unit 101, which indicates whether or not the tumor cells are injured. Further, when the reference values are provided from the network or the auxiliary storage unit 104, these may be temporarily stored.
  • the auxiliary storage unit 104 includes a hard disk, a semiconductor memory device such as a flash memory, an optical disk, and the like.
  • the auxiliary storage unit 104 stores various computer programs to be executed by the CPU 101, such as an operating system and application programs, and various setting data used for executing the computer programs. Specifically, the reference value and the like may be stored in a non-volatile manner.
  • the communication I/F 105 includes a serial interface such as USB, IEEE1394, RS-232C, a parallel interface such as SCSI, IDE, and IEEE1284, an analog interface including a D/A converter and an A/D converter, a network interface controller ( Network interface controller (NIC), etc.
  • the communication I/F 105 receives data from the measurement unit 30 or another external device, and transmits information stored or generated by the presentation device 10 to the measurement unit 30 or the outside as necessary. indicate.
  • the communication I/F 105 may communicate with the measurement unit 30 or another external device via a network. When the reference value is provided from the network, the communication I/F 105 receives the reference value under the control of the CPU 101.
  • the input I/F 106 is composed of, for example, a serial interface such as USB, IEEE1394, RS-232C, a parallel interface such as SCSI, IDE, IEEE1284, and an analog interface including a D/A converter and an A/D converter. It The input I/F 106 receives character input, click, voice input, and the like from the input unit 111. The received input content is stored in the main storage unit 102 or the auxiliary storage unit 104.
  • the input unit 111 includes a touch panel, a keyboard, a mouse, a pen tablet, a microphone, and the like, and performs character input or voice input to the presentation device 10.
  • the input unit 111 may be connected from the outside of the presentation device 10 or integrated with the presentation device 10.
  • the output I/F 107 is composed of the same interface as the input I/F 106, for example.
  • the output I/F 107 outputs the information generated by the CPU 101 to the output unit 112.
  • the output I/F 107 outputs the information generated by the CPU 101 and stored in the auxiliary storage unit 104 to the output unit 112.
  • the output unit 112 is composed of, for example, a display, a printer, and the like, and evaluates the measurement results transmitted from the measurement unit 30, various operation windows in the presentation device 10, and whether or not tumor cells are injured, and presentation of usefulness. Display contents etc.
  • the media I/F 108 reads out, for example, application software stored in the storage medium 113.
  • the read application software and the like are stored in the main storage unit 102 or the auxiliary storage unit 104.
  • the media I/F 108 also writes the information generated by the CPU 101 in the storage medium 113.
  • the media I/F 108 writes the information generated by the CPU 101 and stored in the auxiliary storage unit 104 into the storage medium 113.
  • the storage medium 113 is composed of a flexible disk, a CD-ROM, a DVD-ROM, or the like.
  • the storage medium 113 is connected to the media I/F 108 by a flexible disk drive, a CD-ROM drive, a DVD-ROM drive, or the like.
  • the storage medium 113 may store an application program or the like for a computer to execute an operation.
  • the CPU 101 may acquire application software and various settings necessary for controlling the presentation device 10 via the network instead of reading from the ROM 103 or the auxiliary storage unit 104.
  • the application program is stored in the auxiliary storage unit of the server computer on the network, and the presentation device 10 accesses the server computer to download the computer program and store the computer program in the ROM 103 or the auxiliary storage unit 104. It is possible.
  • the ROM 103 or the auxiliary storage unit 104 an operating system that provides a graphical user interface environment such as Windows (registered trademark) manufactured and sold by Microsoft Corporation in the United States is installed.
  • the application program according to the second embodiment operates on the operating system. That is, the presentation device 10 may be a personal computer or the like.
  • the operation of the presentation device 10 is performed by the processing unit 101 of the presentation device 10 according to a command of a computer program for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor described below in a patient having the malignant tumor. Control.
  • the processing unit 101 is input by the inspector from the input unit 111, or the processing unit 101 is controlled by the inspector and the above 1.
  • the peripheral blood mononuclear cells collected from the patient according to the method described in 1. above are evaluated as to whether or not the tumor cells contacted directly or indirectly in vitro are injured, and the evaluation result is obtained (step S11). ).
  • the processing unit 101 uses the above 1. It is determined whether the evaluation result indicates that the tumor cells are damaged according to the method described in (step 12).
  • step 12 when the evaluation result indicates that the tumor cells are injured (YES), the processing unit 101 proceeds to step S13, determines that the immunotherapeutic agent is useful, and the result is Is presented from the output unit 112.
  • step S12 when the evaluation result indicates that the tumor cells are not injured (NO), the processing unit 101 proceeds to step S14, determines that the immunotherapeutic agent is not useful, and outputs the result. It is presented by outputting from the output unit 112.
  • the processing unit 101 determines in Step S12 that the evaluation result is If it is indicated that the tumor cells are injured (YES), in step S13, the immunotherapeutic agent causes an adverse event for the patient, in which the treatment needs to be interrupted and the administration cannot be restarted. Show that you don't.
  • the processing unit 101 determines in Step S12 that the evaluation result is In the case of (NO) indicating that the tumor cells have not been injured, in step S14, the immunotherapeutic agent causes an adverse event in the patient, which requires treatment interruption and cannot resume administration. Offer to wake up.
  • the processing unit 101 determines in Step S12 that the evaluation result is If the tumor cells are not injured (NO), step S14 is presented to the patient that the immunotherapeutic agent is ineffective.
  • the processing unit 101 may record the result obtained in step S13 or step S14 in the recording medium 113.
  • the processing unit 101 is input by the inspector from the input unit 111.
  • a value that reflects the tumor cytotoxic activity obtained by the method described in (1) is obtained (step S101).
  • the information for calculating the tumor cytotoxic activity is acquired from the measurement unit 30.
  • the information is information that reflects the number of living cells or dead cells of the tumor cells that have come into contact with PBMC, and is, for example, the absorbance of a living cell staining reagent or a dead cell measurement reagent.
  • the information is the absorbance when interferon- ⁇ is measured.
  • the processing unit 101 compares the value reflecting the tumor cell cytotoxic activity acquired in step S101 with the reference value stored in the main storage unit 102 or the auxiliary storage unit 104 (step S102).
  • the processing unit 101 determines whether the value reflecting the tumor cell cytotoxic activity acquired in step S101 is higher than the reference value based on the comparison result in step S102 (step S103).
  • step S103 determines in step S103 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES)
  • the processing unit 101 proceeds to step S104, determines that the immunotherapeutic agent is useful, and Is presented from the output unit 112.
  • step S105 determines that the immunotherapeutic agent is not useful, and Is presented from the output unit 112.
  • the processing unit 101 determines the tumor cytotoxic activity in step S103.
  • the processing unit 101 determines the tumor cytotoxic activity in step S103.
  • the immunotherapeutic agent causes an adverse event for the patient, which requires treatment interruption and resumption of administration is impossible. Offer to wake up.
  • Step S103 determines in Step S103 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES), Step S104. In, it is shown to the patient that the immunotherapeutic agent is effective.
  • the processing unit 101 in step S103, the evaluation result is In the case of indicating that the tumor cells are not injured (NO), when it is determined that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), in step S105, the Present that the immunotherapeutic is ineffective.
  • the processing unit 101 may record the result obtained in step S104 or step S105 in the recording medium 113.
  • a computer program for controlling a device for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor and a storage medium storing the computer program according to an embodiment disclosed herein.
  • the present invention relates to a computer program for presenting the utility of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor.
  • the immunotherapeutic agent for treating the malignant tumor described in 1. it is a computer program for controlling the operation of the presentation device that presents the usefulness in the patient having the malignant tumor.
  • an embodiment disclosed in the present specification relates to a storage medium storing the computer program. That is, the computer program is stored in a storage medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program in the storage medium is not limited as long as the presentation device can read the program.
  • the storage in the storage medium is preferably non-volatile.
  • the device 20 (hereinafter, also referred to as the auxiliary device 20) that assists in determining
  • auxiliary device 20 An example of the hardware configuration of the auxiliary device 20 is the same as that of the presentation device 10 shown in FIGS. 2 and 3.
  • the auxiliary device 20 may be connected to the measurement unit 30 configured by a microplate reader or the like. That is, the auxiliary device 20 assists the determination of the usefulness of the immunotherapeutic agent for treating a malignant tumor, which is connected to the measurement unit 30 directly or via a network or the like, in a patient having the malignant tumor. 50 may be configured.
  • the description of the presentation device 10 is incorporated here.
  • the processing unit (CPU) 101, the main storage unit 102, the ROM 103, the auxiliary storage unit 104, the communication interface (I/F) 105, the input interface (I/F) 106, and the output interface (I) in the description of the presentation device 10 are described.
  • processing unit (CPU) 201 main storage unit 202, ROM 203, auxiliary storage unit 204, communication interface (I/F) 205, input interface (I/F) 206, output interface (I/F) 207, media interface (I/F) 208, bus 209, input unit 211, It should be read as the output unit 212 and the storage medium 213.
  • the operation of the assisting device 20 is performed by the processing unit 201 of the assisting device 20 according to a command of a computer program for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor described below in a patient having the malignant tumor. Control.
  • the processing unit 201 is input by the inspector from the input unit 211, or the processing unit 201 is controlled by the inspector and the above 1.
  • the peripheral blood mononuclear cells collected from the patient are directly or indirectly contacted with the tumor cells according to the method described in 1. to evaluate whether the tumor cells are injured, and obtain the evaluation result (step S21). ).
  • the processing unit 201 uses the above 1. It is determined whether the evaluation result shows that the tumor cells are injured according to the method described in (22).
  • step 22 if the evaluation result indicates that the tumor cells are injured (YES), the processing unit 201 proceeds to step S23, determines that the immunotherapeutic agent is useful, and the result is To be presented by outputting from the output unit 212.
  • step S22 when the evaluation result indicates that the tumor cells are not injured (NO), the processing unit 201 proceeds to step S24, determines that the immunotherapeutic agent is not useful, and outputs the result. It is presented by outputting from the output unit 212.
  • the processing unit 201 in step S22, the evaluation result is If it is indicated that the tumor cells are injured (YES), in step S23, the immunotherapeutic agent causes an adverse event to the patient in which treatment must be interrupted and administration cannot be restarted. Decide not to.
  • the processing unit 201 in step S22, the evaluation result is In the case of (NO) indicating that the tumor cells are not injured, in step S24, the immunotherapeutic agent causes an adverse event in the patient, which requires treatment interruption and cannot resume administration. Decide to wake up.
  • the processing unit 201 When the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the processing unit 201, in step S22, if the evaluation result indicates that the tumor cells are injured (YES), in step S23. , Determine that the immunotherapeutic agent is effective for the patient.
  • the processing unit 201 in step S12, the evaluation result is If the tumor cells are not injured (NO), it is determined in step S24 that the immunotherapeutic agent is ineffective for the patient.
  • the processing unit 201 may record the result obtained in step S23 or step S24 in the recording medium 213.
  • the processing unit 201 is input by the inspector from the input unit 211.
  • a value that reflects the tumor cytotoxic activity obtained by the method described in (1) is obtained (step S201).
  • the information for calculating the tumor cytotoxic activity is acquired from the measurement unit 30.
  • the information is information that reflects the number of living cells or dead cells of the tumor cells that have come into contact with PBMC, and is, for example, the absorbance of a living cell staining reagent or a dead cell measurement reagent.
  • the information is the absorbance when interferon- ⁇ is measured.
  • the processing unit 201 compares the value that reflects the tumor cell cytotoxic activity acquired in step S201 with the reference value stored in the main storage unit 202 or the auxiliary storage unit 204 (step S202).
  • the processing unit 201 determines from the comparison result of step S202 whether or not the value that reflects the tumor cell cytotoxic activity obtained in step S201 is higher than the reference value (step S203).
  • step S203 determines that the value that reflects the tumor cytotoxic activity is higher than the reference value (YES)
  • the processing unit 201 proceeds to step S204, determines that the immunotherapeutic agent is useful, and as a result, To be presented by outputting from the output unit 212.
  • step S204 determines that the immunotherapeutic agent is useful, and as a result, To be presented by outputting from the output unit 212.
  • step S205 determines that the immunotherapeutic agent is not useful, and To be presented by outputting from the output unit 212.
  • the processing unit 201 determines the tumor cytotoxic activity in step S203.
  • the processing unit 201 determines the tumor cytotoxic activity in step S203.
  • the immunotherapeutic agent is administered to the patient for an adverse event requiring treatment interruption and resumption of administration. Show what you don't.
  • the processing unit 201 determines the tumor cytotoxic activity in step S203.
  • the immunotherapeutic agent is administered to the patient for an adverse event that requires treatment interruption and cannot be resumed. Offer to wake up.
  • the processing unit 201 determines in Step S203 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES), Step S204. In, it is shown to the patient that the immunotherapeutic agent is effective.
  • the processing unit 201 in step S203, the evaluation result is In the case of indicating that the tumor cells are not injured (NO), if it is determined that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), in step S205, the Present that the immunotherapeutic is ineffective.
  • the processing unit 201 may record the result obtained in step S204 or step S205 in the recording medium 113.
  • a computer program for assisting the determination of the usefulness of an immunotherapeutic agent for treating a malignant tumor and a storage medium storing the computer program, according to an embodiment disclosed herein.
  • An immunotherapeutic agent for treating a malignant tumor the invention relates to a computer program for assisting in determining the usefulness in a patient having the malignant tumor. Specifically, the above 4.
  • an embodiment disclosed in the present specification relates to a storage medium storing the computer program. That is, the computer program is stored in a storage medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk.
  • the storage format of the program in the storage medium is not limited as long as the presentation device can read the program.
  • the storage in the storage medium is preferably non-volatile.
  • Immunotherapeutic Agents relate to immunotherapeutic agents for treating malignant tumors.
  • the present invention relates to an immunotherapeutic agent that is administered to a patient having a malignant tumor for which the immunotherapeutic agent is shown to be useful by the presenting method described in (including the part incorporating the matters described in 1 above).
  • the immunotherapeutic agent is used for treating a patient having a malignant tumor in which the immunotherapeutic agent is suggested to be useful in the above-mentioned presentation method.
  • the immunotherapy agent in the present disclosure is not limited as long as it can be used for immunotherapy of malignant tumor.
  • Immunotherapeutic agents may include known agents, new agents, agents whose efficacy has been rediscovered by drug repositioning, and the like.
  • the immunotherapeutic agent may be a drug approved as a drug by a predetermined institution or an unapproved drug.
  • the immunotherapeutic agent according to the present disclosure is an anti-PD1 antibody, anti-PD-L1 antibody, anti-CTLA-4 antibody, anti-Tim3 antibody, anti-LAG3 antibody, or the like, which is administered as an immune checkpoint inhibitor; Chimeric antigen receptor-T cells and the like administered for; interleukin-2 and the like administered for cytokine therapy; tumor antigen-derived proteins or peptides (eg WT-1, administered for tumor vaccine therapy) NY-ESO-1) and the like; a drug having an active ingredient such as a bispecific T cell engager (also referred to as BiTE (registered trademark)) or a bispecific antibody (bispecific antibody).
  • the immunotherapeutic agent is preferably an immune checkpoint inhibitor.
  • the active ingredient can be used to produce an immunotherapeutic agent.
  • immunotherapeutic agents containing anti-PD1 antibody include nivolumab, pembrolizumab and the like.
  • immunotherapeutic agents containing anti-PD-L1 antibody include atezolizumab, durvalumab, avelumab and the like.
  • immunotherapeutic agents containing anti-CTLA-4 antibody include ipilimumab and the like.
  • the bispecific T cell engager may include an engager targeting CD19 and CD3 (eg, blinatumomab), and an engager targeting EphA2 and CD3.
  • Examples of bispecific antibodies include antibodies targeting EpCAM and CD3.
  • the antibody may include at least the antigen binding region.
  • the antigen-binding region is not limited as long as it binds to the target antigen.
  • the antigen-binding region is, for example, at least one of the group consisting of complementary chain determining region 1 (CDR1), complementary chain determining region 2 (CDR2) and complementary chain determining region 3 (CDR3) of immunoglobulin that binds to the antigen. Includes CDR.
  • CDR1 complementary chain determining region 1
  • CDR2 complementary chain determining region 2
  • CDR3 complementary chain determining region 3
  • the antigen-binding region preferably comprises at least two CDRs consisting of CDR1, CDR2 and CDR3, more preferably CDR1, CDR2 and CDR3.
  • the CDR may be derived from an immunoglobulin heavy chain or an immunoglobulin light chain.
  • each CDR may be adjacent to the corresponding framework region.
  • the antigen-binding region may be an immunoglobulin variable region (V region), sc-Fv, v-NAR, immunoglobulin Fab region or Fab' region, or the like.
  • the antibody may be a chimeric antibody, a humanized antibody or the like.
  • the antibodies include single domain antibodies, single chain antibodies, maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies and the like.
  • the usage and dose of the immunotherapeutic agent are known. Can be determined according to the usage and dose.
  • the immunotherapeutic agent may consist of 100% by mass of the active ingredient, but usually it contains a pharmaceutically acceptable carrier or additive in addition to the active ingredient.
  • the ratio of the active ingredient of the immunotherapeutic agent is not particularly limited, but it can usually be selected from the range of 5 to 95% by mass. The proportion is preferably 30 to 80% by mass.
  • the dosage form of the immunotherapeutic agent includes oral administration; parenteral administration such as intravenous administration, intramuscular administration, subcutaneous administration, transmucosal administration, transdermal administration, and rectal administration. Oral administration and intravenous administration are preferred, and intravenous administration is more preferred.
  • the immunotherapeutic agent can be prepared into various forms of preparations (forms) depending on the administration method.
  • dosage forms for oral administration include powders, granules, capsules, pills, tablets, elixirs, suspensions, emulsions and syrups, which can be appropriately selected from these. .. Further, these preparations can be modified such as sustained release, stabilization, easy disintegration, difficult disintegration, enteric coating, and easy absorption.
  • the dosage form for intravenous administration, intramuscular administration, or subcutaneous administration includes injections and infusions (including dry products prepared at the time of use), which can be appropriately selected.
  • Dosage forms for transmucosal administration, transdermal administration, or rectal administration include chewing agents, sublingual agents, puckers, troches, ointments, patches, liquids, etc. You can choose here. Further, these preparations can be modified such as sustained release, stabilization, easy disintegration, difficult disintegration, and easy absorption.
  • the immunotherapeutic agent can be mixed with pharmaceutically acceptable carriers and additives depending on its dosage form (oral administration or various parenteral administration dosage forms).
  • pharmaceutically acceptable carriers and additives include solvents, excipients, coating agents, bases, binders, lubricants, disintegrating agents, solubilizing agents, suspending agents, thickening agents, emulsifying agents, Stabilizers, buffers, isotonic agents, soothing agents, preservatives, corrigents, fragrances, and colorants are mentioned.
  • Specific examples of pharmaceutically acceptable carriers and additives are listed below, but the invention is not limited thereto.
  • purified water As the solvent, purified water, sterilized purified water, water for injection, physiological saline, peanut oil, ethanol, glycerin and the like can be mentioned.
  • Excipients include starches (eg potato starch, wheat starch, corn starch), lactose, glucose, sucrose, crystalline cellulose, calcium sulfate, calcium carbonate, sodium hydrogen carbonate, sodium chloride, talc, titanium oxide, trehalose, xylitol. Etc. can be mentioned.
  • starch and its derivatives for example, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, carboxymethyl cellulose), gelatin, sodium alginate, tragacanth, natural polymer compounds such as gum arabic, polyvinylpyrrolidone, polyvinyl alcohol, etc. Examples thereof include synthetic polymer compounds, dextrin, hydroxypropyl starch and the like.
  • Lubricants include light anhydrous silicic acid, stearic acid and salts thereof (eg magnesium stearate), talc, waxes, wheat den bun, macrogol, hydrogenated vegetable oil, sucrose fatty acid ester, polyethylene glycol, silicone oil and the like. be able to.
  • disintegrant examples include starch and its derivatives, agar, gelatin powder, sodium hydrogen carbonate, calcium carbonate, cellulose and its derivatives, hydroxypropyl starch, carboxymethyl cellulose and its salts and cross-linked products thereof, low-substituted hydroxypropyl cellulose and the like. be able to.
  • solubilizing agent examples include cyclodextrin, ethanol, propylene glycol, polyethylene glycol and the like.
  • suspending agent examples include sodium carboxymethyl cellulose, polypinylpyrrolidone, gum arabic, tragacanth, sodium alginate, aluminum monostearate, citric acid and various surfactants.
  • thickener examples include sodium carboxymethylcellulose, polypinylpyrrolidone, methylcellulose, hydroxypropylmethylcellulose, polyvinyl alcohol, tragacanth, gum arabic, sodium alginate and the like.
  • emulsifier examples include gum arabic, cholesterol, tragacanth, methyl cellulose, lecithin, various surfactants (for example, polyoxyl 40 stearate, sorbitan sesquioleate, polysorbate 80, sodium lauryl sulfate).
  • the stabilizer examples include tocopherol, chelating agents (eg EDTA, thioglycolic acid), inert gases (eg nitrogen, carbon dioxide), reducing substances (eg sodium bisulfite, sodium thiosulfate, ascorbic acid, rongalite) and the like. be able to.
  • chelating agents eg EDTA, thioglycolic acid
  • inert gases eg nitrogen, carbon dioxide
  • reducing substances eg sodium bisulfite, sodium thiosulfate, ascorbic acid, rongalite
  • buffer examples include sodium hydrogen phosphate, sodium acetate, sodium citrate, boric acid and the like.
  • tonicity agent sodium chloride, glucose and the like can be mentioned.
  • soothing agents include local anesthetics (procaine hydrochloride, lidocaine), penzyl alcohol, glucose, sorbitol, amino acids and the like.
  • sucrose, saccharin, licorice extract, sorbitol, xylitol, glycerin and the like can be mentioned.
  • aromatic agents include spruce tincture and rose oil.
  • colorants include water-soluble food dyes and lake dyes.
  • preservatives examples include benzoic acid and its salts, paraoxybenzoic acid esters, chlorobutanol, inverted soap, benzyl alcohol, phenol, tiromesal, dehydroacetic acid, boric acid, and the like.
  • sucrose hydroxypropylcellulose (HPC), shellac, gelatin, glycerin, sorbitol, hydroxypropylmethylcellulose (HPMC), ethylcellulose, polyvinylpyrrolidone (PVP), hydroxypropylmethylcellulose phthalate (HPMCP), cellulose acetate phthalate (CAP). ), a methylmethacrylate-methacrylic acid copolymer, and the polymers described above.
  • HPMC hydroxypropylmethylcellulose
  • HPMC hydroxypropylmethylcellulose
  • PVP polyvinylpyrrolidone
  • HPMCP hydroxypropylmethylcellulose phthalate
  • CAP cellulose acetate phthalate
  • a methylmethacrylate-methacrylic acid copolymer a methylmethacrylate-methacrylic acid copolymer, and the polymers described above.
  • petrolatum liquid paraffin, carnauba wax, beef tallow, hydrogenated oil, paraffin, beeswax, vegetable oil, macrogol, macrogol fatty acid ester, stearic acid, sodium carboxymethyl cellulose, bentonite, cacao butter, witepsol, gelatin, stearyl.
  • DDS drug delivery system
  • the DDS preparations include sustained-release preparations, topical preparations (troches, buccal tablets, sublingual tablets, etc.), controlled-release preparations, enteric-coated preparations and gastric-soluble preparations, administration routes, bioavailability. In consideration of side effects, etc., it is a formulation in an optimal formulation form.
  • the dose depends on the type and location of the cancer to be treated, the stage (progression) of the cancer, and the patient. It may vary depending on the disease state, age, sex, body weight, type of anticancer drug used in combination, etc., and can be appropriately set according to these factors.
  • the immunotherapeutic agent is orally administered to humans, the dose can be appropriately set within the range of 0.03 to 300 mg/kg/day in terms of the amount of the active ingredient.
  • the immunotherapeutic agent is an intravenous administration agent
  • the effective blood concentration of is 0.1 to 3000 ⁇ g/mL, more preferably 1 to 1000 ⁇ g/mL. It can be administered at ⁇ 50 mg/kg.
  • the immunotherapeutic agent is preferably, for the patient having a malignant tumor for which the immunotherapeutic agent is suggested to be useful by the presenting method (hereinafter, also referred to as “administration target patient”), Administered for first line treatment.
  • the first-line treatment is chemotherapy that is first administered to a patient having a malignant tumor.
  • the method of administration is intravenous administration at intervals of 2 to 4 weeks.
  • the immunotherapeutic agent is preferably administered to the target patient for preoperative chemotherapy before surgical operation.
  • the term “before surgery” refers to preferably 90 days to 1 day before, and more preferably 60 days to 7 days before, the day of surgery.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • the immunotherapeutic agent is preferably administered to the target patient for postoperative adjuvant chemotherapy after surgical operation.
  • the term “after surgery” means preferably 1 to 180 days, more preferably 7 to 90 days from the date of operation.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • the immunotherapeutic agent is preferably administered to the target patient before the radiotherapy is performed.
  • the administration method is preferably 42 days to 1 day before, more preferably 30 days to 7 days before the date of radiotherapy.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • the immunotherapeutic agent is preferably administered to the subject patient who has undergone radiotherapy.
  • the administration method is preferably 1 to 180 days, more preferably 3 to 90 days from the date of radiotherapy.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • the radiation treatment can be performed by irradiating the affected area with X-rays or electron rays, preferably using a linear accelerator.
  • the conditions of X-ray irradiation vary depending on the degree of progression and size of the tumor, but usually one dose of 1.5 to 3 Gy, preferably about 2 Gy, is applied 2 to 5 times a week, preferably 4 to a week.
  • the method may be performed about 5 times over 1 to 5 weeks.
  • the total dose may be 20 to 70 Gy, preferably about 40 to 70 Gy, and more preferably about 50 to 60 Gy.
  • the electron beam irradiation conditions also depend on the degree of tumor progression and size, but usually one dose of 2 to 5 Gy, preferably about 4 Gy, is irradiated 1 to 5 times a week, preferably 2 to 3 times a week.
  • the method may be carried out for about 1 to 5 weeks.
  • the total dose may be about 30 to 70 Gy, preferably about 40 to 60 Gy.
  • the immunotherapeutic agent is preferably administered to the target patient before the chemoradiotherapy is performed.
  • the administration method is preferably 42 days to 1 day before, more preferably 30 days to 3 days before the date of radiotherapy.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • the immunotherapeutic agent is preferably administered to the target patient who has undergone chemoradiotherapy.
  • the administration method is preferably 1 day to 12 months, more preferably 3 days to 240 days from the date of radiotherapy.
  • the administration method is intravenous administration at intervals of 2 to 4 weeks.
  • Chemoradiotherapy is a combination therapy of radiation therapy and anticancer drug. Radiation therapy is 2 Gy for one dose, 5 times a week for 6 weeks, total dose is 60 Gy, and anticancer drug treatment is two types of anticancer drug treatment including platinum anticancer drug (eg, cisplatin or carboplatin) For at least 2 cycles.
  • platinum anticancer drug eg, cisplatin or carboplatin
  • Ipilimumab malignant melanoma, preferably unresectable malignant melanoma
  • renal cell carcinoma unresectable or metastatic renal cell carcinoma
  • Nivolumab malignant melanoma
  • non-small cell lung cancer preferably unresectable advanced or recurrent non-small cell lung cancer
  • renal cell carcinoma preferably unresectable or metastatic renal cell carcinoma
  • Hodgkin lymphoma preferably relapsed or refractory Classic Hodgkin lymphoma
  • head and neck cancer preferably head and neck cancer with recurrent or distant metastases
  • gastric cancer preferably unresectable advanced or recurrent gastric cancer exacerbated after cancer chemotherapy
  • malignant pleural mesothelioma Unresectable advanced or recurrent
  • two or more different immunotherapeutic agents can be administered in combination.
  • the immune checkpoint inhibitor can be administered in combination with an immunotherapeutic agent other than one or more other immune checkpoint inhibitors.
  • the immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor can be administered in combination.
  • an immune checkpoint inhibitor containing an anti-PD1 antibody and another antibody eg, an anti-PD-L1 antibody, Anti-CTLA-4 antibody, anti-Tim3 antibody, or anti-LAG3 antibody-containing immune checkpoint inhibitor.
  • an immune checkpoint inhibitor containing an anti-PD1 antibody can be combined with an anti-CTLA-4 antibody.
  • One or more is preferably two or more, more preferably three or more.
  • “combination” or “combination” means (I) Includes both an active ingredient of an immune checkpoint inhibitor from the beginning and an active ingredient of one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors). If it is in a state (combination), (Ii) A combination product in which the immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) exist in separate packaging forms, respectively. Or (iii) an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors), respectively. It is used in the meaning of including a case where they are present in the market in separate packaging forms and in separate distribution channels and used in combination when used.
  • the expression "combining an immune checkpoint inhibitor with one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors)" means “immunity checkpoint inhibitor". Including an active ingredient of an inhibitor and an active ingredient of one or more immunotherapeutic agents (or one or more other immune checkpoint inhibitors) other than the immune checkpoint inhibitor", “substantially immune checkpoint inhibition” Agent and one or more immunotherapeutic agents other than immune checkpoint inhibitors (or other one or more immune checkpoint inhibitors)", “Immune checkpoint inhibitors and 1 other than immune checkpoint inhibitors" It consists of one or more immunotherapeutic agents (or one or more other immune checkpoint inhibitors)".
  • the timing of administration of an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) is different from that of the immune checkpoint inhibitor.
  • Immune check consisting of separate packaging forms, each having a form in which it is separately administered with one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors)
  • a form in which a point inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or other one or more immune checkpoint inhibitors) are administered to the patient at the same time or at staggered times be able to.
  • an immune checkpoint inhibitor having an oral dosage form and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral dosage form (or one or more other immune checkpoint inhibitors) Agent
  • an immune checkpoint inhibitor having a parenteral dosage form and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having a parenteral dosage form (or one or more other immunotherapeutic agents) (Immune checkpoint inhibitor) and a form of parenteral administration at the same time.
  • the above-mentioned immune checkpoint inhibitor may be parenterally administered at different times.
  • one is an oral administration form and the other is a parenteral administration form a form in which both are administered simultaneously and in parallel can be mentioned.
  • one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral administration form and the immune checkpoint inhibitor having a parenteral administration form are administered at different times; an immune checkpoint inhibitor having a parenteral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral dosage form (Or one or more other immune checkpoint inhibitors) may be administered at staggered times.
  • one or more immunotherapeutic agents other than the immune checkpoint inhibitor may be administered prior to the immune checkpoint inhibitor, or in any order.
  • one or more immunotherapeutic agents other than the immune checkpoint inhibitor may be administered after the administration of the immune checkpoint inhibitor, or in any order.
  • the dose of the immune checkpoint inhibitor is 0.003 to 3000 mg/kg/day. It can be set appropriately from the range.
  • the dose of other immunotherapeutic agents to be combined is also 0.1 to 5000 mg/kg/day, 0.1 to 5000 mg/kg/dose, 0.1 to 5000 mg/m2/d or 100,000 to 5 million JRU/dose. It can be set appropriately.
  • the frequency of administration may be 1 to 8 days, and a drug holiday of 5 days to 9 weeks may be provided.
  • the therapeutic agent that combines an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) is an intravenous administration agent
  • the dose in which the effective blood concentration of the active ingredient of the checkpoint inhibitor is 0.1 to 3000 ⁇ g/mL, more preferably 1 to 1000 ⁇ g/mL, and the daily dose is 0.5 to 50 mg/kg.
  • the dose of one or more immunotherapeutic agents (or other one or more immune checkpoint inhibitors) other than the immune checkpoint inhibitor is also 10 to 100%, preferably 10 to 95% of the normally administered dose. It can be appropriately set within the range.
  • the therapeutic agent is preferably a combination of one or more immunotherapeutic agents and one or more anti-cancer agents other than the immunotherapeutic agents.
  • One or more is preferably two or more, more preferably three or more.
  • “combining” or “combining” means (I) In the case where the active ingredient of the immunotherapeutic agent and the active ingredient of one or more anti-cancer agents other than the immunotherapeutic agent are contained from the beginning (combination drug), (Ii) When the immunotherapeutic agent and one or more anti-cancer agents other than the immunotherapeutic agent exist in separate packaging forms and are sold as a combination (kit), or (iii) the immunotherapeutic agent One or more anti-cancer agents other than immunotherapeutic agents are used in the meaning of including the case where they are present in the market in separate packaging forms and in separate distribution channels and used in combination at the time of use.
  • the expression "combining an immunotherapeutic agent with one or more anticancer agents other than the immunotherapeutic agent” means “the active ingredient of the immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent”.
  • the timing of administration of the immunotherapeutic agent and the one or more anti-cancer agents other than the immuno-therapeutic agent is such that the immuno-therapeutic agent and the one or more anti-cancer agents other than the immunotherapeutic agent are separately administered.
  • examples of those having different forms include those in which the immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent are administered to the patient at the same time or at different times from separate packaging forms. it can.
  • an immunotherapeutic agent having an oral administration form and one or more anti-cancer agents other than the immunotherapeutic agent having an oral administration form are orally administered simultaneously; immunity having a parenteral administration form
  • An example is a form in which a therapeutic agent and one or more anticancer agents other than the immunotherapeutic agent having a parenteral form are administered parenterally at the same time.
  • an immunotherapeutic agent having an oral dosage form and one or more anticancer agents other than the immunotherapeutic agent having an oral dosage form are orally administered at staggered times;
  • the immunotherapy agent having an oral dosage form and one or more anti-cancer agents other than the immunotherapy agent having a parenteral dosage form may be parenterally administered at different times.
  • a form in which both are administered simultaneously and in parallel can be mentioned.
  • a mode to administer both of them with a staggered time a mode in which an immunotherapy agent having an oral dosage form and one or more anti-cancer agents other than the immunotherapy agent to be parenterally administered are staggered respectively; parenteral An immunotherapy agent having a dosage form and a form in which one or more anticancer agents other than the immunotherapy agent to be orally administered are administered at staggered times, respectively.
  • one or more anti-cancer agents other than the immunotherapeutic agent may be administered prior to the immunotherapeutic agent, and one or more anti-cancer agents other than the immunotherapeutic agent may be administered after the immunotherapy agent is administered.
  • the drug may be administered in any order.
  • the dose of the immunotherapeutic agent is 0.003 to 3000 mg. It can be appropriately set within the range of /kg/day.
  • the dose of one or more anti-cancer agents to be combined is also 0.1 to 5000 mg/kg/day, 0.1 to 5000 mg/kg/dose, 0.1 to 5000 mg/m2/dose or 100,000 to 5 million JRU. It can be appropriately set from / times.
  • the frequency of administration may be 1 to 8 days, and a drug holiday of 5 days to 9 weeks may be provided.
  • the effective blood concentration of the active ingredient of the immunotherapeutic agent is 0.1 to 3000 ⁇ g. /ML, more preferably 1 to 1000 ⁇ g/mL, and the daily dose may be 0.5 to 50 mg/kg.
  • the dose of one or more anti-cancer agents can also be appropriately set within the range of 10 to 100%, preferably 10 to 95% of the normally administered dose.
  • the one or more anticancer agents other than the immunotherapeutic agents are not limited as long as they are agents intended to treat tumors.
  • other anticancer agents include alkylating agents, antimetabolites, antitumor antibiotics, microvascular inhibitors, hormones or hormone analogs, platinum preparations, topoisomerase inhibitors, cytokines, antibody drugs, molecular targets. It can be appropriately selected from a drug, a steroid drug, a non-specific immunostimulant, and other anti-cancer agents according to the target cancer.
  • alkylating agents include cyclophosphamide, ifosfamide, busulfan, melphalan, bendamustine hydrochloride, nimustine hydrochloride, ranimustine, carmustine, streptozocin, dagalbazine, procarbazine hydrochloride, temozolomide; as antimetabolites, Methotrexate, pemetrexed sodium, pralatrexate, fluorouracil, doxyfluridine, capecitabine, tegafur, cytarabine, cytarabine ocfosfate hydrate, enocitabine, gemcitabine hydrochloride, mercaptopurine hydrate, fludarabine phosphate, nelarabine, pentostatin, pentostatin.
  • Hormones or hormone analogs include anastrozole, exemestane, letrozole, tamoxifen citrate, toremifene citrate, fulvestrant, flutamide, bicalutamide, enzalutamide, chlormadinone acetate, Abiraterone acetate, medroxyprogesterone acetate, estramustine phosphate sodium hydrate, goserelin acetate, leuprorelin acetate, degarelix acetate, etc.; platinum preparations include cisplatin, miriplatin hydrate, carboplatin, Nedaplatin, oxaliplatin, etc.; topoisomerase inhibitors include topoisomerase I inhibitors such as irinotecan hydrochloride hydrate and nogitecan hydrochloride, and doxorubicin hydrochloride, daunorubicin hydrochloride, pirarubicin, epi
  • Topoisomerase II inhibitors such as salts, amrubicin hydrochloride, mitoxantrone hydrochloride, etoposide, and sobuzoxane; as cytokines, interferon gamma-1a , Teseleukin, sermoleukin, etc.; antibody drugs include trastuzumab, rituximab, gemtuzumab ozogamicin, bevacizumab, cetuximab, etc.; , Dalatumumab, alemtuzumab, erotuzumab, transtuzumab emtansine, gemtuzumab odigamicin, inotuzumab ozogamicin, ibritumomab, gefitinib, erlotinib hydrochloride, afatinib maleate, otimertinib mesilibate, imate , Bortezomib, bosut
  • an immunotherapeutic agent for treating a malignant tumor and one or more anti-cancer agents other than the immunotherapeutic agent for example, an immune checkpoint inhibitor (including, for example, an anti-PD1 antibody) and a platinum preparation , Antimetabolites (eg, pemetrexed), and the like.
  • an immune checkpoint inhibitor eg, including anti-PD1 antibody
  • a platinum agent eg, carboplatin
  • a microvascular inhibitor eg, paclitaxel or nab-paclitaxel
  • an immune checkpoint inhibitor eg, including anti-PD-L1 antibody
  • a platinum agent eg, carboplatin
  • a microvascular inhibitor eg, paclitaxel or nab-paclitaxel
  • a targeted therapeutic agent for example, an angiogenesis inhibitor, bevacizumab, etc.
  • Test Reagent and Kit for Evaluating Whether Tumor Cells are Damaged are disclosed. It relates to a test reagent 81 for evaluating whether or not.
  • the test reagent 81 is the same as in 2. above. And the presentation method described in 3. above. It can also be used to carry out the auxiliary methods described in.
  • FIG. 8A shows the structure of the test reagent 81.
  • the structure of the kit 8 is shown in FIG. 8B.
  • the test reagent 81 may include a buffer solution or the like for dissolving the engager.
  • the buffer solution may contain a reducing agent ( ⁇ -mercaptoethanol, dithiothreitol, etc.) for stabilizing the engager, a surfactant, albumin and the like. It may also contain a preservative such as sodium azide.
  • the engager may be in a dry state or may be dissolved in the buffer solution.
  • kits 8 including a test reagent 81 for evaluating whether tumor cells are injured.
  • the kit 8 can also be used to carry out the presentation method described in 2 above.
  • the kit 8 of the present embodiment includes at least a test reagent 81 and a protocol describing a measurement method performed using the test reagent, or an attached document 82 describing information such as a URL and a Q code for accessing the protocol. Including etc.
  • a test reagent 91a and a kit 9 for evaluating whether or not a tumor cell is damaged, the anti-interferon- ⁇ antibody is included.
  • the inspection reagent 91a is the same as in 2. above. And the presentation method described in 3. above. It can also be used to carry out the auxiliary methods described in.
  • the structure of the kit 9 is shown in FIG. 8C. Kit 9 is an example of an ELISA kit, but is not limited to this.
  • the kit 9 includes a test reagent 91a containing an anti-interferon- ⁇ antibody, a microplate 92 (an antibody having an epitope different from that of the anti-interferon antibody contained in the test reagent 91a may be fixed), and an anti-interferon- ⁇ .
  • Detection including an enzyme-labeled antibody for detecting an antibody, or an enzyme-labeled detector (enzyme-labeled avidin, or enzyme-labeled streptavidin when the anti-interferon antibody contained in the test reagent 91a is labeled with biotin) It includes a reagent 91b and a protocol that describes a measurement method using the kit 9, or an attachment 93 that describes information such as a URL and a Q code for accessing the protocol.
  • the kit 9 may include a substrate that reacts with the enzyme.
  • the anti-interferon- ⁇ antibody contained in the test reagent 91a is 1. Those described in can be used.
  • the test reagent 91a may contain at least one anti-interferon- ⁇ antibody.
  • the anti-interferon- ⁇ antibody is a polyclonal antibody, it may be a polyclonal antibody obtained by immunizing with one kind of antigen, or may be obtained by immunizing the same individual with two or more kinds of antigens in parallel. It may be a prepared polyclonal antibody.
  • the respective polyclonal antibodies obtained by inoculating two or more antigens into different animals may be mixed.
  • the anti-interferon- ⁇ antibody when the anti-interferon- ⁇ antibody is a monoclonal antibody, it may be a monoclonal antibody produced by one kind of hybridoma, but it is a monoclonal antibody produced by two or more kinds of hybridoma, and each monoclonal antibody Two or more kinds of monoclonal antibodies that recognize the same or different epitopes may be contained. In addition, one or more polyclonal antibodies and one or more monoclonal antibodies may be mixed and included.
  • the form of the anti-interferon- ⁇ antibody contained in the test reagent 91a is not particularly limited, and may be a dry state or a liquid state of antiserum or ascites fluid containing the anti-interferon- ⁇ antibody. Further, the form of the anti-interferon- ⁇ antibody may be a purified anti-interferon- ⁇ antibody, an immunoglobulin fraction containing the anti-interferon- ⁇ antibody or an IgG fraction containing the anti-interferon- ⁇ antibody in a dry state or an aqueous solution. ..
  • a stabilizer such as ⁇ -mercaptoethanol or DTT
  • a protective agent such as albumin
  • polyoxyethylene 20
  • It may contain at least one of a surfactant such as sorbitan monolaurate and polyoxyethylene (10) octyl phenyl ether, and a preservative such as sodium azide.
  • the form of the anti-interferon- ⁇ antibody is a purified anti-interferon- ⁇ antibody, an immunoglobulin fraction containing the anti-interferon- ⁇ antibody or an IgG fraction containing the anti-interferon- ⁇ antibody in a dry state or an aqueous solution, , Buffer components such as phosphate buffer, stabilizers such as ⁇ -mercaptoethanol and DTT, protective agents such as albumin, salts such as sodium chloride, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (10 ) It may contain at least one of preservatives such as sodium azide and the like, such as surfactants such as octylphenyl ether and the like.
  • the anti-interferon- ⁇ antibody when the test reagent 91a is supplied alone, the anti-interferon- ⁇ antibody may be unlabeled or labeled with the above-mentioned labeling substance. It is preferably labeled with the labeling substance described in 1 above.
  • a microplate 92 having an anti-interferon- ⁇ antibody for capturing an antigen immobilized on a solid phase surface or the like may be provided as a test reagent.
  • Example 1 Measurement of percent tumor cytotoxic activity using peripheral blood mononuclear cells 1.
  • Method (1) A cell suspension was prepared by suspending the U251 cell line in RPMI1640 medium (RPMI1640 medium containing 10% FBS) supplemented with 10% FBS at 1 ⁇ 10 5 cells/mL. The cell suspension was seeded on a 96-well plate at 100 ⁇ L/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
  • Peripheral blood heparin blood sampling
  • Lymphoprep TM Lymphoprep TM
  • the number of cells of the PBMC is 5 ⁇ 10 4
  • the final concentration of EphA2-CD3 BiTE is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added.
  • a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1).
  • the total volume of RPMI medium containing 10% FBS was 200 ⁇ L per well.
  • the mixture was incubated for 48 hours.
  • each well was washed 4 times with RPMI medium containing 10% FBS. After washing, 100 ⁇ L of RPMI medium containing 10% FBS and 20 ⁇ L of CellTiter 96 (registered trademark) A Queous One Solution Reagent (MTS reagent: Promega) were added to each well from which the medium had been removed. Then, the 96-well microplate was incubated at 37° C. for 20 minutes in a wet incubator in the presence of 5% carbon dioxide.
  • RPMI medium containing 10% FBS 100 ⁇ L of RPMI medium containing 10% FBS and 20 ⁇ L of CellTiter 96 (registered trademark) A Queous One Solution Reagent (MTS reagent: Promega) were added to each well from which the medium had been removed. Then, the 96-well microplate was incubated at 37° C. for 20 minutes in a wet incubator in the presence of 5% carbon dioxide.
  • Tumor cytotoxic activity percentage (%) [(A-B)/A] x 100
  • FIG. 9 shows the percent tumor cytotoxic activity of PBMC in each patient. Since BiTE that binds to CD3 is used, the tumor cytotoxic activity of this PBMC is considered to be the tumor cytotoxic activity of peripheral blood T cells. As can be seen from FIG. 9, the tumor cytotoxic activity of peripheral blood T cells in lung cancer patients varies depending on the patient, and while some patients have tumor cytotoxic activity of only a few %, a high value of 80% or more is obtained. There were also patients presenting. From this, it was found that the tumor cytotoxic activity of peripheral blood T cells of each patient varies among individuals.
  • T cells in lung cancer tissue The percent tumor cytotoxic activity of T cells in lung cancer tissue was determined by collecting cells from lung cancer tissue according to the following method and determining the cytotoxic activity of the cell group. It was measured. The cancer tissue was transferred to a 6 cm Dish and shredded, then placed in a tissue agitation solution (HBSS(-) + 2% FBS + 10 mM HEPES with Tumor Dissociation Kit (Miltenyi Biotec) added) and gentleMACS TM Dissociator (Miltenyi Biotec) After stirring at 37° C., the cells were incubated for 30 minutes while rotating in an incubator maintained at 37° C.
  • tissue agitation solution HBSS(-) + 2% FBS + 10 mM HEPES with Tumor Dissociation Kit (Miltenyi Biotec) added
  • gentleMACS TM Dissociator Miltenyi Biotec
  • the stirred solution containing cells was passed through a 70 ⁇ m mesh to remove the tissue residue, and the filtrate was centrifuged at 600 ⁇ g for 10 minutes to recover the precipitate.
  • BD Pharm lyse (BD Biosciences) was added to the cell-containing pellet and left standing for 2 minutes.
  • HBSS Buffer was added to the lysate containing cells after standing, and the mixture was centrifuged at 600 xg for 10 minutes, and the precipitate was collected again.
  • a 30% Percoll solution was added to the recollected precipitate, and the mixture was centrifuged at 12000 xg for 30 seconds to collect the precipitate again.
  • the recollected precipitate was washed with HBSS Buffer and centrifuged at 12000 xg for 30 seconds to collect cells in the tissue.
  • the percent tumor cytotoxic activity of the recovered cells was determined according to I. It measured by the method similar to. Since BiTE that binds to CD3 is used, the tumor cytotoxic activity of the cells recovered from the lung cancer tissue is considered to be the tumor cytotoxic activity of T cells in the lung cancer tissue.
  • T cell receptor repertoire analysis In order to confirm whether or not a T cell clone common to tumor and peripheral blood exists in the same patient, repertoire analysis of T cell receptor (TCR) was performed. CD8-positive T cells were collected from cells in peripheral blood PBMCs of lung cancer patients and lung cancer tissues using FACSAria (Becton Dickinson). RNA was extracted from the collected CD8-positive T cells, the TCR gene was amplified using the Adapter-Ligation PCR method, and then the nucleotide sequence was determined using a next-generation sequencer. The TCR repertoire analysis was outsourced to Repertoire Genesis Co., Ltd. (http://www.repertoire.co.jp/detailed_repertoire.html). The repertoire analysis was performed for each of TCR ⁇ and TCR ⁇ . For patients 1-4, we compared the repertoires of the top 30 most frequently occurring clones of PBMC and T cells in lung cancer tissues.
  • Patient 1 and patient 2 were cases in which the percent tumor cytotoxic activity of CD8-positive T cells in lung cancer tissue was high, and patient 3 was an example in which the percent tumor cytotoxic activity was moderate, and patient 4 Is an example where the tumor cell cytotoxic activity percentage was low. The results are shown in Fig. 10.
  • Example 3 Correlation between tumor cytotoxic activity of T cells in lung cancer tissue and tumor cytotoxic activity of peripheral blood T cells Next, the percentage of tumor cytotoxic activity of T cells in lung cancer tissue and the peripheral blood T cells derived from PBMC were examined. The correlation of percent tumor cytotoxic activity was determined. The results are shown in Fig. 11. The correlation coefficient R 2 and the p value were obtained using statistical software JMP. The p-value shows a test value for the presence or absence of correlation.
  • the concentration of interferon ⁇ (IFN ⁇ ) showing an antitumor effect was determined for the culture supernatant obtained by adding EphA2-CD3BiTE to PBMC and U251 cell lines collected from patients for 2 days, and The correlation of percent tumor cytotoxic activity of T cells in lung cancer tissue was determined.
  • the IFN ⁇ concentration was measured using the Bio-Plex Pro human cytokine GI27-plex panel (Bio-Rad).
  • the tumor cytotoxic activity of peripheral blood T cells was more strongly correlated with the tumor cytotoxic activity of T cells in lung cancer tissues than that of IFN ⁇ .
  • the tumor cytotoxic activity of T cells in tumor tissue can be predicted by measuring the tumor cytotoxic activity of peripheral blood T cells, which in turn leads to the tumor cytotoxic activity of PBMC in peripheral blood. It was considered that the effect of tumor immunotherapy could be predicted by measuring
  • Example 4 Correlation between the effect of nivolumab and tumor cytotoxic activity of peripheral blood T cells 1.
  • Method The effect of nivolumab was compared with the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC measured according to Example 1 in 18 patients.
  • the effect of nivolumab was evaluated by the following method. According to the method described in Example 1, the U251 cell line was seeded on a 96-well plate and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
  • Tumor cytotoxic activity percentage (%) [(A-B)/A] x 100
  • the correlation coefficient R 2 and the p value were obtained using statistical software JMP.
  • the p-value shows a test value for the presence or absence of correlation.
  • the tumor cytotoxic activity of peripheral blood T cells measured using PBMC has a high correlation with the effect of tumor immunotherapy. Further, it was shown that the tumor cytotoxic activity of peripheral blood T cells can be used as a marker for evaluating the effect of tumor immunotherapy.
  • Example 5 Correlation between Tumor Cytotoxic Activity of Peripheral Blood T Cells and Effect of Tumor Immunotherapy
  • the correlation of The percent tumor cytotoxic activity of PBMC collected from the patients was measured according to Example 1, and three patients having a percent tumor cytotoxic activity of 25% or more were extracted.
  • tumor shrinkage was observed by tumor immunotherapy using anti-PD-1 antibody, and the duration of treatment was 140 days or longer, which enabled long-term treatment. The reduction of the tumor and the ability to continue the treatment in this way indicate that tumor immunotherapy is effective.
  • the positive rate of PD-L1-expressing cells in tumor tissue is used as an index when determining whether to apply tumor immunotherapy.
  • the positive rate of PD-L1-expressing lung cancer cells in the tumor tissue was measured for the 3 patients, the positive rate was 50% or more for the 2 patients, but the positive rate for 1 patient was It was a very low value of 2%.
  • the positive rate of PD-L1 expressing cells is evaluated by performing immunostaining (immunoantibody method) of PD-L1 protein. Patients with only 2% PD-L1 expressing cells cannot be predicted by PD-L1 protein immunostaining to be effective for tumor immunotherapy.
  • Example 6 Comparison of prediction accuracy of therapeutic effect between tumor cytotoxic activity and positive rate of PD-L1 expressing cells 1.
  • METHODS Peripheral blood T cells derived from PBMCs prior to treatment with pembrolizumab or nivolumab were assayed for percent tumor cytotoxic activity in 30 patients with non-small cell lung cancer, followed by treatment efficacy, progression-free survival and the need for treatment discontinuation. , And the relationship with the occurrence of adverse events (serious adverse events) that made it impossible to resume administration. Percentage of tumor cytotoxic activity of peripheral blood T cells derived from PBMC was evaluated by the following method. Currently, in determining whether to apply tumor immunotherapy, the positive rate of PD-L1 expressing cells (Tumor Proportion Score: TPS) in tumor tissues is used as one index, and this was used as a comparison target.
  • TPS Tumor Proportion Score
  • RPMI1640 medium (10% FBS-containing RPMI1640 medium) containing 10% FBS of U251 cell line at 1 ⁇ 10 5 cells/mL
  • a cell suspension suspended in was prepared.
  • the cell suspension was seeded on a 96-well plate at 100 ⁇ L/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
  • Peripheral blood (heparin blood sampling) was collected from a lung cancer patient, and a cell group containing peripheral blood mononuclear cells (referred to as "PBMC") was collected using Lymphoprep TM (Alere Technologies AS) according to the attached protocol.
  • PBMC peripheral blood mononuclear cells
  • the PBMCs were collected immediately before the step (3) described later.
  • the number of cells of the PBMC is 5 ⁇ 10 4
  • the final concentration of EphA2-CD3 BiTE (registered trademark) is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added.
  • a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1).
  • the total volume of RPMI medium containing 10% FBS was 200 ⁇ L per well.
  • the cutoff value of the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC before treatment with pembrolizumab or nivolumab was calculated to be 18.98%, the sensitivity was 78.26%, and the specificity was 60.00%, The AUC was 0.65217 ( Figure 13A).
  • the cut-off value for the positive rate of PD-L1 expressing cells in the tumor tissue before treatment with pembrolizumab or nivolumab was calculated to be 55.00%, the sensitivity was 69.57%, the specificity was 60.00%, and the AUC was 0.60870. (FIG. 13B). From these results, it was shown that the tumor cytotoxic activity was excellent in predicting the therapeutic effects of nivolumab and pembrolizumab.
  • the patient cytotoxic activity percentage of peripheral blood T cells derived from PBMC before treatment was 18.98% or more. It showed significantly prolonged progression-free survival (Figs. 16A and 17A).
  • patients with a PD-L1-expressing cell positive cut-off value of 55.00% or higher, or 50.00% or higher did not have a significant prolongation of progression-free survival for both pembrolizumab and nivolumab ( 16B, C and 17B, C).
  • PBMC peripheral blood T cells
  • the cutoff value in this example was calculated using the ROC curve (Receiver Operator Characteristic Curve). ) was used to compare sensitivity and specificity, and the area under the ROC curve (Area Under Curve: AUC). Regarding the cutoff value, the percent value of the tumor cytotoxicity that maximizes the value of "true positive sensitivity-(1-specificity)" was taken as the optimum cutoff value.
  • the cut-off value for the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMCs before treatment with pembrolizumab or nivolumab was calculated to be 35.60%, sensitivity was 69.57%, specificity was 100.00%, and AUC was 0.77640.
  • the cut-off value of the positive rate of PD-L1 expressing cells in the tumor tissue before the treatment of pembrolizumab or nivolumab was calculated to be 65.00%, the sensitivity was 73.91%, the specificity was 71.43%, and the AUC was 0.65528.
  • FIG. 19B show that the tumor cytotoxicity is excellent in predicting the occurrence of serious adverse events resulting in treatment discontinuation of nivolumab and pembrolizumab.
  • Example 7 Prediction of usefulness of immunotherapeutic agents by the concentration of interferon- ⁇ Method
  • the percentage of tumor cytotoxic activity of peripheral blood T cells derived from PBMC was measured, and the correlation with the concentration of interferon- ⁇ (IFN ⁇ ) in the culture supernatant was determined.
  • the tumor cytotoxic activity percentage of peripheral blood T cells derived from PBMC and the concentration of interferon ⁇ in the culture supernatant were evaluated by the following methods.
  • RPMI1640 medium containing 10% FBS of U251 cell line (1 ⁇ 10 5 cells/mL) A cell suspension suspended in RPMI1640 medium containing 10% FBS was prepared. The cell suspension was seeded on a 96-well plate at 100 ⁇ L/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
  • Peripheral blood heparin blood sampling
  • Lymphoprep TM Lymphoprep TM
  • the PBMCs were collected immediately before the step (3) described later.
  • the number of cells of the PBMC is 5 ⁇ 10 4
  • the final concentration of EphA2-CD3 BiTE is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added.
  • a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1).
  • the total volume of RPMI medium containing 10% FBS was 200 ⁇ L per well.
  • the mixture was incubated for 48 hours. (4) After the incubation, the culture supernatant was collected and stored (-20°C), and then each well was washed 4 times with RPMI medium containing 10% FBS.
  • Processing Unit 10 Presentation Device 201 Processing Unit 20 Auxiliary Device

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Pathology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Toxicology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)

Abstract

The present invention addresses the problem of presenting information required in companion diagnostics for predicting the effectiveness of an immunotherapeutic agent for individual patients. To solve the problem, the present invention provides a method for presenting the effectiveness of an immunotherapeutic agent for treating a malignant tumor for a patient having a malignant tumor, the method including the following steps 1 and 2. Step 1: a step for assessing whether tumor cells are damaged after directly or indirectly contacting, in vitro, peripheral blood mononuclear cells collected from the patient. Step 2: a step for presenting that the immunotherapeutic agent is effective for the patient if the assessment indicates that the tumor cells are damaged, and/or a step for presenting that the immunotherapeutic agent is not effective for the patient if the assessment indicates that the tumor cells are not damaged.

Description

悪性腫瘍を治療するための免疫療法剤の有用性の提示方法及び悪性腫瘍を治療するための免疫療法剤Method for presenting usefulness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor

 本開示は、悪性腫瘍を治療するための免疫療法剤の有用性の提示方法、悪性腫瘍を治療するための免疫療法剤について有用性の決定を補助する方法、悪性腫瘍を治療するための免疫療法剤、悪性腫瘍を治療するための免疫療法、悪性腫瘍を治療するための免疫療法剤の有用性の提示装置、悪性腫瘍を治療するための免疫療法剤について有用性の決定を補助する装置、及び悪性腫瘍を治療するための免疫療法剤の有用性を評価するための検査試薬を含む。 The present disclosure provides a method of presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor, a method for assisting in determining the usefulness of an immunotherapeutic agent for treating a malignant tumor, an immunotherapy for treating a malignant tumor. Agent, immunotherapy for treating malignant tumor, device for presenting usefulness of immunotherapeutic agent for treating malignant tumor, device for assisting determination of usefulness of immunotherapeutic agent for treating malignant tumor, and It includes test reagents for assessing the usefulness of immunotherapeutic agents for treating malignant tumors.

 腫瘍免疫療法は、腫瘍患者自身の腫瘍細胞に対する免疫応答を利用した腫瘍の治療方法である。腫瘍免疫療法の一例では、T細胞上の蛋白質に結合する抗CTLA-4抗体、抗PD-1抗体等を使って、腫瘍患者自身の細胞傷害性T細胞を活性化し、その機能によりがん細胞を殺傷する(非特許文献1及び非特許文献2)。 Tumor immunotherapy is a tumor treatment method that uses the immune response of tumor patients to tumor cells. In an example of tumor immunotherapy, anti-CTLA-4 antibody, anti-PD-1 antibody, etc. that bind to a protein on T cells are used to activate cytotoxic T cells of the tumor patient's own, and cancer cell is activated by the function. Are killed (Non-Patent Document 1 and Non-Patent Document 2).

 また、腫瘍組織内に存在する細胞傷害性T細胞が腫瘍細胞に対して効率よく細胞傷害活性を示すことができるよう、細胞傷害性T細胞を腫瘍細胞に特異的にエンゲージさせる方法も開発されている。そのエンゲージャーとして、現在、腫瘍細胞の細胞表面マーカーとT細胞の表面マーカーとを1分子で特異的に認識する二重特異性分子(バイスペシフィック抗体)が使用されている(非特許文献2及び3)。 In addition, a method for specifically engaging cytotoxic T cells with tumor cells has been developed so that the cytotoxic T cells present in tumor tissue can efficiently exhibit cytotoxic activity against tumor cells. There is. A bispecific molecule (bispecific antibody) that specifically recognizes a cell surface marker of a tumor cell and a surface marker of a T cell with one molecule is currently used as the engager (Non-Patent Document 2 and 3).

PD1/PD-L1 Combination Therapies, 2015, Evaluate Ltd.PD1/PD-L1 Combination Therapies, 2015, Evaluate Ltd. ONCOIMMUNOLOGY, 2016, VOL. 5, NO. 2, e1062969ONCOIMMUNOLOGY, 2016, VOL.5, NO. 2, e1062969 Cancer Research, 2013, 73(15) August 1, p4663-4673Cancer Research, 2013, 73 (15) August 1, 1, p4663-4673

 一方で、例えば、非小細胞肺癌で保険適用となっている腫瘍免疫療法治療薬ペムブロリズマブ(商品名キイトルーダ)は、奏効率19%であり、多くの患者には無効であるのが現状である。このため、当該腫瘍免疫療法治療薬が有効な患者群を選別するためコンパニオン診断が行われている。現在、ペムブロリズマブにおいては、生検検体を用いたPD-L1染色がコンパニオン診断薬として用いられているが、PD-L1染色で50%以上陽性の患者群でも奏効率は45%である。このことから、PD-L1染色より優れたコンパニオン診断の開発が切望されている。 On the other hand, for example, the tumor immunotherapeutic drug pembrolizumab (trade name KEYTRUDA), which is covered by insurance for non-small cell lung cancer, has a response rate of 19% and is currently ineffective for many patients. For this reason, companion diagnosis is performed in order to select patient groups in which the therapeutic drug for tumor immunotherapy is effective. Currently, in pembrolizumab, PD-L1 staining using a biopsy sample is used as a companion diagnostic agent, but the response rate is 45% even in a patient group positive for PD-L1 staining by 50% or more. Therefore, development of companion diagnostics superior to PD-L1 staining has been earnestly desired.

 さらに、腫瘍免疫療法治療薬の投与による悪性腫瘍の治療では、間質性肺疾患、脳炎等の治療中止となる有害事象を引き起こすケースがあるが、現時点では、腫瘍免疫療法治療薬の投与による重篤な有害事象を予測することはできない。 Furthermore, the treatment of malignant tumors by the administration of tumor immunotherapeutic drugs may cause adverse events such as interstitial lung disease and encephalitis, which are treatment discontinuation. No serious adverse events can be predicted.

 上記のような奏効率、悪性腫瘍に対する免疫療法剤の効果予測、及び/又は悪性腫瘍に対する免疫療法を中止せざるを得ない有害事象の発生も含めた、悪性腫瘍を標的とする免疫療法の有用性を予測することの困難性は、多くの悪性腫瘍を標的とする免疫療法に共通の課題である。このような腫瘍免疫療法の現状から、本発明は、個々の患者について免疫療法剤の有用性を予測するためのコンパニオン診断に必要な情報を提示することを一課題とする。また、免疫療法剤が有用性であることが提示された患者に投与される、免疫療法剤を提供することを一課題とする。 Usefulness of immunotherapy targeting malignant tumors, including the above-mentioned response rate, prediction of the effect of immunotherapeutic agents on malignant tumors, and/or occurrence of adverse events inevitably stopping immunotherapy for malignant tumors The difficulty of predicting sex is a common problem in immunotherapy targeting many malignancies. In view of the current state of tumor immunotherapy, an object of the present invention is to present information necessary for companion diagnosis for predicting the usefulness of immunotherapeutic agents for individual patients. Another object is to provide an immunotherapeutic agent to be administered to a patient who is suggested to be useful.

 本発明者は、鋭意研究を重ねたところ、in vitroで、個々の患者から採取した末梢血中の単核細胞を使用して腫瘍細胞が傷害されているか否かを評価することにより、免疫療法剤の有用性を各患者について提示できることを見出した。 The inventors of the present invention have conducted extensive studies and found that immunotherapy can be performed in vitro by evaluating whether tumor cells are injured by using mononuclear cells in peripheral blood collected from individual patients. It was found that the usefulness of the agent can be presented for each patient.

 本明細書には、下記実施形態が開示される。
(I)悪性腫瘍を治療するための免疫療法剤の有用性の提示方法、有用性の決定の補助方法及び検査試薬
(I-1) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(I-2) 前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
前記工程2が、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は
前記工程2が、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、
を含む、(I-1)に記載の提示方法。
(I-3) 前記有用性が、前記免疫療法剤の効果の有無であり、
前記工程2が、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は
前記工程2が、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、
を含む、(I-1)又は(I-2)に記載の提示方法。
(I-4) 前記工程1の腫瘍細胞が傷害されているか否かを評価する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程、及び
前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高い場合に前記腫瘍細胞が傷害されていることを示す工程、及び/又は前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より低い場合に前記腫瘍細胞が傷害されていないことを示す工程、
を含む、(I-1)から(I-3)のいずれか一項に記載の提示方法。
(I-5) 腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、(I-4)に記載の提示方法。
(I-6) 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、(I-4)に記載の提示方法。
(I-7) 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、(I-1)から(I-6)のいずれか一項に記載の提示方法。
(I-8) 前記エンゲージャーが、二重特異性分子である、(I-7)に記載の提示方法。
(I-9) 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、(I-8)に記載の提示方法。
(I-10) 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、(I-1)から(I-9)に記載のいずれか一項に提示方法。
(I-11) (I-7)から(I-10)のいずれか一項に記載の提示方法において、前記免疫療法剤について、前記悪性腫瘍を有する患者における有用性を評価するための、エンゲージャーを含む、
検査試薬。
(I-12) 下記工程iから工程iiを含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助する方法:
工程i:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程ii:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(I-13) 前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
工程iiが、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は
工程iiが、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、
を含む、(I-12)に記載の補助方法。
(I-14) 前記有用性が、前記免疫療法剤の効果の有無であり、
工程iiが、前記評価において前記腫瘍細胞が傷害されていると決定された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は
工程iiが、前記評価において前記腫瘍細胞が傷害されていないと決定された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、
を含む、(I-12)又は(I-13)に記載の補助方法。
(I-15) 前記工程iの腫瘍細胞が傷害されているか否かを評価する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程、及び
前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高い場合に腫瘍細胞が傷害されていることを示す工程、及び/又は前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より低い場合に前記腫瘍細胞が傷害されていないことを示す工程、
を含む、(I-12)から(I-14)のいずれか一項に記載の補助方法。
(I-16) 腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、(I-15)に記載の補助方法。
(I-17) 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、(I-15)に記載の補助方法。
(I-18) 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、(I-12)から(I-17)のいずれか一項に記載の補助方法。
(I-19) 前記エンゲージャーが、二重特異性分子である、(I-18)に記載の補助方法。
(I-20) 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、(I-19)に記載の補助方法。
(I-21) 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、(I-12)から(I-20)に記載のいずれか一項に補助方法。
(I-22) (I-18)から(I-21)のいずれか一項に記載の補助方法において、前記免疫療法剤について、前記悪性腫瘍を有する患者における有用性を評価するための、エンゲージャーを含む、
検査試薬。
The following embodiments are disclosed herein.
(I) Method of presenting usefulness of immunotherapeutic agent for treating malignant tumor, method of assisting determination of usefulness and test reagent (I-1) Treating malignant tumor including the following step 1 and step 2 Method for presenting usefulness of an immunotherapeutic agent for use in a patient having the malignant tumor:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(I-2) The usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and cannot resume administration,
When the step 2 shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent for the patient needs to be interrupted and the administration cannot be restarted. If the step of presenting that no adverse event occurs, and/or the step 2 is shown in the evaluation that the tumor cells are not injured, the patient is treated with the immunotherapeutic agent. Presenting an adverse event that requires discontinuation and that makes it impossible to resume administration,
The presentation method according to (I-1), which comprises:
(I-3) The usefulness is presence or absence of the effect of the immunotherapeutic agent,
Said step 2 presents to said patient that said immunotherapeutic agent is effective when said tumor cells are shown to be injured, and/or said step 2 Presenting to the patient that the immunotherapeutic agent is ineffective if the evaluation indicates that the tumor cells are not injured,
The presentation method according to (I-1) or (I-2), which comprises:
(I-4) The step of evaluating whether or not the tumor cells in step 1 above are injured,
A step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells contacted with the tumor cells, and comparing the value reflecting the tumor cytotoxic activity with a corresponding reference value, and the reference corresponding to the value If the value is lower than the corresponding reference value, the step of indicating that the tumor cells are injured when the value is higher than the value, and/or the value reflecting the tumor cytotoxic activity is compared with the corresponding reference value. A step showing that the tumor cells are not injured,
The presentation method according to any one of (I-1) to (I-3), including:
(I-5) The presentation according to (I-4), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. Method.
(I-6) In (I-4), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measurement value of interferon-γ in the culture medium. How to present the description.
(I-7) The in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, from (I-1) to (I) -The presentation method according to any one of 6).
(I-8) The presentation method according to (I-7), wherein the engager is a bispecific molecule.
(I-9) The bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (I-8).
(I-10) The method according to any one of (I-1) to (I-9), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
(I-11) In the presentation method according to any one of (I-7) to (I-10), the method for evaluating the usefulness of the immunotherapeutic agent in a patient having the malignant tumor, Including gadgets,
Testing reagent.
(I-12) A method for assisting in determining the usefulness of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor, comprising the following steps i to ii:
Step i: a step of evaluating whether or not tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured, and step ii: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(I-13) The usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and cannot resume administration,
If the step ii shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent is harmful to the patient because the treatment needs to be interrupted and the administration cannot be restarted. If the step of presenting no event and/or step ii is shown in the assessment that the tumor cells are not injured, the immunotherapeutic agent is directed to the patient for treatment interruption. The need to present an adverse event that makes it impossible to resume administration,
The auxiliary method according to (I-12), which comprises:
(I-14) The usefulness is whether or not the immunotherapeutic agent is effective,
Step ii presenting to said patient that said immunotherapeutic agent is effective if said tumor cells are determined to be injured in said evaluation, and/or step ii comprises said evaluation Presenting to the patient that the immunotherapeutic agent is ineffective if it is determined that the tumor cells are not injured in
The auxiliary method according to (I-12) or (I-13), which comprises:
(I-15) The step of evaluating whether the tumor cells in step i are injured,
A step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cell, and comparing the value reflecting the tumor cytotoxic activity with a corresponding reference value, and the value corresponds to the criterion A step indicating that the tumor cells are injured when higher than the value, and/or a value reflecting the tumor cytotoxic activity is compared with a corresponding reference value, and when the value is lower than the corresponding reference value, A step showing that the tumor cells are not injured,
The method according to any one of (I-12) to (I-14), which comprises:
(I-16) The aid according to (I-15), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. Method.
(I-17) In (I-15), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measurement value of interferon-γ in the culture medium. Described assistance method.
(I-18) The in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, (I-12) to (I-12) -The auxiliary method according to any one of 17).
(I-19) The assisting method according to (I-18), wherein the engager is a bispecific molecule.
(I-20) The bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (I-19).
(I-21) The assisting method according to any one of (I-12) to (I-20), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
(I-22) The adjuvant method according to any one of (I-18) to (I-21), wherein the immunotherapeutic agent is used to evaluate the usefulness in a patient having the malignant tumor. Including a gadget,
Testing reagent.

(II)悪性腫瘍を治療するための免疫療法剤
(II-1) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、悪性腫瘍を治療するための免疫療法剤。
(II-2) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して1次治療のために投与される、免疫療法剤。
(II-3) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して術前化学療法のために投与される、免疫療法剤。
(II-4) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して術後補助化学療法のために投与される、免疫療法剤。
(II-5) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、放射線治療後の前記患者に投与される免疫療法剤。
(II-6) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、化学放射線治療後の前記患者に投与される免疫療法剤。
(II-7) 免疫療法剤が、免疫チェックポイント阻害剤である、(II-1)から(II-6)のいずれか一項に記載の免疫療法剤。
(II-8) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(II-7)に記載の免疫療法剤。
(II-9) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(II-1)から(II-8)のいずれか一項に記載の免疫療法剤。
(II-10) 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、(II-9)に記載の免疫療法剤。
(II-11) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、免疫チェックポイント阻害剤であって、他の1種以上の免疫チェックポイント阻害剤と組み合わせて投与される、免疫チェックポイント阻害剤。
(II-12) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、免疫チェックポイント阻害剤であって、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与される、免疫チェックポイント阻害剤。
(II-13) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(II-11)又は(II-12)に記載の免疫チェックポイント阻害剤。
(II-14) 免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(II-11)から(II-13)のいずれか一項に記載の免疫チェックポイント阻害剤。
(II-15) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して、免疫療法剤以外の1種以上の抗がん剤と組み合わせて投与される免疫療法剤。
(II-16) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(II-15)に記載の免疫療法剤。
(II-17) 免疫療法剤が、免疫チェックポイント阻害剤である、(II-15)又は(II-16)に記載の免疫療法剤。
(II) Immunotherapeutic agent for treating malignant tumor (II-1) It is shown that the immunotherapeutic agent is useful in the presenting method according to any one of (I-1) to (I-10). An immunotherapeutic agent for treating a malignant tumor, which is administered to a patient having an established malignant tumor.
(II-2) First treatment for a patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for.
(II-3) Preoperative Chemistry for Patients with Malignant Tumors Presented as Useful for Immunotherapeutic Agents in the Presenting Method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for therapy.
(II-4) Postoperative assistance for a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) An immunotherapeutic agent administered for chemotherapy.
(II-5) A patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and radiation treatment An immunotherapeutic agent to be subsequently administered to the patient.
(II-6) A patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and the actinic radiation An immunotherapeutic agent administered to the patient after treatment.
(II-7) The immunotherapeutic agent according to any one of (II-1) to (II-6), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
(II-8) The immunotherapy according to (II-7), wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. Agent.
(II-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (II-1) to (II-8) The immunotherapeutic agent according to any one of 1) above.
(II-10) The immunotherapy agent according to (II-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapy agent.
(II-11) Administered to a patient having a malignant tumor for whom an immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) , An immune checkpoint inhibitor, which is administered in combination with one or more other immune checkpoint inhibitors.
(II-12) Administered to a patient having a malignant tumor for whom the immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) , An immune checkpoint inhibitor, which is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor.
(II-13) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (II-11) or (II-12) Immune checkpoint inhibitor described in (4).
(II-14) The immune checkpoint inhibitor includes an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody (II-11) to (II-13) The immune checkpoint inhibitor according to any one of 1.
(II-15) Immunotherapy for a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10) An immunotherapeutic agent that is administered in combination with one or more anti-cancer agents other than the agent.
(II-16) The immunotherapy according to (II-15), wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. Agent.
(II-17) The immunotherapeutic agent according to (II-15) or (II-16), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.

(III)悪性腫瘍を治療するための免疫療法
(III-1) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して、免疫療法剤を投与する、悪性腫瘍の免疫療法。
(III-2) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して免疫療法剤を1次治療として投与する、悪性腫瘍を治療するための方法。
(III-3) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して免疫療法剤を術前化学療法として投与する、悪性腫瘍の免疫療法。
(III-4) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して免疫療法剤を術後補助化学療法として投与する、悪性腫瘍の免疫療法。
(III-5) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、放射線治療後の前記患者に免疫療法剤を投与する、悪性腫瘍の免疫療法。
(III-6) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、化学放射線治療後の前記患者に免疫療法剤を投与する、悪性腫瘍の免疫療法。
(III-7) 免疫療法剤が、免疫チェックポイント阻害剤である、(III-1)から(III-6)のいずれか一項に記載の悪性腫瘍の免疫療法。
(III-8) 免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、免疫療法剤を投与する、悪性腫瘍の免疫療法。
(III-9) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(III-1)から(III-8)のいずれか一項に記載の悪性腫瘍の免疫療法。
(III-10) 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、(III-9)に記載の悪性腫瘍の免疫療法。
(III-11) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して、2種以上の免疫チェックポイント阻害剤を組み合わせて投与する、悪性腫瘍の免疫療法。
(III-12) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して、免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与する、悪性腫瘍の免疫療法。
(III-13) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(III-11)又は(III-12)に記載の悪性腫瘍の免疫療法。
(III-14) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(III-11)から(III-14)のいずれか一項に記載の悪性腫瘍の免疫療法。
(III-15) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、免疫療法剤及び免疫療法剤以外の1種以上の抗がん剤を組み合わせて投与する、悪性腫瘍の免疫療法。
(III-16) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(III-15)に記載の悪性腫瘍の免疫療法。
(III-17) 免疫療法剤が、免疫チェックポイント阻害剤である、(III-15)又は(III-16)に記載の悪性腫瘍の免疫療法。
(III) Immunotherapy for treating malignant tumor (III-1) It has been proposed that the immunotherapeutic agent is useful in the presentation method according to any one of (I-1) to (I-10). Immunotherapy of a malignant tumor, wherein an immunotherapeutic agent is administered to a patient with a malignant tumor.
(III-2) An immunotherapeutic agent for a patient having a malignant tumor, which is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Is administered as a first line treatment for treating malignant tumors.
(III-3) An immunotherapeutic agent for a patient having a malignant tumor, which is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Immunotherapy of malignant tumors given as preoperative chemotherapy.
(III-4) An immunotherapeutic agent for a patient having a malignant tumor, which is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Is a postoperative adjuvant chemotherapy for malignant tumors.
(III-5) A patient with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), and radiation treatment Immunotherapy of a malignant tumor, wherein an immunotherapeutic agent is subsequently administered to the patient.
(III-6) A patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10), wherein the actinic radiation is Immunotherapy of malignant tumors, wherein the patient is treated with an immunotherapeutic agent.
(III-7) The immunotherapy for malignant tumor according to any one of (III-1) to (III-6), wherein the immunotherapy agent is an immune checkpoint inhibitor.
(III-8) Immunity to malignant tumor, wherein the immune checkpoint inhibitor is an immunotherapeutic agent containing an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody Therapy.
(III-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (III-1) to (III-8) The immunotherapy of the malignant tumor as described in any one of 1) above.
(III-10) The immunotherapy for malignant tumor according to (III-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapeutic agent.
(III-11) Two types for patients with a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presenting method according to any one of (I-1) to (I-10) Immunotherapy for malignant tumors, which is administered in combination with the above immune checkpoint inhibitors.
(III-12) Immune check for a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of (I-1) to (I-10) Immunotherapy for malignant tumors, which is administered in combination with a point inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor.
(III-13) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (III-11) or (III-12) ) Immunotherapy for malignant tumors described in (1) above.
(III-14) The (III-11) to (III-14), wherein the immune checkpoint inhibitor includes an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. The immunotherapy of the malignant tumor as described in any one of 1) above.
(III-15) Administered to a patient having a malignant tumor for whom the immunotherapeutic agent has been shown to be useful in the presentation method according to any one of (I-1) to (I-10) Immunotherapy of malignant tumors, which comprises administering an immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent in combination.
(III-16) The malignant tumor according to (III-15), wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. Immunotherapy.
(III-17) The immunotherapy for malignant tumor according to (III-15) or (III-16), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.

(IV)悪性腫瘍を治療するための免疫療法剤
(IV-1) (I-1)から(I-10)のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、悪性腫瘍を治療するための免疫療法剤。
(IV-2) 悪性腫瘍の1次治療のために投与される、(IV-1)に記載の免疫療法剤。
(IV-3) 術前化学療法のために投与される、(IV-1)に記載の免疫療法剤。
(IV-4) 術後補助化学療法のために投与される、(IV-1)に記載の免疫療法剤。
(IV-5) 放射線治療後の前記患者に投与される、(IV-1)に記載の免疫療法剤。
(IV-6) 化学放射線治療後の前記患者に投与される、(IV-1)に記載の免疫療法剤。
(IV-7) 免疫療法剤が、免疫チェックポイント阻害剤である、(IV-1)から(IV-6)のいずれか一項に記載の免疫療法剤。
(IV-8) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(IV-7)に記載の免疫療法剤。
(IV-9) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(IV-1)から(IV-8)のいずれか一項に記載免疫療法剤。
(IV-10) 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、(IV-9)に記載の免疫療法剤。
(IV-11) 前記免疫療法剤が免疫チェックポイント阻害剤であり、他の1種以上の免疫チェックポイント阻害剤と組み合わせて投与される、(IV-11)から(IV-10)のいずれか一項に記載の免疫治療剤。
(IV-12) 前記免疫療法剤が免疫チェックポイント阻害剤であり、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与される、(IV-11)から(IV-10)のいずれか一項に記載の免疫治療剤。
(IV-13) 免疫療法剤以外の1種以上の抗がん剤と組み合わせて投与される、(IV-11)から(IV-10)のいずれか一項に記載の免疫治療剤。
(IV) Immunotherapeutic agent for treating malignant tumor (IV-1) It is shown that the immunotherapeutic agent is useful in the presentation method according to any one of (I-1) to (I-10). An immunotherapeutic agent for treating a malignant tumor, which is administered to a patient having an established malignant tumor.
(IV-2) The immunotherapeutic agent according to (IV-1), which is administered for the primary treatment of malignant tumor.
(IV-3) The immunotherapy agent according to (IV-1), which is administered for preoperative chemotherapy.
(IV-4) The immunotherapy agent according to (IV-1), which is administered for postoperative adjuvant chemotherapy.
(IV-5) The immunotherapy agent according to (IV-1), which is administered to the patient after radiation treatment.
(IV-6) The immunotherapy agent according to (IV-1), which is administered to the patient after chemoradiotherapy.
(IV-7) The immunotherapy agent according to any one of (IV-1) to (IV-6), wherein the immunotherapy agent is an immune checkpoint inhibitor.
(IV-8) The immunotherapy according to (IV-7), wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. Agent.
(IV-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (IV-1) to (IV-8) An immunotherapy agent according to any one of (1) to (4).
(IV-10) The immunotherapy agent according to (IV-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapy agent.
(IV-11) Any one of (IV-11) to (IV-10), wherein the immunotherapeutic agent is an immune checkpoint inhibitor, and is administered in combination with one or more other immune checkpoint inhibitors. The immunotherapeutic agent according to one item.
(IV-12) The immunotherapeutic agent is an immune checkpoint inhibitor, and is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor, (IV-11) to (IV-10) The immunotherapeutic agent according to any one of 1.
(IV-13) The immunotherapeutic agent according to any one of (IV-11) to (IV-10), which is administered in combination with one or more anticancer agents other than the immunotherapeutic agent.

(V)悪性腫瘍を治療するための免疫療法剤の製造のための使用
(V-1) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して投与される、悪性腫瘍を治療するための免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-2) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して1次治療のために投与される、免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-3) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して術前化学療法のために投与される、免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-4) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して術後補助化学療法のために投与される、免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-5) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者であって、放射線治療後の前記患者に投与される免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-6) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者であって、化学放射線治療後の前記患者に投与される免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-7) 免疫療法剤が、免疫チェックポイント阻害剤である、(V-1)から(V-6)のいずれか一項に記載の免疫療法剤の製造のための有効成分の使用。
(V-8) 前記有効成分が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体に由来する、(V-1)から(V-7)のいずれか一項に記載の免疫療法剤の製造のための有効成分の使用。
(V-9) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(V-1)から(V-7)のいずれか一項に記載の免疫療法剤の製造のための有効成分の使用。
(V-10) 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、(V-9)に記載の免疫療法剤の製造のための有効成分の使用。
(V-11) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して投与される、免疫チェックポイント阻害剤であって、他の1種以上の免疫チェックポイント阻害剤と組み合わせて投与される免疫チェックポイント阻害剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-12) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して投与される、免疫チェックポイント阻害剤であって、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与される免疫チェックポイント阻害剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-13) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(V-11)又は(V-12)に記載の免疫チェックポイント阻害剤の製造のための有効成分の使用。
(V-14) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(V-11)から(V-13)のいずれか一項に記載の免疫チェックポイント阻害剤の製造のための有効成分の使用。
(V-15) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法によって、免疫療法剤が有用であることが提示された前記患者に対して、免疫療法剤以外の1種以上抗がん剤と組み合わせて投与される免疫療法剤の製造のための有効成分の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(V-15) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(V-14)に記載の免疫療法剤の製造のための有効成分の使用。
(V-16) 免疫療法剤が、免疫チェックポイント阻害剤である、(V-14)又は(V-15)に記載の免疫療法剤の製造のための有効成分の使用。
(V) Use for producing an immunotherapeutic agent for treating a malignant tumor (V-1) An immunotherapeutic agent for treating a malignant tumor, comprising the following Step 1 and Step 2, wherein: Of an active ingredient for the manufacture of an immunotherapeutic agent for treating a malignant tumor, which is administered to said patient for whom the immunotherapeutic agent has been shown to be useful by a method of presenting utility in a patient having use:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-2) Regarding the immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by the method of presenting the usefulness in the patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent, which is administered for the first-line treatment to the patient presented with:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-3) Regarding the immunotherapeutic agent for treating a malignant tumor, comprising the following Step 1 and Step 2, the immunotherapeutic agent being useful by the method of presenting its usefulness in a patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent, which is administered for preoperative chemotherapy to said patient presented with:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-4) Regarding the immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent, which is administered for postoperative adjuvant chemotherapy to said patient presented with:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-5) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent to be administered to said patient after radiation therapy, wherein:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-6) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent to be administered to a patient following chemoradiotherapy, wherein:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-7) Use of an active ingredient for producing the immunotherapeutic agent according to any one of (V-1) to (V-6), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
(V-8) The active ingredient is derived from anti-PD1 antibody, anti-PD-L1 antibody, anti-CTLA-4 antibody, anti-Tim3 antibody, or anti-LAG3 antibody, (V-1) to (V-7) Use of an active ingredient for the manufacture of the immunotherapeutic agent according to any one of claims.
(V-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma, (V-1) to (V-7) Use of the active ingredient for the manufacture of the immunotherapeutic agent according to any one of (1) to (4).
(V-10) The use of the active ingredient for producing the immunotherapeutic agent according to (V-9), wherein the lung cancer is complicated by interstitial pneumonia at the start of administration of the immunotherapeutic agent.
(V-11) Use of an immunotherapeutic agent for treating a malignant tumor, which comprises the following step 1 and step 2 by a method of presenting usefulness in a patient having the malignant tumor: For the manufacture of an immune checkpoint inhibitor, which is administered to the patient presented with, wherein the immune checkpoint inhibitor is administered in combination with one or more other immune checkpoint inhibitors Use of ingredients:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-12) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor. Of an immune checkpoint inhibitor, which is administered to said patient presented with, wherein the immune checkpoint inhibitor is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor. Use of active ingredients for:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-13) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (V-11) or (V-12) Use of an active ingredient for the manufacture of an immune checkpoint inhibitor as described in 1.).
(V-14) The immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody, (V-11) to (V-13) Use of an active ingredient for the manufacture of the immune checkpoint inhibitor according to any one of (1) to (4).
(V-15) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful by a method of presenting its usefulness in a patient having the malignant tumor. Use of an active ingredient for the manufacture of an immunotherapeutic agent, which is administered in combination with one or more anticancer agents other than the immunotherapeutic agent to the patient presented with:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(V-15) The immunotherapy according to (V-14), wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. Use of the active ingredient for the manufacture of a drug.
(V-16) Use of the active ingredient for producing the immunotherapeutic agent according to (V-14) or (V-15), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.

(VI)悪性腫瘍を治療するための免疫療法剤の有用性を提示するために情報を取得する方法、及び検査試薬の製造のための使用
(VI-1) 下記工程を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示するための情報の取得方法:
前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを示す情報を取得する工程。
(VI-2) 前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無である、(VI-1)に記載の取得方法。
(VI-3) 前記有用性が、前記免疫療法剤の効果の有無である、(VI-1)又は(VI-2)に記載の取得方法。
(VI-4) 前記腫瘍細胞が傷害されているか否かを反映する情報を取得する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程であって、腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、工程
を含む、(VI-1)から(VI-3)のいずれか一項に記載の取得方法。
(VI-5) 腫瘍細胞が傷害されているか否かを反映する情報を取得する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程であって、前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、工程
を含む、(VI-1)から(VI-3)のいずれか一項に記載の取得方法。
(VI-6) 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、(VI-1)から(VI-5)のいずれか一項に記載の取得方法。
(VI-7) 前記エンゲージャーが、二重特異性分子である、(VI-6)に記載の取得方法。
(VI-8) 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、(VI-7)に記載の取得方法。
(VI-9) 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、(VI-1)から(VI-8)に記載のいずれか一項に取得方法。
(VI-10) (VI-6)から(VI-9)のいずれか一項に記載の取得方法において、免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示するための情報を取得する検査試薬を製造するための、エンゲージャーの使用。
(VI) Method of obtaining information for presenting usefulness of immunotherapeutic agent for treating malignant tumor, and use for producing test reagent (VI-1) Treating malignant tumor, comprising the following steps Regarding the immunotherapeutic agent for achieving the above, a method for obtaining information for presenting usefulness in the patient having the malignant tumor:
A step of obtaining information indicating whether or not the tumor cells, which are directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro, are injured.
(VI-2) The above-mentioned usefulness is the presence or absence of an adverse event resulting from the immunotherapeutic agent that requires discontinuation of treatment and is incapable of resuming administration. Acquisition method.
(VI-3) The acquisition method according to (VI-1) or (VI-2), wherein the usefulness is the presence or absence of the effect of the immunotherapeutic agent.
(VI-4) The step of acquiring information reflecting whether or not the tumor cells are injured,
In the step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, the value reflecting the tumor cytotoxic activity is the number of living cells or dead cells of the tumor cells. The acquisition method according to any one of (VI-1) to (VI-3), which comprises the step of being a value of the tumor cytotoxic activity calculated based on the step.
(VI-5) The step of acquiring information that reflects whether tumor cells are injured,
A step of obtaining a value that reflects the tumor cytotoxic activity of peripheral blood mononuclear cells that have come into contact with the tumor cells, wherein the in vitro contact is contact in a culture medium and reflects the tumor cytotoxic activity. The acquisition method according to any one of (VI-1) to (VI-3), which comprises a step in which the value is a measurement value of interferon-γ in the culture medium.
(VI-6) The in vitro contact is indirect contact of peripheral blood mononuclear cells collected from the patient with tumor cells via an engager, (VI-1) to (VI) -The acquisition method according to any one of 5).
(VI-7) The acquisition method according to (VI-6), wherein the engager is a bispecific molecule.
(VI-8) The bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (VI-7).
(VI-9) The method according to any one of (VI-1) to (VI-8), wherein the tumor cell is a cultured cell line derived from a malignant tumor.
(VI-10) In the acquisition method according to any one of (VI-6) to (VI-9), the information for presenting the usefulness of the immunotherapeutic agent in the patient having the malignant tumor is acquired. Use of an engager to produce a test reagent that

(VII)悪性腫瘍を治療するための免疫療法剤の有用性の提示装置
(VII-1) 悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する装置であって、
前記装置は処理部を備え、
前記処理部は、
前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価した評価結果を取得し、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が有用であることを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていないことを示す場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する、
提示装置。
(VII-2) 前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
前記処理部は、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていないことを示す場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する、
(VII-1)に記載の提示装置。
(VII-3) 前記有用性が、前記免疫療法剤の効果の有無であり、
前記処理部は、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が有効であることを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が無効であることを提示する、
(VII-1)又は(VII-2)に記載の提示装置。
(VII-4) 前記末梢血単核細胞と接触した腫瘍細胞が傷害されているか否かが、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得し、前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高いか否かにより評価される、(VII-1)から(VII-3)のいずれか一項に記載の提示装置。
(VII-5) 前記値が対応する基準値より高いか否かの評価が、
前記値が対応する基準値より高い場合に腫瘍細胞が傷害されていることを示すこと、及び/又は、
前記値が対応する基準値より低い場合に腫瘍細胞が傷害されていないことを示すこと
を含む、(VII-6)に記載の提示装置。
(VII-6) 腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、(VII-5)に記載の提示装置。
(VII-7) 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、(VII-5)に記載の提示装置。
(VII-8) 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、(VII-1)から(VII-7)のいずれか一項に記載の提示装置。
(VII-9) 前記エンゲージャーが、二重特異性分子である、(VII-8)に記載の提示装置。
(VII-10) 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、(VII-9)に記載の提示装置。
(VII-11) 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、(VII-1)から(VII-10)に記載のいずれか一項に提示装置。
(VII) Device for presenting usefulness of immunotherapeutic agent for treating malignant tumor (VII-1) A device for presenting usefulness of immunotherapeutic agent for treating malignant tumor in a patient having the malignant tumor There
The apparatus includes a processing unit,
The processing unit is
Peripheral blood mononuclear cells collected from the patient, directly or indirectly, to obtain an evaluation result of evaluating whether tumor cells contacted in vitro are injured,
If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is useful to the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is not useful to the patient, if not indicated,
Presentation device.
(VII-2) The usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires discontinuation of treatment and cannot resume administration,
The processing unit is
When the evaluation result shows that the tumor cells are injured, the immunotherapeutic agent does not cause an adverse event for the patient, which requires discontinuation of treatment and resumption of administration is impossible. And/or the evaluation result indicates that the tumor cells are not injured, the immunotherapeutic agent requires treatment interruption to the patient and resumption of administration is not possible. Suggesting that a possible adverse event will occur,
The presentation device according to (VII-1).
(VII-3) The usefulness is presence or absence of the effect of the immunotherapeutic agent,
The processing unit is
If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is effective for the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is ineffective to the patient,
The presentation device according to (VII-1) or (VII-2).
(VII-4) Whether the tumor cells contacted with the peripheral blood mononuclear cells are damaged or not,
Obtain a value that reflects the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, compare the value that reflects the tumor cytotoxic activity with the corresponding reference value, the value from the corresponding reference value The presentation device according to any one of (VII-1) to (VII-3), which is evaluated by whether or not it is high.
(VII-5) Evaluation of whether or not the value is higher than the corresponding reference value is
Indicating that the tumor cells are injured when said value is higher than the corresponding reference value, and/or
The presentation device according to (VII-6), comprising indicating that the tumor cells are not injured when the value is lower than a corresponding reference value.
(VII-6) The presentation according to (VII-5), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. apparatus.
(VII-7) In (VII-5), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measurement value of interferon-γ in the culture medium. The presentation device described.
(VII-8) The in vitro contact is indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, (VII-1) to (VII) -The presentation device according to any one of 7).
(VII-9) The presentation device according to (VII-8), wherein the engager is a bispecific molecule.
(VII-10) The bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (VII-9).
(VII-11) The presentation device according to any one of (VII-1) to (VII-10), wherein the tumor cell is a cultured cell line derived from a malignant tumor.

(VIII)悪性腫瘍を治療するための免疫療法剤の有用性の決定補助装置
(VIII-1) 悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を決定することを補助する装置であって、
前記装置は処理部を備え、
前記処理部は、
前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価した評価結果を取得し、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が有用であることを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていないことを示す場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する、
決定補助装置。
(VIII-2) 前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
前記処理部は、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていないことを示す場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する、
(VIII-1)に記載の決定補助装置。
(VIII-3) 前記有用性が、前記免疫療法剤の効果の有無であり、
前記処理部は、
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が有効であることを提示する、及び/又は
前記評価結果が、前記腫瘍細胞が傷害されていることを示す場合に、前記患者に対して前記免疫療法剤が無効であることを提示する、
(VIII-1)又は(VIII-2)に記載の決定補助装置。
(VIII-4) 前記末梢血単核細胞と接触した腫瘍細胞が傷害されているか否かが、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得し、前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高いか否かにより評価される、(VII-1)から(VIII-3)のいずれか一項に記載の決定補助装置。
(VIII-5) 前記値が対応する基準値より高いか否かの評価が、
前記値が対応する基準値より高い場合に腫瘍細胞が傷害されていることを示すこと、及び/又は、
前記値が対応する基準値より低い場合に腫瘍細胞が傷害されていないことを示すこと
を含む、(VIII-4)に記載の決定補助装置。
(VIII-6) 腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、(VIII-5)に記載の決定補助装置。
(VIII-7) 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、(VIII-5)に記載の決定補助装置。
(VIII-8) 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、(VIII-1)から(VIII-7)のいずれか一項に記載の決定補助装置。
(VIII-9) 前記エンゲージャーが、二重特異性分子である、(VIII-8)に記載の決定補助装置。
(VIII-10) 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、(VIII-9)に記載の決定補助装置。
(VIII-11) 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、(VIII-1)から(VIII-10)に記載のいずれか一項に決定補助装置。
(VIII) Auxiliary Device for Determining Utility of Immunotherapeutic Agent for Treating Malignant Tumor (VIII-1) Determining Utility of Immunotherapeutic Agent for Treating Malignant Tumor in Patients Having the Malignant Tumor A device for assisting
The apparatus includes a processing unit,
The processing unit is
Peripheral blood mononuclear cells collected from the patient, directly or indirectly, to obtain an evaluation result of evaluating whether tumor cells contacted in vitro are injured,
If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is useful to the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is not useful to the patient, if not indicated,
Decision aid.
(VIII-2) The usefulness is presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and cannot resume administration,
The processing unit is
When the evaluation result shows that the tumor cells are injured, the immunotherapeutic agent does not cause an adverse event for the patient, which requires discontinuation of treatment and resumption of administration is impossible. And/or the evaluation result indicates that the tumor cells are not injured, the immunotherapeutic agent requires treatment interruption to the patient and resumption of administration is not possible. Suggesting that a possible adverse event will occur,
The determination assisting device according to (VIII-1).
(VIII-3) The usefulness is whether or not the immunotherapeutic agent has an effect,
The processing unit is
If the evaluation result indicates that the tumor cell is injured, it indicates that the immunotherapeutic agent is effective for the patient, and/or the evaluation result indicates that the tumor cell is injured. Indicating that the immunotherapeutic agent is ineffective to the patient,
The determination assisting device according to (VIII-1) or (VIII-2).
(VIII-4) Whether the tumor cells contacted with the peripheral blood mononuclear cells are damaged or not,
Obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cells, comparing the value reflecting the tumor cytotoxic activity with the corresponding reference value, the value from the corresponding reference value The determination assisting device according to any one of (VII-1) to (VIII-3), which is evaluated according to whether or not it is high.
(VIII-5) Evaluation of whether or not the value is higher than the corresponding reference value is
Indicating that the tumor cells are injured when said value is higher than the corresponding reference value, and/or
The decision aid of (VIII-4), comprising indicating that the tumor cells are not injured when the value is below the corresponding reference value.
(VIII-6) The determination according to (VIII-5), wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. Auxiliary device.
(VIII-7) In (VIII-5), the contact in vitro is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is the measured value of interferon-γ in the culture medium. The described decision aid device.
(VIII-8) The in vitro contact is indirect contact of peripheral blood mononuclear cells collected from the patient with tumor cells via an engager, (VIII-1) to (VIII) -The determination assisting device according to any one of 7).
(VIII-9) The determination aid device according to (VIII-8), wherein the engager is a bispecific molecule.
(VIII-10) The bispecific molecule is a molecule containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. , (VIII-9).
(VIII-11) The determination assisting device according to any one of (VIII-1) to (VIII-10), wherein the tumor cell is a cultured cell line derived from a malignant tumor.

(IX)悪性腫瘍を治療するための免疫療法剤の使用方法
(IX-1) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して投与される、悪性腫瘍を治療するための免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-2) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して1次治療のために投与する、免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-3) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して術前化学療法のために投与する、免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-4) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む、前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して術後補助化学療法のために投与する、免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-5) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者であって、放射線治療後の前記患者に投与する、免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-6) 悪性腫瘍を治療するための免疫療法剤を、下記工程1、及び工程2を含む前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者であって、化学放射線治療後の前記患者に投与する、免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-7) 免疫療法剤が、免疫チェックポイント阻害剤である、(IX-1)から(IX-6)のいずれか一項に記載の免疫療法剤の使用。
(IX-8) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(IX-7)に記載の免疫療法剤の使用。
(IX-9) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、又は腎細胞癌、尿路上皮癌、胃癌、悪性胸膜中皮腫である、(IX-1)から(IX-8)のいずれか一項に記載免疫療法剤の使用。
(IX-10) 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、(IX-9)に記載の免疫療法剤の使用。
(IX-11) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して投与する、免疫チェックポイント阻害剤であって、他の1種以上の免疫チェックポイント阻害剤と組み合わせて投与される、免疫チェックポイント阻害剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-12) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して投与する、免疫チェックポイント阻害剤であって、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与される、免疫チェックポイント阻害剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-13) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(IX-11)又は(IX-12)に記載の免疫チェックポイント阻害剤の使用。
(IX-14) 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、(IX-11)から(IX-13)に記載の免疫チェックポイント阻害剤の使用。
(IX-15) 下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法において免疫療法剤が有用であることが提示された前記患者に対して投与される、免疫療法剤以外の1種以上の抗がん剤免疫療法剤と組み合わせて使用される免疫療法剤の使用:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
(IX-16) 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、(IX-14)又は(IX-15)に記載の免疫療法剤の使用。
(IX-17) 免疫療法剤が、免疫チェックポイント阻害剤である、(IX-15)又は(IX-16)に記載の免疫療法剤の使用。
(IX) Method of Using Immunotherapeutic Agent for Treating Malignant Tumor (IX-1) Usefulness of Immunotherapeutic Agent for Treating Malignant Tumor in a Patient Having the Malignant Tumor Including Steps 1 and 2 below Use of an immunotherapeutic agent to treat a malignant tumor administered to said patient who has been shown to be useful in the method of presenting sex:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-2) Presenting that an immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2. Use of an immunotherapeutic agent for the first-line treatment of said patient:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-3) Presenting that an immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2. Use of immunotherapeutic agents administered to the patient for preoperative chemotherapy:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-4) An immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2. Use of immunotherapeutic agents given to the presented patient for postoperative adjuvant chemotherapy:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-5) Presenting that an immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2. Use of an immunotherapeutic agent administered to said patient following radiation therapy:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-6) Presenting that an immunotherapeutic agent for treating a malignant tumor is useful in a method of presenting usefulness in a patient having the malignant tumor, including the following Step 1 and Step 2. Use of an immunotherapeutic agent administered to said patient after chemoradiotherapy, wherein:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-7) Use of the immunotherapeutic agent according to any one of (IX-1) to (IX-6), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
(IX-8) The immunotherapy according to (IX-7), wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. Use of agents.
(IX-9) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, or renal cell carcinoma, urothelial cancer, gastric cancer, malignant pleural mesothelioma, (IX-1) to (IX-8) Use of the immunotherapeutic agent according to any one of (1) to (4).
(IX-10) Use of the immunotherapeutic agent according to (IX-9), wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapeutic agent.
(IX-11) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful in a method of presenting usefulness in a patient having the malignant tumor. Use of an immune checkpoint inhibitor administered to said presented patient, which is administered in combination with one or more other immune checkpoint inhibitors:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-12) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following step 1 and step 2, the immunotherapeutic agent is useful in a method of presenting usefulness in a patient having the malignant tumor. Use of an immune checkpoint inhibitor, which is administered to the presented patient and is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-13) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell cancer, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (IX-11) or (IX-12) Use of the immune checkpoint inhibitor described in 1.).
(IX-14) The immune checkpoint inhibitor contains an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody (IX-11) to (IX-13) Use of the immune checkpoint inhibitor described in 1.).
(IX-15) Regarding an immunotherapeutic agent for treating a malignant tumor, which comprises the following Step 1 and Step 2, the immunotherapeutic agent is useful in a method of presenting usefulness in a patient having the malignant tumor. Use of immunotherapeutic agents used in combination with one or more anti-cancer agent immunotherapeutic agents other than the immunotherapeutic agent administered to said presented patient:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
(IX-16) The malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma (IX-14) or (IX-15) Use of the immunotherapeutic agent described in (1).
(IX-17) Use of the immunotherapeutic agent according to (IX-15) or (IX-16), wherein the immunotherapeutic agent is an immune checkpoint inhibitor.

 本発明によれば、in vitroで、個々の患者から採取された末梢血中の単核細胞(PBMC)を使用し腫瘍細胞が傷害されているか否かを評価することにより、免疫療法剤の有用性を各患者について提示できる。免疫療法剤の有用性が評価された患者に対して投与される免疫療法剤を提供することができる。 INDUSTRIAL APPLICABILITY According to the present invention, usefulness of an immunotherapeutic agent is evaluated by in vitro evaluating whether tumor cells are injured by using mononuclear cells (PBMC) in peripheral blood collected from individual patients. Gender can be presented for each patient. It is possible to provide an immunotherapeutic agent to be administered to a patient whose usefulness of the immunotherapeutic agent has been evaluated.

図1は、本発明の概要を示す。FIG. 1 shows an outline of the present invention. 図2は、提示装置及び決定補助装置の概略を例示す。FIG. 2 shows an example of the outline of the presentation device and the determination assisting device. 図3は、提示装置及び決定補助装置のハードウェアの構成を示すブロック図の例である。FIG. 3 is an example of a block diagram showing a hardware configuration of a presentation device and a decision assisting device. 図4は、提示装置の処理部が行う処理を示すフローチャートの例である。FIG. 4 is an example of a flowchart showing processing performed by the processing unit of the presentation device. 図5は、提示装置の処理部が行う処理を示すフローチャートの例である。FIG. 5 is an example of a flowchart showing processing performed by the processing unit of the presentation device. 図6は、決定補助装置の処理部が行う処理を示すフローチャートの例である。FIG. 6 is an example of a flowchart showing a process performed by the processing unit of the determination assisting device. 図7は、決定補助装置の処理部が行う処理を示すフローチャートの例である。FIG. 7 is an example of a flowchart showing a process performed by the processing unit of the determination assisting device. 図8は、検査試薬及び検査キットの構成の例を示す。FIG. 8 shows an example of the configuration of a test reagent and a test kit. 図9は、肺癌患者PBMCの腫瘍細胞傷害活性を示す。FIG. 9 shows the tumor cytotoxic activity of lung cancer patient PBMC. 図10は、末梢血CD8陽性T細胞と肺癌組織内CD8陽性T細胞のレパトア解析の結果を示す。FIG. 10 shows the results of repertoire analysis of peripheral blood CD8-positive T cells and lung cancer tissue CD8-positive T cells. 図11は、腫瘍(肺癌)組織内T細胞の腫瘍細胞傷害活性とエンゲージャーを介したPBMCによるIFNγ産生量(図11A)、又は末梢血T細胞の腫瘍細胞傷害活性(図11B)との相関関係を示すグラフである。FIG. 11 shows the correlation between the tumor cytotoxic activity of T cells in tumor (lung cancer) tissue and the amount of IFNγ production by PBMC via the engager (FIG. 11A), or the tumor cytotoxic activity of peripheral blood T cells (FIG. 11B). It is a graph which shows a relationship. 図12は、腫瘍(肺癌)組織内T細胞のニボルマブの効果と末梢血T細胞の腫瘍細胞傷害活性との相関関係(図12A)、又は末梢血T細胞の腫瘍細胞傷害活性のカットオフ値を28%とした場合の各群の腫瘍(肺癌)組織内T細胞のニボルマブの効果(図12B)を示すグラフである。FIG. 12 shows the correlation between the effect of nivolumab on T cells in tumor (lung cancer) tissue and the tumor cytotoxic activity of peripheral blood T cells (FIG. 12A), or the cutoff value of the tumor cytotoxic activity of peripheral blood T cells. FIG. 12B is a graph showing the effect of nivolumab on T cells in tumor (lung cancer) tissues of each group when the percentage is 28% (FIG. 12B). (A)はPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントのROC曲線を示す。(B)PD-L1発現細胞の陽性率のROC曲線を示す。(A) shows a ROC curve of percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC. (B) shows an ROC curve of the positive rate of PD-L1 expressing cells. PBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントと腫瘍組織におけるPD-L1発現細胞の陽性率の比較結果を示す。The comparison result of the tumor cell cytotoxic activity percentage of the peripheral blood T cell derived from PBMC and the positive rate of the PD-L1 expression cell in a tumor tissue is shown. (A)ペムブロリズマブ又はニボルマブ投与患者における、腫瘍細胞傷害活性パーセントのカットオフ値18.98%以上の患者群と18.98%未満の患者群の無増悪生存期間を示す。(B)ペムブロリズマブ又はニボルマブ投与患者における、腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値55.00%以上の患者群と55.00%未満の患者群の無増悪生存期間を示す。(C)ペムブロリズマブ又はニボルマブ投与患者における、腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値50.00%以上の患者群と50.00%未満の患者群の無増悪生存期間を示す。(A) Progression-free survival time of a patient group with a cut-off value of percent tumor cytotoxicity of 18.98% or more and a patient group of less than 18.98% in patients treated with pembrolizumab or nivolumab. (B) Shows progression-free survival of patients with pembrolizumab or nivolumab who had PD-L1-expressing cells in the tumor tissue with a cutoff value of 55.00% or more and less than 55.00%. (C) Shows progression-free survival in patients with pembrolizumab or nivolumab administered, with a cut-off value of PD-L1 expressing cells in the tumor tissue of 50.00% or more and less than 50.00%. (A)ペムブロリズマブ投与患者における腫瘍細胞傷害活性パーセントのカットオフ値18.98%以上の患者群と18.98%未満の患者群の無増悪生存期間を示す。(B)ペムブロリズマブ投与患者における腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値55.00%以上の患者群と55.00%未満の患者群の無増悪生存期間を示す。(C)ペムブロリズマブ投与患者における腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値50.00%以上の患者群と50.00%未満の患者群の無増悪生存期間を示す。(A) Shows the progression-free survival of patients with pembrolizumab-treated patients with a cut-off value of percent tumor cytotoxicity of 18.98% or more and patients with a cutoff value of less than 18.98%. (B) Shows progression-free survival in a group of patients with a positive cutoff value of PD-L1 expressing cells in tumor tissues of pembrolizumab-treated patients of 55.00% or more and a patient group of less than 55.00%. (C) The progression-free survival time of the patient group with a positive cut-off value of PD-L1 expressing cells in the tumor tissue of pembrolizumab-treated patients of 50.00% or more and the patient group of less than 50.00%. (A)ニボルマブ投与患者における腫瘍細胞傷害活性パーセントのカットオフ値18.98%以上の患者群と18.98%未満の患者群の無増悪生存期間を示す。(B)ニボルマブ投与患者における腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値55.00%以上の患者群と55.00%未満の患者群の無増悪生存期間を示す。(C)ニボルマブ投与患者における腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値50.00%以上の患者群と50.00%未満の患者群の無増悪生存期間を示す。(A) Progression-free survival of a patient group with a cutoff value of 18.98% or more and a patient group of less than 18.98% for the percent tumor cytotoxic activity in patients treated with nivolumab. (B) Shows progression-free survival in a group of patients with a positive cutoff value of PD-L1 expressing cells in tumor tissues of nivolumab-treated patients of 55.00% or more and a patient group of less than 55.00%. (C) The progression-free survival time of the patient group with a positive cutoff value of PD-L1 expressing cells in the tumor tissue of nivolumab-treated patients of 50.00% or more and the patient group of less than 50.00%. (A)ペムブロリズマブ又はニボルマブの治療中止となる有害事象が出現した患者群と治療中止となる有害事象が出現しなかった患者群のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントを示す。(B)ペムブロリズマブ又はニボルマブの治療中止となる有害事象が出現した患者群と治療中止となる有害事象が出現しなかった腫瘍組織におけるPD-L1発現細胞の陽性率を示す。(A) Percent tumor cell cytotoxic activity of PBMC-derived peripheral blood T cells in a patient group in which an adverse event resulting in discontinuation of treatment with pembrolizumab or nivolumab and in a patient group in which an adverse event resulting in discontinuation of treatment did not occur. (B) shows the positive rate of PD-L1 expressing cells in a patient group in which an adverse event resulting in discontinuation of treatment with pembrolizumab or nivolumab and in a tumor tissue in which an adverse event resulting from discontinuation of treatment did not occur. (A)は有害事象の発生に関するPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントのROC曲線を示す。(B)は有害事象の発生に関するPD-L1発現細胞の陽性率のROC曲線を示す。(A) shows a ROC curve of percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC regarding the occurrence of adverse events. (B) shows the ROC curve of the positive rate of PD-L1 expressing cells regarding the occurrence of adverse events. 末梢血T細胞の腫瘍細胞傷害活性パーセントパーセントと培養上清のインターフェロン-γ濃度の相関を示す。The correlation between the percentage of tumor cytotoxic activity of peripheral blood T cells and the concentration of interferon-γ in the culture supernatant is shown.

1.腫瘍細胞の傷害の評価
 本開示においては、図1に示すように、個々の患者から採取された末梢血単核細胞(Peripheral Blood Mononuclear Cells:PBMC)と、腫瘍細胞とを、直接又はエンゲージャー等を介して間接的に接触させ、前記腫瘍細胞が傷害されているか否かを評価し、悪性腫瘍に対する免疫療法剤(以下、単に「免疫療法剤」ともいう)の有用性の評価に利用する。図1は、エンゲージャーを介してPBMCに含まれるT細胞と、予めシャーレに播種されて培養された腫瘍細胞とを接触させ、腫瘍細胞傷害活性を反映する値を取得する例を示す。
1. Evaluation of Tumor Cell Injury In the present disclosure, as shown in FIG. 1, peripheral blood mononuclear cells (PBMC) collected from an individual patient and tumor cells are directly or by an engager. It is used to evaluate the usefulness of an immunotherapeutic agent (hereinafter also simply referred to as “immunotherapeutic agent”) against a malignant tumor by contacting the cells indirectly through the cells to evaluate whether or not the tumor cells are injured. FIG. 1 shows an example in which T cells contained in PBMCs are brought into contact with tumor cells that have been seeded and cultured in a petri dish in advance via an engager, and a value that reflects tumor cytotoxic activity is obtained.

 本実施形態は、具体的には、患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する。好ましくは、患者から採取された末梢血単核細胞と、腫瘍細胞とを、直接、又は間接的にin vitroで接触させる工程(工程1)と、前記工程1で末梢血単核細胞と接触した腫瘍細胞が傷害されているか否かを評価する工程(工程2)により、前記腫瘍細胞が傷害されているか否かを評価する。本実施形態は、ヒトによって実施されても、後述する提示装置10又は補助装置20において実施されてもよい。 In the present embodiment, specifically, it is evaluated whether or not the tumor cells contacted with the peripheral blood mononuclear cells collected from the patient directly or indirectly in vitro are injured. Preferably, the step of contacting the peripheral blood mononuclear cells collected from the patient with the tumor cells directly or indirectly in vitro (step 1), and the contact with the peripheral blood mononuclear cells in step 1 above By the step of evaluating whether the tumor cells are injured (step 2), it is evaluated whether the tumor cells are injured. The present embodiment may be carried out by a human or may be carried out by the presentation device 10 or the auxiliary device 20 described later.

 「有用性」とは、患者に存在する悪性腫瘍を免疫療法剤を用いて治療するにあたり、前記免疫療法剤が患者にとって好ましい結果をもたらすことを意図する。有用性には、例えば、免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の有無、及び/又は免疫療法剤の治療効果の有無等が含まれ得る。 “Usefulness” means that the immunotherapeutic agent brings favorable results to the patient when treating a malignant tumor existing in the patient with the immunotherapeutic agent. The usefulness can include, for example, the presence or absence of an adverse event resulting from the immunotherapeutic agent that requires treatment interruption and that makes it impossible to resume administration, and/or the therapeutic effect of the immunotherapeutic agent. ..

 有害事象とは、免疫療法剤の投与に起因して起こる患者にとって有害な事象である。有害事象とは、例えば、間質性肺疾患(好ましくは、患者が免疫療法剤投与開始時に既に間質性肺炎を発症している場合には、間質性肺炎の増悪を含む、以下有害事象において同じ)等の肺障害;大腸炎、下痢、腸閉塞等の腸障害;皮膚粘膜眼症候群等の上皮系組織障害;多形紅斑;類天疱瘡;神経障害(末梢性ニューロパチー、ギラン・バレー症候群等);肝機能障害、肝炎、硬化性胆管炎等の肝胆管障害;甲状腺機能障害(症候性の甲状腺機能低下症、甲状腺機能亢進症、甲状腺炎等)、下垂体機能障害(症候性の下垂体炎、症候性の下垂体機能低下症等)、副腎機能障害(症候性の副腎機能不全、副腎クリーゼ等)等の内分泌障害;1 型糖尿病(劇症1 型糖尿病を含む)等の血糖値異常;腎障害(腎不全、尿細管間質性腎炎等);膵炎;筋炎、横紋筋融解症、重症筋無力症、心筋炎等の筋障害;心臓障害;脳炎、髄膜炎、脳症、白質脳症等の中枢神経系障害;免疫性血小板減少性紫斑病、溶血性貧血、赤芽球癆等の造血器障害;中毒性表皮壊死融解症(Toxic Epidermal Necrolysis:TEN);Infusion reaction(発熱、悪寒、そう痒、発疹、高血圧、低血圧、呼吸困難、過敏症(アナフィラキシーを含む)等)及び静脈血栓塞栓症(深部静脈血栓症、肺塞栓症)よりなる群から選択される少なくとも一種を挙げることができる。ここで、前記障害、又は疾患は無症候性であっても、症候性であってもよい。 Adverse events are adverse events for patients that result from the administration of immunotherapeutic agents. Adverse events include, for example, interstitial lung disease (preferably, if the patient has already developed interstitial pneumonia at the start of immunotherapeutic agent administration, the following adverse events including exacerbation of interstitial pneumonia). Same)); intestinal disorders such as colitis, diarrhea, intestinal obstruction; epithelial tissue disorders such as mucocutaneous ocular syndrome; erythema multiforme; pemphigoid; neuropathy (peripheral neuropathy, Guillain-Barre syndrome, etc.) ); Hepatobiliary disorders such as liver dysfunction, hepatitis, and sclerosing cholangitis; thyroid dysfunction (symptomatic hypothyroidism, hyperthyroidism, thyroiditis, etc.), pituitary dysfunction (symptomatic pituitary gland) Endocrine disorders such as inflammation, symptomatic hypopituitarism) and adrenal dysfunction (symptomatic adrenal insufficiency, adrenal crisis, etc.); abnormal blood glucose levels such as type 1 diabetes (including fulminant type 1 diabetes) Renal disorders (renal insufficiency, renal interstitial nephritis, etc.); pancreatitis; myopathy such as myositis, rhabdomyolysis, myasthenia gravis, myocarditis; heart disorder; encephalitis, meningitis, encephalopathy, white matter Central nervous system disorders such as encephalopathy; hematopoietic disorders such as immune thrombocytopenic purpura, hemolytic anemia, and erythroblastosis; toxic epidermal necrolysis (TEN); Infusion reaction (fever, chills) , Pruritus, rash, hypertension, hypotension, dyspnea, hypersensitivity (including anaphylaxis), and venous thromboembolism (deep vein thrombosis, pulmonary embolism) You can Here, the disorder or disease may be asymptomatic or symptomatic.

 治療中断(若しくは治療中止)の必要がある有害事象とは、免疫療法剤による治療中断(若しくは治療中止)の必要がある有害事象であり、例えば、間質性肺疾患(CTCAE v4.0における肺臓炎のGrade 1、Grade 2、Grade 3、Grade 4、又はGrade 5)等の肺障害;大腸炎、下痢、腸閉塞(CTCAE v4.0における大腸炎及び下痢のGrade 2、Grade 3、Grade 4、又はGrade 5、腸閉塞のGrade 1、Grade 2、Grade 3、Grade 4、又はGrade 5)等の腸障害;皮膚粘膜眼症候群(Stevens-Johnson 症候群)等の上皮系組織障害(CTCAE v4.0におけるGrade 3、Grade 4、又はGrade 5);多形紅斑(CTCAE v4.0におけるGrade 1、Grade 2、Grade 3、Grade 4、又はGrade 5);類天疱瘡;神経障害(末梢性ニューロパチー、ギラン・バレー症候群等)(CTCAE v4.0における神経障害のGrade 2、Grade 3、Grade 4、又はGrade 5);肝機能障害、肝炎、硬化性胆管炎(CTCAE v4.0における肝機能検査値上昇のGrade 2、Grade 3、Grade 4、又はGrade 5)等の肝胆管障害;甲状腺機能障害(症候性の甲状腺機能低下症、甲状腺機能亢進症、甲状腺炎等)、下垂体機能障害(症候性の下垂体炎、症候性の下垂体機能低下症等)、副腎機能障害(症候性の副腎機能不全、副腎クリーゼ等)等の内分泌障害;1 型糖尿病(劇症1 型糖尿病を含む)の血糖値異常;腎障害(腎不全、尿細管間質性腎炎等)(CTCAE v4.0における急性腎障害のGrade 2、Grade 3、Grade 4、又はGrade 5);膵炎;筋炎、横紋筋融解症、重症筋無力症、心筋炎等の筋障害;心臓障害;脳炎、髄膜炎、脳症、白質脳症等の中枢神経系障害;免疫性血小板減少性紫斑病、溶血性貧血、赤芽球癆等の造血器障害;中毒性表皮壊死融解症(Toxic Epidermal Necrolysis:TEN);Infusion reaction(発熱、悪寒、そう痒、発疹、高血圧、低血圧、呼吸困難、過敏症(アナフィラキシーを含む)等)、及び静脈血栓塞栓症(深部静脈血栓症、肺塞栓症)よりなる群から選択される少なくとも一種を挙げることができる。ここで、前記障害、又は疾患は無症候性であっても、症候性であってもよい。 An adverse event that requires treatment interruption (or treatment discontinuation) is an adverse event that requires treatment interruption (or treatment discontinuation) with an immunotherapeutic agent, such as interstitial lung disease (CTCAE v4.0 lung). Pulmonary disorders such as Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5) of inflammation; colitis, diarrhea, bowel obstruction (CTCAE v4.0 colitis and diarrhea Grade 2, Grade 3, Grade 4), or Grade 5, bowel obstruction Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5 and other intestinal disorders; cutaneous mucosal-eye syndrome (Stevens-Johnson syndrome) and other epithelial tissue disorders (CTCAE v4.0 Grade 3) , Grade 4, or Grade 5); erythema multiforme (Grade 1, Grade 2, Grade 3, Grade 4, or Grade 5 in CTCAE v4.0); pemphigoid; neuropathy (peripheral neuropathy, Guillain-Barre syndrome) Etc.) (Grade 2, Grade 3, Grade 4, or Grade 5 of neuropathy in CTCAE v4.0); liver dysfunction, hepatitis, sclerosing cholangitis (Grade 2, with increased liver function test value in CTCAE v4.0, Hepatobiliary disorders such as Grade 3, Grade 4, or Grade 5); thyroid dysfunction (symptomatic hypothyroidism, hyperthyroidism, thyroiditis, etc.), pituitary dysfunction (symptomatic hypophysitis, Endocrine disorders such as symptomatic hypopituitarism) and adrenal insufficiency (symptomatic adrenal insufficiency, adrenal crisis, etc.); abnormal blood glucose level of type 1 diabetes (including fulminant type 1 diabetes); renal disorder (Kidney failure, renal interstitial nephritis, etc.) (Grade 2, Grade 3, Grade 4, or Grade 5 of acute kidney injury in CTCAE v4.0); Pancreatitis; Myositis, rhabdomyolysis, myasthenia gravis , Myopathy such as myocarditis; heart disorder; central nervous system disorders such as encephalitis, meningitis, encephalopathy, leukoencephalopathy; hematopoietic disorders such as immune thrombocytopenic purpura, hemolytic anemia, and erythroblastosis; Toxic epidermal necrolysis (TEN); Infusion reaction (fever, chills, pruritus, rash, hypertension, hypotension, dyspnea, hypersensitivity (including anaphylaxis), etc., and venous thromboembolism (TEN). Deep vein thrombosis, pulmonary embolism) There may be at least one selected. Here, the disorder or disease may be asymptomatic or symptomatic.

 治療中断の必要があり、かつ投与再開が不可能となる有害事象(重篤な有害事象)とは、免疫療法剤による治療中断(若しくは治療中止)の必要があり、かつ免疫療法剤の投与再開が不可能となる有害事象であり、例えば、間質性肺疾患(CTCAE v4.0における肺臓炎のGrade 1、Grade 2、Grade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状が改善しない場合)等の肺障害;大腸炎、下痢、(CTCAE v4.0における大腸炎/下痢のGrade 2、Grade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状がGrade 1まで改善しない場合)、腸閉塞等の腸障害;皮膚粘膜眼症候群(Stevens-Johnson 症候群)等の上皮系組織障害(CTCAE v4.0における発疹のGrade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状がGrade 1まで改善しない場合);多形紅斑;類天疱瘡;神経障害(末梢性ニューロパチー、ギラン・バレー症候群等)(CTCAE v4.0における神経障害のGrade 2、Grade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状がベースライン状態まで改善しない場合);肝機能障害、肝炎、硬化性胆管炎(CTCAE v4.0における肝機能検査値上昇のGrade 2、Grade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても肝機能がベースライン状態まで改善しない場合)等の肝胆管障害;甲状腺機能障害(症候性の甲状腺機能低下症、症候性の甲状腺機能亢進症、症候性の甲状腺炎等の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状が改善しない場合)、下垂体機能障害(症候性の下垂体炎、症候性の下垂体機能低下症等)、副腎機能障害(症候性の副腎機能不全、副腎クリーゼ等)等の内分泌障害が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状が改善しない場合;1 型糖尿病(劇症1 型糖尿病を含む)の血糖値異常;腎障害(腎不全、尿細管間質性腎炎等)(CTCAE v4.0における急性腎障害のGrade 2、Grade 3、又はGrade 4の症状が出現し、悪性腫瘍の免疫療法剤の投与を中断しても、或いは投与を中断し対処療法を行っても症状がGrade 1まで改善しない場合);膵炎;筋炎、横紋筋融解症、重症筋無力症、心筋炎等の筋障害;心臓障害;脳炎、髄膜炎、脳症、白質脳症等の中枢神経系障害;免疫性血小板減少性紫斑病、溶血性貧血、赤芽球癆等の造血器障害;中毒性表皮壊死融解症(Toxic Epidermal Necrolysis:TEN);高血圧(Grade 2、Grade 3、Grade 4、又はGrade 5);低血圧(Grade 3、Grade 4、又はGrade 5);呼吸困難(Grade 2、Grade 3、Grade 4、又はGrade 5);アナフィラキシー及び静脈血栓塞栓症(深部静脈血栓症、肺塞栓症)よりなる群から選択される少なくとも一種を挙げることができる。また、治療中断の必要があり、かつ投与再開が不可能となる有害事象(重篤な有害事象)には、死亡する場合を含む。 Adverse events (serious adverse events) that require discontinuation of treatment and incapable of resuming administration include treatment interruption (or discontinuation of treatment) with immunotherapeutic agents and resumption of administration of immunotherapeutic agents. It is an adverse event that makes it impossible to, for example, interstitial lung disease (Grade 1, Grade 2, Grade 3, or Grade 4 of pneumonitis in CTCAE v4.0 appears, and immunotherapeutic agent for malignant tumor Disorders such as discontinuation of administration of the drug or discontinuation of administration and coping therapy does not improve symptoms); colitis, diarrhea, (CTCAE v4.0 colitis/diarrhea Grade 2, Grade) 3 or Grade 4 symptoms appear, and even if the administration of the immunotherapeutic agent for malignant tumor is interrupted, or the symptoms do not improve to Grade 1 even if the administration is discontinued and coping therapy is performed), bowel obstruction, etc. Disorders: Cutaneous mucosal-eye syndrome (Stevens-Johnson syndrome) and other epithelial tissue disorders (Grade 3 or Grade 4 of rash in CTCAE v4.0 appear, and administration of immunotherapeutic agent for malignant tumor is discontinued. Or, if the symptoms do not improve to Grade 1 even after discontinuation of administration and coping therapy); erythema multiforme; pemphigus vulgaris; neuropathy (peripheral neuropathy, Guillain-Barre syndrome, etc.) (CTCAE v4.0 Symptoms of Grade 2, Grade 3, or Grade 4 of neuropathy appear, and even if the administration of the immunotherapeutic agent for malignant tumor is discontinued, or the treatment is discontinued and coping therapy is applied, the symptoms improve to the baseline state. If not); hepatic dysfunction, hepatitis, sclerosing cholangitis (Grade 2, Grade 3, or Grade 4 symptoms of elevated liver function test value in CTCAE v4.0 appear, and immunotherapeutic agent for malignant tumor is administered. Hepatobiliary disorders such as hepatic function not improving to baseline even after discontinuation or discontinuation of administration and coping therapy; thyroid dysfunction (symptomatic hypothyroidism, symptomatic thyroid function) Symptoms such as hyperactivity and symptomatic thyroiditis appear, and the symptoms do not improve even if the administration of immunotherapeutic agents for malignant tumors is stopped, or if the treatment is stopped and coping therapy is performed), pituitary function Endocrine disorders such as disorders (symptomatic pituitaryitis, symptomatic hypopituitarism, etc.), adrenal dysfunction (symptomatic adrenal insufficiency, adrenal crisis, etc.) appear, and immunotherapeutic agents for malignant tumors appear. Symptoms do not improve after discontinuation of administration or discontinuation of administration and coping therapy Case: Abnormal blood glucose level of type 1 diabetes (including fulminant type 1 diabetes); renal disorder (renal failure, tubulointerstitial nephritis, etc.) (Grade 2, Grade 3, or acute renal disorder in CTCAE v4.0) Grade 4 symptoms appear, and if the administration of the immunotherapeutic agent for malignant tumor is discontinued, or if the treatment is discontinued and coping therapy is used, the symptoms do not improve to Grade 1); pancreatitis; myositis, striated muscle Myopathy such as lysis, myasthenia gravis, myocarditis; heart disorders; central nervous system disorders such as encephalitis, meningitis, encephalopathy, leukoencephalopathy; immune thrombocytopenic purpura, hemolytic anemia, erythroblasts Hematopoietic disorders such as lichen; toxic epidermal necrolysis (TEN); hypertension (Grade 2, Grade 3, Grade 4 or Grade 5); hypotension (Grade 3, Grade 5) or Grade 4 or Grade 5 or At least one selected from the group consisting of dyspnea (Grade 2, Grade 3, Grade 4, or Grade 5); anaphylaxis and venous thromboembolism (deep vein thrombosis, pulmonary embolism). In addition, adverse events (serious adverse events) that require discontinuation of treatment and incapable of resuming administration include death.

 本開示において、免疫療法剤は、T細胞の腫瘍細胞に対する傷害活性を高めることにより悪性腫瘍を治療ができる限り制限されない。ここで、治療することには、悪性腫瘍を改善すること、治癒すること及び/又は予防することが含まれ得る。好ましくは、治療することとは、悪性腫瘍を改善すること、及び/又は治癒することである。より好ましくは、治療することには、悪性腫瘍を改善すること、又は治癒することが含まれる。ここで改善することとは、悪性腫瘍の大きさの縮小又は拡大の抑制、及び/又は転移巣の縮小又は拡大の抑制等を含む。治癒することには、悪性腫瘍細胞の消失、好ましくは、1年間、2年間、3年間、5年間、又は7年間以上再発がないことを含む。予防することには、悪性腫瘍の再発を起こさないことが含まれる。前記再発は、悪性腫瘍の原発巣における再発であっても、転移巣における再発であってもよい。 In the present disclosure, the immunotherapeutic agent is not limited as long as it can treat a malignant tumor by increasing the damaging activity of T cells on tumor cells. Here, treating may include ameliorating, curing and/or preventing a malignant tumor. Preferably, treating is ameliorating and/or curing a malignant tumor. More preferably, treating includes ameliorating or curing a malignant tumor. Improving here includes suppressing the size reduction or expansion of malignant tumors, and/or suppressing the reduction or expansion of metastases. Curing includes the disappearance of malignant tumor cells, preferably the absence of recurrence for more than 1 year, 2 years, 3 years, 5 years, or 7 years. Preventing includes preventing recurrence of malignant tumors. The recurrence may be a recurrence in the primary lesion of the malignant tumor or a recurrence in the metastatic lesion.

 本開示における免疫療法剤は、悪性腫瘍の免疫療法に使用できる限り制限されない。免疫療法剤は、後述する「8.免疫療法剤」で述べる免疫療法剤を挙げることができる。 The immunotherapy agent in the present disclosure is not limited as long as it can be used for immunotherapy of malignant tumor. Examples of the immunotherapeutic agent include the immunotherapeutic agents described in "8. Immunotherapeutic agents" described later.

 本開示において、悪性腫瘍には、上皮性悪性腫瘍及び非上皮性悪性腫瘍が含まれるが、好ましくは上皮性悪性腫瘍である。 In the present disclosure, malignant tumors include epithelial malignant tumors and non-epithelial malignant tumors, preferably epithelial malignant tumors.

 患者が有する悪性腫瘍としては、例えば、気管、気管支又は肺等から発生する呼吸器系悪性腫瘍;上咽頭、食道、胃、十二指腸、空腸、回腸、盲腸、虫垂、上行結腸、横行結腸、S状結腸、直腸又は肛門部等から発生する消化管系悪性腫瘍;肝臓癌;膵臓癌;膀胱、尿管又は腎臓から発生する泌尿器系悪性腫瘍;卵巣、卵管及び子宮等のから発生する女性生殖器系悪性腫瘍;乳癌:前立腺癌;皮膚癌;視床下部、下垂体、甲状腺、副甲状腺、副腎等の内分泌系悪性腫瘍;中枢神経系悪性腫瘍;骨軟部組織から発生する悪性腫瘍等の固形腫瘍、悪性黒色腫、及び骨髄異形成症候群、急性リンパ性白血病、急性骨髄性白血病、慢性リンパ性白血病、慢性骨髄性白血病、急性骨髄単球性白血病、慢性骨髄単球性白血病、急性単球性白血病、慢性単球性白血病、急性全骨髄性白血病、急性巨核球性白血病、赤白血病、好酸球性白血病、慢性好酸球性白血病、慢性好中球性白血病、成人T細胞白血病、ヘアリー細胞白血病、形質細胞性白血病、多発性骨髄腫、悪性リンパ腫等の造血系悪性腫瘍;リンパ系悪性腫瘍等の造血器腫瘍が挙げられる。より好ましくは、肺癌(扁平上皮癌、小細胞癌、大細胞癌、腺癌)等の呼吸器系上皮性悪性腫瘍;胃癌、十二指腸癌、大腸癌(S上結腸癌、直腸癌等)等の消化管系上皮性悪性腫瘍;肝臓癌;膵臓癌;膀胱癌;甲状腺癌;卵巣癌;乳癌:前立腺癌;頭頚部癌;悪性黒色腫;腎細胞癌、尿路上皮癌、古典的ホジキンリンパ腫、メルケル細胞癌又は悪性胸膜中皮腫を挙げることができる。最も好ましくは、肺癌である。肺癌は、免疫療法剤の投与開始時に間質性肺炎を伴っていてもよい。 Examples of malignant tumors that a patient has include respiratory malignant tumors that develop from the trachea, bronchi, lungs, etc.; nasopharynx, esophagus, stomach, duodenum, jejunum, ileum, cecum, appendix, ascending colon, transverse colon, sigmoid Gastrointestinal malignant tumors arising from the colon, rectum or anus; liver cancer; pancreatic cancer; urinary system malignant tumors arising from the bladder, ureter or kidney; female reproductive system arising from ovaries, fallopian tubes, uterus, etc. Malignant tumor; Breast cancer: Prostate cancer; Skin cancer; Endocrine malignant tumor such as hypothalamus, pituitary gland, thyroid, parathyroid gland, adrenal gland, central nervous system malignant tumor, solid tumor such as malignant tumor originating from bone and soft tissue, malignant tumor Melanoma and myelodysplastic syndrome, acute lymphocytic leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, acute myelomonocytic leukemia, chronic myelomonocytic leukemia, acute monocytic leukemia, chronic Monocytic leukemia, acute myelocytic leukemia, acute megakaryocytic leukemia, erythroleukemia, eosinophilic leukemia, chronic eosinophilic leukemia, chronic neutrophilic leukemia, adult T cell leukemia, hairy cell leukemia, trait Hematopoietic malignancies such as cell leukemia, multiple myeloma and malignant lymphoma; hematopoietic tumors such as lymphoid malignant tumors. More preferably, respiratory epithelial malignant tumor such as lung cancer (squamous cell cancer, small cell cancer, large cell cancer, adenocarcinoma); gastric cancer, duodenal cancer, colon cancer (S colon cancer, rectal cancer, etc.) Gastrointestinal epithelial malignancies; liver cancer; pancreatic cancer; bladder cancer; thyroid cancer; ovarian cancer; breast cancer: prostate cancer; head and neck cancer; malignant melanoma; renal cell carcinoma, urothelial cancer, classic Hodgkin lymphoma, Mention may be made of Merkel cell carcinoma or malignant pleural mesothelioma. Most preferably, it is lung cancer. The lung cancer may be associated with interstitial pneumonia at the start of administration of the immunotherapeutic agent.

 また、肺癌には、扁平上皮癌、小細胞癌、大細胞癌又は腺癌を含む。好ましくは、非小細胞性の肺癌である。 Also, lung cancer includes squamous cell carcinoma, small cell carcinoma, large cell carcinoma or adenocarcinoma. Preferred is non-small cell lung cancer.

 本発明において「患者」は、免疫療法剤が適用されうる者である限り制限されない。例えば、患者としては、上記に記載のいずれかの悪性腫瘍を有し、かつ未治療である患者、又は既に何らかの悪性腫瘍の治療を受けた患者、悪性腫瘍の治療中の患者、悪性腫瘍が再発又は転移している患者を挙げることができる。前記、悪性腫瘍の治療には、好ましくは、外科的腫瘍切除、化学療法(より好ましくは、免疫療法剤以外の抗がん剤投与による化学療法)、放射療法等を含む。また患者は、好ましくは、投与される免疫療法剤の適用に応じて、例えば非小細胞癌性の悪性腫瘍;切除不能な進行性の非小細胞癌性の悪性腫瘍、及び/又は再発性の非小細胞癌性の悪性腫瘍;切除不能な悪性黒色腫;切除不能又は転移性の腎細胞癌);再発又は難治性の古典的ホジキンリンパ腫;再発又は遠隔転移を有する頭頸部癌;がん化学療法後に増悪した治癒切除不能な進行又は再発の胃癌;がん化学療法後に増悪した切除不能な進行又は再発の悪性胸膜中脾腫;がん化学療法後に増悪した切除不能な尿路上皮癌;切除不能なメルケル細胞癌)を有する患者等から選択されてもよい。ここで、「切除不能」には、好ましくは根治切除不能又は治癒切除不能な状態を含む。具体的な免疫療法剤との組み合わせは、後述する「8.免疫療法剤」で説明する。 In the present invention, the “patient” is not limited as long as it is a person to whom an immunotherapy agent can be applied. For example, as a patient, a patient who has any of the malignant tumors described above and is untreated, a patient who has already been treated for some malignant tumor, a patient who is undergoing treatment for a malignant tumor, or a malignant tumor has recurred. Or a patient with metastasis can be mentioned. The above-mentioned treatment of malignant tumor preferably includes surgical tumor resection, chemotherapy (more preferably, chemotherapy by administration of an anti-cancer agent other than immunotherapeutic agent), radiation therapy and the like. The patient is also preferably dependent on the application of the administered immunotherapeutic agent, eg, non-small cell carcinoma malignancies; unresectable, advanced non-small cell carcinoma malignancies, and/or recurrent Non-small cell malignancies; unresectable malignant melanoma; unresectable or metastatic renal cell carcinoma); relapsed or refractory classic Hodgkin lymphoma; head and neck cancer with recurrent or distant metastases; cancer chemistry Unresectable advanced or recurrent gastric cancer that worsened after therapy; unresectable advanced or recurrent malignant pleural splenomegaly that worsened after cancer chemotherapy; unresectable urothelial cancer that worsened after cancer chemotherapy; unresectable Merkel cell carcinoma). Here, "unresectable" preferably includes radical unresectable or curable unresectable state. Specific combinations with immunotherapeutic agents will be described in “8. Immunotherapeutic agents” described later.

 本発明において末梢血は、T細胞を含む単核細胞を取得できる限り制限されない。末梢血は、動脈血であっても静脈血であってもよいが、好ましくは静脈血である。末梢血の採血は、例えば四肢の血管から行うことができる。また、手術や生検の際に、四肢以外の血管から採血しても良い。 In the present invention, peripheral blood is not limited as long as mononuclear cells including T cells can be obtained. The peripheral blood may be arterial blood or venous blood, but is preferably venous blood. Peripheral blood can be collected, for example, from the blood vessels of the extremities. In addition, blood may be collected from blood vessels other than the extremities at the time of surgery or biopsy.

 末梢血の採血方法は、単核細胞を取得できる限り制限されないが、抗凝固剤を使用して採血することが好ましい。抗凝固剤としては、エチレンジアミン四酢酸塩、クエン酸塩、ヘパリン塩等を挙げることができる。好ましくはヘパリンである。 The method of collecting peripheral blood is not limited as long as mononuclear cells can be obtained, but it is preferable to collect blood using an anticoagulant. Examples of the anticoagulant include ethylenediaminetetraacetate, citrate, heparin salt and the like. Heparin is preferred.

 PBMCは、公知の方法にしたがって取得することができる。例えば、ヒトリンパ球分離用の比重液(1.077±0.001g/ml程度の密度を有する)を使って遠心分離により取得することができる。また、溶血剤を使って末梢血中の赤血球を溶血させ、得られる有核細胞をPBMCとして使用してもよい。あるいは、セルソータ、磁気細胞分離法を使って所望の細胞を取得してもよい。前記所望の細胞としては、例えば、T細胞、好ましくはCD3陽性T細胞又はCD8陽性T細胞を挙げることができる。取得されたPBMCは、取得後培養等の工程を経ることなく腫瘍細胞と接触させることが好ましい。例えば、PBMCを取得してから、24時間以内、18時間以内、12時間以内、6時間以内、4時間以内、3時間以内、2時間以内、1時間以内、30分以内、又は15分以内に腫瘍細胞と接触させることが好ましい。 -PBMC can be obtained according to a known method. For example, it can be obtained by centrifugation using a specific gravity liquid for human lymphocyte separation (having a density of about 1.077±0.001 g/ml). Alternatively, the red blood cells in the peripheral blood may be hemolyzed using a hemolytic agent, and the resulting nucleated cells may be used as PBMC. Alternatively, desired cells may be obtained using a cell sorter or magnetic cell separation method. Examples of the desired cells include T cells, preferably CD3 positive T cells or CD8 positive T cells. The obtained PBMC is preferably brought into contact with tumor cells without undergoing a step such as culturing after the acquisition. For example, within 24 hours, within 18 hours, within 12 hours, within 6 hours, within 4 hours, within 3 hours, within 2 hours, within 1 hour, within 30 minutes, or within 15 minutes after obtaining PBMC. Contacting with tumor cells is preferred.

 PBMCと接触する腫瘍細胞は、上記腫瘍に由来する細胞である限り制限されない。腫瘍細胞として好ましくは、培養細胞株を挙げることができる。培養細胞株としては特に制限されないが、例えば、U251細胞株、RERF細胞株、A549細胞株等の悪性腫瘍由来の培養細胞株を挙げることができる。前記腫瘍細胞は、患者が有する悪性腫瘍と同種の細胞である必要はないが、同種の細胞であってもよい。好ましくは、PBMCと同じ培養条件で発育できる細胞であることが好ましい。 The tumor cells that come into contact with PBMC are not limited as long as they are cells derived from the above tumor. A preferable example of the tumor cell is a cultured cell line. The cultured cell line is not particularly limited, and examples thereof include cultured cell lines derived from malignant tumors such as U251 cell line, RERF cell line, and A549 cell line. The tumor cells do not have to be cells of the same species as the malignant tumor of the patient, but may be cells of the same species. Preferably, the cells can grow under the same culture conditions as PBMC.

 in vitroにおけるPBMCと、腫瘍細胞との接触方法は、PBMCと腫瘍細胞が直接、又は間接的に接触する限り制限されない。 The contact method between PBMC and tumor cells in vitro is not limited as long as PBMC and tumor cells directly or indirectly contact each other.

 in vitroにおいて、PBMCと腫瘍細胞とを接触させる際には、腫瘍細胞を予めin vitroでプレート、又はカルチャーボトルに播種しておくことが好ましい。腫瘍細胞が接着性の細胞である場合には、前記腫瘍細胞が、in vitroで生存可能な状態で、シャーレ、又はカルチャーボトルに接着している状態であることがより好ましい。さらには、前記腫瘍細胞が接着性の細胞である場合には、前記腫瘍細胞が発育可能な条件下(例えば、35℃~37℃、5%程度の炭酸ガス存在下)で12時間~48時間程度培養された状態であることが好ましい。前記腫瘍細胞を培養する培養培地は、腫瘍細胞が生存できる、又は発育できる培地である限り制限されない。例えば、ウシ胎児血清を8%~20%程度含む、RPMI1640培地(改変培地含む)、α-MEM培地(改変培地含む)、ダルベッコMEM培地(改変培地含む)等を挙げることができる。 When contacting PBMCs with tumor cells in vitro, it is preferable to seed the tumor cells in vitro on a plate or culture bottle in advance. When the tumor cells are adhesive cells, it is more preferable that the tumor cells are in a viable state in vitro and adhere to a petri dish or a culture bottle. Furthermore, when the tumor cells are adherent cells, the tumor cells can grow for 12 hours to 48 hours under conditions (eg, 35° C. to 37° C. in the presence of about 5% carbon dioxide gas). It is preferably in a state of being cultured to some extent. The culture medium for culturing the tumor cells is not limited as long as the tumor cells can survive or develop. For example, RPMI1640 medium (including modified medium), α-MEM medium (including modified medium), Dulbecco MEM medium (including modified medium), etc. containing about 8% to 20% fetal bovine serum can be mentioned.

 PBMCと腫瘍細胞を接触させる際のPBMCの細胞数は、例えば96ウェルプレート1ウェルに対して、1×10~1×10個程度、好ましくは5×10~5×10個程度の腫瘍細胞に対して、PBMCが1×10~1×10個程度、好ましくは5×10~5×10個程度となるように接触させることが好ましい。 The number of PBMC cells when contacting PBMCs with tumor cells is, for example, about 1×10 3 to 1×10 5 , preferably about 5×10 3 to 5×10 4 cells per well of a 96-well plate. It is preferable that the tumor cells are brought into contact with PBMC at about 1×10 3 to 1×10 5 , preferably about 5×10 3 to 5×10 4 .

 PBMCと腫瘍細胞を直接接触させる方法としては、腫瘍細胞が播種されたプレート、又はカルチャーボトルに、PBMCを播種する方法を挙げることができる。好ましくは、播種したPBMCが浮遊している場合には、1,000から2,000 r.p.m.程度で5から10分程度遠心することができる。 As a method of directly contacting PBMC with tumor cells, a method of seeding PBMC on a plate or culture bottle seeded with tumor cells can be mentioned. Preferably, if the seeded PBMC are floating, then 1,000 to 2,000 r.p.m. p. m. It can be centrifuged for about 5 to 10 minutes.

 PBMCと腫瘍細胞を間接的に接触させる方法としては、エンゲージャーを介して接触させる方法を挙げることができる。本実施形態においてエンゲージャーとは、PBMCを、好ましくはリンパ球を、より好ましくはT細胞を、さらに好ましくはCD8陽性T細胞(細胞障害性T細胞)を腫瘍細胞にエンゲージできる分子である限り制限されない。エンゲージとは、少なくとも細胞同士(例えば、T細胞と腫瘍細胞)を架橋すること、好ましくは、T細胞と腫瘍細胞とをエンゲージャーを介して結合させることをいい、好ましくは、細胞障害性T細胞が架橋した腫瘍細胞に対して細胞傷害活性を示すことができる状態をいう。 As a method for indirectly contacting PBMC with tumor cells, a method for contacting them via an engager can be mentioned. In the present embodiment, the term "engager" is limited as long as it is a molecule capable of engaging PBMC, preferably lymphocytes, more preferably T cells, and even more preferably CD8-positive T cells (cytotoxic T cells) with tumor cells. Not done. Engage means cross-linking at least cells (for example, T cells and tumor cells), preferably binding T cells and tumor cells via an engager, and preferably cytotoxic T cells Means a state capable of exhibiting cytotoxic activity against cross-linked tumor cells.

 エンゲージャーとして、好ましくは二重特異性分子、三重特異性分子等を挙げることができる。エンゲージャーとしてより好ましくは二重特異性分子である。二重特性分子及び三重特異性分子は、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子であることが好ましい。前記抗原結合領域は、標的とする抗原と結合する限り制限されない。前記抗原結合領域は、例えば、前記抗原に結合する免疫グロブリンの相補鎖決定領域1(CDR1)、相補鎖決定領域2(CDR2)及び相補鎖決定領域3(CDR3)よりなる群からなる少なくとも一つのCDRを含む。前記抗原結合領域は、好ましくは、CDR1、CDR2及びCDR3よりなる群からなる少なくとも二つのCDRを含み、より好ましくは、CDR1、CDR2及びCDR3を含む。前記CDRは、免疫グロブリンの重鎖由来であっても、免疫グロブリンの軽鎖由来であってもよい。さらに好ましくは、前記抗原結合領域において、各CDRは、それぞれに対応するフレームワーク領域に隣接していてもよい。また、前記抗原結合領域は、免疫グロブリンの可変領域(V領域)、sc-Fv、免疫グロブリンのFab領域等であってもよい。 As the engager, preferably, a bispecific molecule, a trispecific molecule and the like can be mentioned. A bispecific molecule is more preferable as an engager. The bispecific molecule and the trispecific molecule are molecules containing an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. preferable. The antigen-binding region is not limited as long as it binds to the target antigen. The antigen-binding region is, for example, at least one of the group consisting of complementary chain determining region 1 (CDR1), complementary chain determining region 2 (CDR2) and complementary chain determining region 3 (CDR3) of immunoglobulin that binds to the antigen. Includes CDR. The antigen-binding region preferably comprises at least two CDRs consisting of CDR1, CDR2 and CDR3, more preferably CDR1, CDR2 and CDR3. The CDR may be derived from an immunoglobulin heavy chain or an immunoglobulin light chain. More preferably, in the antigen-binding region, each CDR may be adjacent to the corresponding framework region. The antigen-binding region may be an immunoglobulin variable region (V region), sc-Fv, an immunoglobulin Fab region, or the like.

 表面抗原の少なくとも一種と結合する抗原結合領域は、1つの抗原に対して、1つであっても2以上であってもよい。例えば、表面抗原の少なくとも一種と結合する抗原結合領域は、同種のFab領域の組み合わせ、同種の可変領域の組み合わせ、同種のsc-Fvの組み合わせ、1種の抗体に由来する重鎖Fab領域と軽鎖Fab領域の組み合わせ1種の抗体に由来する重鎖可変領域と軽鎖可変領域の組み合わせ、1種の抗体に由来する重鎖sc-Fvの組み合わせであってもよい。 The number of antigen-binding regions that bind to at least one surface antigen may be one or two or more for one antigen. For example, an antigen-binding region that binds to at least one surface antigen is a combination of Fab regions of the same species, a combination of variable regions of the same species, a combination of sc-Fv of the same species, a heavy chain Fab region derived from one antibody, and a light chain Fab region. Combination of chain Fab regions Combination of heavy chain variable region and light chain variable region derived from one type of antibody may be a combination of heavy chain sc-Fv derived from one type of antibody.

 T細胞の表面抗原は、T細胞の表面に存在し、二重特異性分子又は三重特異性分子に含まれる少なくとも一つの抗原結合領域が結合できる限り制限されない。T細胞の表面抗原として、好ましくは細胞障害性T細胞の表面抗原を挙げることができる。例えば、T細胞の表面抗原は、CD3、CD8、TCR、CTLA-4、PD1、Tim3、CD27、CD28、CD40、CD134(OX40)、CD137(4-1BB)、CD278(ICOS)を挙げることができる。好ましくはCD3である。 The surface antigen of T cell is not limited as long as it exists on the surface of T cell and can bind to at least one antigen-binding region contained in the bispecific molecule or trispecific molecule. Preferable examples of the T cell surface antigen include cytotoxic T cell surface antigens. For example, T cell surface antigens include CD3, CD8, TCR, CTLA-4, PD1, Tim3, CD27, CD28, CD40, CD134 (OX40), CD137 (4-1BB), CD278 (ICOS). .. It is preferably CD3.

 腫瘍細胞の表面抗原は、腫瘍細胞の表面に存在し、二重特異性分子又は三重特異性分子に含まれる少なくとも一つの抗原結合領域が結合できる限り制限されない。腫瘍細胞の表面抗原としては、EphA1(ephrin type-A receptor 1)、EphA2、FolR1(folate receptor 1)、EpCAM(Epithelial cell adhesion molecule)、CD19、Her1 (v-erb-b2 erythroblastic leukemia viral oncogene homolog 1: ErbB1)、Her2、CD20、EGFR(Epidermal Growth Factor Receptor)、CCR4(C-C chemokine receptor type 4)、CEA(Carcinoembryonic antigen)、GD2、GD3,CD22、CD30、CD33、CD70、CD123、EGFRvIII、MUC1(Mucin1)、PSCA(Prostate stem cell antigen)、PSMA(Prostate-specific membrane antigen)、HLA-A1+NY-ESO1(HLA-A1 restricted NY-ESO1)、HLA-A2+NY-ESO1(HLA-A2 restricted NY-ESO1)、HLA-A3+NY-ESO1(HLA-A3 restricted NY-ESO1)等を挙げることができる。 The tumor cell surface antigen is not limited as long as it exists on the surface of the tumor cell and can bind to at least one antigen-binding region contained in the bispecific molecule or the trispecific molecule. As surface antigens of tumor cells, EphA1 (ephrin type-A receptor 1), EphA2, FolR1 (folate receptor 1), EpCAM (Epithelial cellulae berbereion molecule), CD19, Herb 1-herb (2). : ErbB1), Her2, CD20, EGFR (Epidermal Growth Factor Receptor), CCR4 (C-C chemokine receptor type 4), CEA (CarcinoembryonicCD, EG, CD2, CD2G, D22, G22, CDD, G22, CDG, G22, G22, G22, G22, G22, G2, G22, G22, G22, G22, G22, G2, G2, G2, G2, G2, G2, G2, G2, G2, G3, G2, G2, G2, G2, G2, G2, G2, G2, G2, G2, G2, G3, G2, G3, G2, G3, G2, G3, G2, G2, G2, G2. (Mucin1), PSCA (Prostate system cell antigen), PSMA (Prostate-specific membrane antigen), HLA-A1+NY-ESO1 (HLA-A1 resty-NY-HLA-A1+HN-ESO1+HLA-A2+HLA-A2+HLA-A2+HLA-A2). , HLA-A3+NY-ESO1 (HLA-A3 restricted NY-ESO1) and the like.

 二重特異性分子には、二重特異性T細胞エンゲージャー、二重特異性抗体等が含まれる。二重特異性T細胞エンゲージャーとしては、CD19とCD3を標的とするエンゲージャー、EphA2とCD3を標的とするエンゲージャーを挙げることができる。また二重特異性抗体としては、EpCAMとCD3を標的とする抗体を挙げることができる。 Bispecific molecules include bispecific T cell engagers, bispecific antibodies, etc. Examples of bispecific T cell engagers include those that target CD19 and CD3, and those that target EphA2 and CD3. Examples of bispecific antibodies include antibodies targeting EpCAM and CD3.

 PBMCと腫瘍細胞を接触させる際の細胞数は、例えば96ウェルプレート1ウェルに対して、1×10~1×10個程度、好ましくは5×10~5×10個程度の腫瘍細胞に対して、PBMCが1×10~1×10個程度、好ましくは5×10~5×10個程度となるように接触させることが好ましい。二重特異性分子を添加する場合には、96ウェルプレート1ウェルに対して50~200ng/mlの終濃度となるように添加することが好ましい。 The number of cells when contacting PBMCs with tumor cells is, for example, about 1×10 3 to 1×10 5 cells, preferably about 5×10 3 to 5×10 4 tumors per well of a 96-well plate. The cells are preferably contacted with about 1×10 3 to 1×10 5 PBMCs, preferably about 5×10 3 to 5×10 4 PBMCs. When the bispecific molecule is added, it is preferable to add it at a final concentration of 50 to 200 ng/ml per well of a 96-well plate.

 PBMCと腫瘍細胞の直接又は間接的な接触は、PBMCと腫瘍細胞のそれぞれが、通常発育可能な条件で行うことが好ましい。例えば、発育可能な条件下(例えば、35℃~37℃、5%程度の炭酸ガス存在下)で16時間~72時間、好ましくは36~50時間程度行うことができる。前記PBMCと腫瘍細胞を接触させる時の培養培地は、PBMCと腫瘍細胞が生存できる、又は発育できる培地である限り制限されない。例えば、ウシ胎児血清を8%~20%程度含む、RPMI1640培地(改変培地含む)を挙げることができる。 Direct or indirect contact between PBMC and tumor cells is preferably performed under conditions in which PBMC and tumor cells can normally develop. For example, it can be performed for 16 hours to 72 hours, preferably for 36 hours to 50 hours under conditions that allow growth (eg, 35° C. to 37° C. in the presence of 5% carbon dioxide gas). The culture medium for contacting the PBMC with the tumor cells is not limited as long as the culture medium allows the PBMC and the tumor cells to survive or develop. For example, RPMI1640 medium (including modified medium) containing about 8% to 20% fetal bovine serum can be mentioned.

 前記腫瘍細胞が傷害されているか否かの評価には、前記腫瘍細胞が傷害されていること、及び/又は前記腫瘍細胞が傷害されていないことを評価することが含まれる。好ましくは、前記腫瘍細胞が傷害されているか否かの評価には、前記腫瘍細胞が傷害されているか否かを決定することが含まれる。前記腫瘍細胞が傷害されているか否かを決定することには、前記腫瘍細胞が傷害されていると決定すること、及び/又は前記腫瘍細胞が傷害されていないと決定することが含まれる。前記腫瘍細胞が傷害されているか否かの評価結果には、腫瘍細胞が傷害されているか否かを示す情報として、後述する腫瘍細胞に対する細胞傷害活性(腫瘍細胞傷害活性)を反映する値と、前記値に対応する基準値に基づいて決定された腫瘍細胞が傷害されていることを示す情報(例えば「傷害あり」、「傷害陽性」、「+(プラス)」等)、又は腫瘍細胞が傷害されていないことを示す情報(例えば「傷害なし」、「傷害陰性」、「-(マイナス)」等)が含まれる。 Assessment of whether or not the tumor cells are injured includes assessing that the tumor cells are injured and/or that the tumor cells are not injured. Preferably, assessing whether the tumor cells are injured comprises determining whether the tumor cells are injured. Determining whether the tumor cells are injured includes determining that the tumor cells are injured and/or determining that the tumor cells are not injured. The evaluation result of whether the tumor cells are injured, as information indicating whether the tumor cells are injured, a value reflecting the cytotoxic activity against the tumor cells described below (tumor cytotoxic activity), Information indicating that the tumor cells are injured based on the reference value corresponding to the above value (for example, "injury present", "injury positive", "+ (plus)", etc.), or tumor cells are injured It includes information indicating that it has not been done (for example, "no injury", "negative injury", "-(minus)", etc.).

 好ましくは、前記腫瘍細胞が傷害されているか否かの評価することには、腫瘍細胞が傷害されているか否かを示す情報を取得することと、取得した情報に基づいて、前記腫瘍細胞が傷害されていること、及び/又は前記腫瘍細胞が傷害されていないことを評価することが含まれ得る。 Preferably, in assessing whether the tumor cells are injured, obtaining information indicating whether the tumor cells are injured, and based on the obtained information, the tumor cells are injured And/or assessing that the tumor cells are not injured.

 腫瘍細胞が傷害されているか否かを評価するために、前記患者から採取された末梢血単核細胞と、腫瘍細胞とを、直接、又は間接的にin vitroで接触させ後に、前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する。 In order to evaluate whether the tumor cells are injured or not, the peripheral blood mononuclear cells collected from the patient are directly or indirectly contacted with the tumor cells in vitro, and then the tumor cells are A value that reflects the tumor cytotoxic activity of contacted peripheral blood mononuclear cells is obtained.

 取得された腫瘍細胞傷害活性を反映する値は、その対応する基準値と比較される。比較の結果、前記腫瘍細胞傷害活性を反映する値が対応する基準値より高い場合、前記腫瘍細胞が傷害されていることを示す。比較の結果、前記腫瘍細胞傷害活性を反映する値が対応する基準値より低い場合、前記腫瘍細胞が傷害されていないことを示す。 The value that reflects the acquired tumor cytotoxic activity is compared with its corresponding reference value. As a result of the comparison, when the value reflecting the tumor cytotoxic activity is higher than the corresponding reference value, it indicates that the tumor cells are injured. As a result of the comparison, when the value reflecting the tumor cell cytotoxicity is lower than the corresponding reference value, it indicates that the tumor cells are not injured.

 前記基準値は、腫瘍細胞が傷害されているか否かを判定できる値である限り制限されない。例えば、基準値は、腫瘍免疫が活性化していない陰性対照のPBMCの腫瘍細胞傷害活性を反映する値や、腫瘍免疫が活性化している陽性対照のPBMCの腫瘍細胞傷害活性を反映する値から求めることができる。陰性対照のPBMCの腫瘍細胞傷害活性を反映する値、及び陽性対照のPBMCの腫瘍細胞傷害活性を反映する値を測定する方法は、治療対象患者のPBMCの腫瘍細胞傷害活性を反映する値を測定する方法と同じであることが好ましい。基準値は、陰性対照のPBMCの腫瘍細胞傷害活性を反映する値の上限値、陽性対照のPBMCの腫瘍細胞傷害活性を反映する値、陰性対照のPBMCの腫瘍細胞傷害活性を反映する値と陽性対照のPBMCの腫瘍細胞傷害活性を反映する値の中央値、平均値、最頻値等とすることができる。あるいは、基準値は、ROC曲線(Receiver Operatorating Characteristic curve、受信者動作特性曲線)、判別分析法、モード法、Kittler法、3σ法、p‐tile法等により算出してもよい。 The above-mentioned reference value is not limited as long as it can determine whether or not tumor cells are injured. For example, the reference value is obtained from a value that reflects the tumor cytotoxic activity of the negative control PBMC in which tumor immunity is not activated, or a value that reflects the tumor cytotoxic activity of the positive control PBMC in which tumor immunity is activated. be able to. The method for measuring the value reflecting the tumor cytotoxic activity of negative control PBMC and the value reflecting the tumor cytotoxic activity of positive control PBMC is performed by measuring the value reflecting the tumor cytotoxic activity of PBMC of the patient to be treated. The method is preferably the same as the method described above. The reference value is the upper limit of the value that reflects the tumor cytotoxic activity of the negative control PBMC, the value that reflects the tumor cytotoxic activity of the positive control PBMC, and the value that reflects the tumor cytotoxic activity of the negative control PBMC and is positive. The median, mean, mode, etc. of the values reflecting the tumor cytotoxic activity of the control PBMC can be used. Alternatively, the reference value may be calculated by a ROC curve (Receiver Operating Characteristic curve, receiver operating characteristic curve), discriminant analysis method, mode method, Kittler method, 3σ method, p-tile method, or the like.

 例えば、基準値は、腫瘍細胞傷害活性を反映する値として、例えば、後述する腫瘍細胞傷害活性の値を用いる場合には、前記基準値を生細胞数に基づく又は死細胞数に基づく腫瘍細胞傷害活性パーセントとして5%、10%、15%、20%、25%、30%、35%、40%、45%又は50%から選択してもよい。基準値は、治療中断の必要があり、かつ投与再開が不可能となる有害事象の有無を決定する場合と、治療効果の有無を決定する場合とで、同じであっても、異なっていてもよい。基準値が異なる場合に、治療中断の必要があり、かつ投与再開が不可能となる有害事象の有無を決定する場合の基準値を20%、25%、30%、35%、40%、45%又は50%から選択してもよい。基準値が異なる場合に、治療効果の有無を決定する場合の基準値を10%、15%、20%、25%、30%、35%又は40%から選択してもよい。 For example, the reference value is a value that reflects the tumor cytotoxic activity, and for example, when the value of the tumor cytotoxic activity described later is used, the reference value is based on the number of living cells or the number of dead cells. The percent activity may be selected from 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50%. The reference value may or may not be the same in the case of deciding the presence or absence of an adverse event that requires discontinuation of treatment and in which resumption of administration is impossible, and the case of deciding the presence or absence of a therapeutic effect. Good. 20%, 25%, 30%, 35%, 40%, 45% of the reference value when determining the presence or absence of adverse events that require treatment interruption and inability to resume administration when the reference values differ % Or 50% may be selected. When the reference values are different, the reference value for determining the presence or absence of the therapeutic effect may be selected from 10%, 15%, 20%, 25%, 30%, 35% or 40%.

 また基準値は、腫瘍細胞傷害活性を反映する値として、例えば、後述するインターフェロン-γの測定値を用いる場合には、1,000pg/ml、1,500pg/ml、2,000pg/ml、2,500pg/ml、3,000pg/ml、3,500pg/ml、4,000pg/ml、4,500pg/ml、5,000pg/ml、5,500pg/ml、6,000pg/ml、又は6,500pg/mlから選択しても良い。基準値は、治療中断の必要があり、かつ投与再開が不可能となる有害事象の有無を決定する場合と、治療効果の有無を決定する場合とで、同じであっても、異なっていてもよい。基準値が異なる場合に、治療中断の必要があり、かつ投与再開が不可能となる有害事象の有無を決定する場合の基準値を3,500pg/ml、4,000pg/ml、4,500pg/ml、5,000pg/ml、5,500pg/ml、6,000pg/ml、又は6,500pg/mlから選択しても良い。基準値が異なる場合に、治療効果の有無を決定する場合の基準値を2,000pg/ml、2,500pg/ml、3,000pg/ml、3,500pg/ml、4,000pg/ml、4,500pg/ml、5,000pg/ml、5,500pg/ml、6,000pg/ml、又は6,500pg/mlから選択しても良い。 The reference value is a value that reflects the tumor cytotoxicity. For example, when the measurement value of interferon-γ described later is used, 1,000 pg/ml, 1,500 pg/ml, 2,000 pg/ml, 2 , 500 pg/ml, 3,000 pg/ml, 3,500 pg/ml, 4,000 pg/ml, 4,500 pg/ml, 5,000 pg/ml, 5,500 pg/ml, 6,000 pg/ml, or 6, You may select from 500 pg/ml. The reference value may or may not be the same in the case of deciding the presence or absence of an adverse event that requires discontinuation of treatment and in which resumption of administration is impossible, and the case of deciding the presence or absence of a therapeutic effect. Good. If the reference values are different, the reference values for determining the presence or absence of adverse events that require discontinuation of treatment and incapable of resuming administration are 3,500 pg/ml, 4,000 pg/ml, 4,500 pg/ml. It may be selected from ml, 5,000 pg/ml, 5,500 pg/ml, 6,000 pg/ml, or 6,500 pg/ml. When the reference value is different, the reference value for determining the presence or absence of the therapeutic effect is 2,000 pg/ml, 2,500 pg/ml, 3,000 pg/ml, 3,500 pg/ml, 4,000 pg/ml, 4 , 500 pg/ml, 5,000 pg/ml, 5,500 pg/ml, 6,000 pg/ml, or 6,500 pg/ml.

 腫瘍細胞に対する細胞傷害活性を反映する値には、例えば、腫瘍細胞傷害活性の値、又はインターフェロン-γの測定値等が含まれ得る。 The value that reflects the cytotoxic activity on tumor cells may include, for example, the value of tumor cytotoxic activity or the measured value of interferon-γ.

 例えば、腫瘍細胞傷害活性の値は、前記PBMCと接触後の腫瘍細胞の生細胞数を測定し、その測定結果から腫瘍細胞傷害活性を算出することにより求めることができる。腫瘍細胞の生細胞数の測定方法は、PBMCがどのくらい腫瘍細胞を傷害できたかを評価できる限り制限されない。例えば、PBMCと腫瘍細胞とを直接接触させる場合には、PBMCと腫瘍細胞とを接触させた直後の腫瘍細胞の生細胞数とPBMCと腫瘍細胞とを接触させてから48時間後の腫瘍細胞の生細胞数の値から求めることができる。また、PBMCと腫瘍細胞とをエンゲージャーを介して、間接的に接触させる場合には、PBMCと腫瘍細胞とを接触させる際にエンゲージャーを添加するウェル(測定ウェル)と、PBMCと腫瘍細胞とを接触させる際にエンゲージャーを添加しないウェル(陰性対照ウェル)を作成し、測定ウェルの生細胞数と、陰性対照ウェルの生細胞数とに基づいて、腫瘍細胞傷害活性の値を取得することができる。この場合、例えば、腫瘍細胞傷害活性の値は、生細胞に基づく腫瘍細胞傷害活性パーセント(%)として、下式により求めることができる。 For example, the value of the tumor cytotoxic activity can be determined by measuring the number of viable tumor cells after contact with the PBMC and calculating the tumor cytotoxic activity from the measurement result. The method for measuring the viable cell number of tumor cells is not limited as long as it can evaluate how much PBMC can injure the tumor cells. For example, in the case of directly contacting PBMC with tumor cells, the viable cell number of the tumor cells immediately after contacting PBMC with the tumor cells and the tumor cell 48 hours after contacting PBMC with the tumor cells It can be determined from the value of the number of viable cells. Further, when PBMC and tumor cells are contacted indirectly via an engager, a well (measurement well) to which the engager is added when contacting PBMC and tumor cells, and PBMC and tumor cells To create a well (negative control well) to which no engager is added when contacting, and obtain the value of tumor cytotoxic activity based on the number of viable cells in the measurement well and the number of viable cells in the negative control well You can In this case, for example, the value of the tumor cytotoxic activity can be determined by the following formula as a percentage (%) of the tumor cytotoxic activity based on living cells.

[数1]
生細胞に基づく腫瘍細胞傷害活性パーセント(%)
={([陰性対照ウェルの生細胞数]-[測定ウェルの生細胞数])/[陰性対照ウェルの生細胞数]}×100
[Equation 1]
Tumor cytotoxic activity percentage based on live cells (%)
={([live cell number in negative control well]-[live cell number in measurement well])/[live cell number in negative control well]}×100

 また、例えばCellTiter 96(登録商標) AQueous One Solution Reagent (MTS reagent:プロメガ)等の生細胞染色試薬を用いて上記生細胞数を計数する場合には、上記生細胞数に変えて、各ウェルの吸光度に基づいて生細胞に基づく腫瘍細胞傷害活性パーセント(%)を算出してもよい。 When counting the number of viable cells using a viable cell staining reagent such as CellTiter96 (registered trademark) AQueousOne Solution Reagent (MTS reagent: Promega), change the viable cell number to The percentage of tumor cell cytotoxic activity based on live cells may be calculated based on the absorbance.

 さらに、上記生細胞数に変えて死細胞に基づいて、腫瘍細胞傷害活性パーセント(%)を算出し、この値を腫瘍細胞傷害活性の値として用いてもよい。PBMCと腫瘍細胞とをエンゲージャーを介して、間接的に接触させる場合において、死細胞に基づいて、腫瘍細胞傷害活性パーセント(%)を求める場合には、例えば以下の方法により、死細胞に基づく腫瘍細胞傷害活性パーセント(%)を算出することができる。具体的には、PBMCと腫瘍細胞とを接触させる際にエンゲージャーを添加するウェル(測定ウェル)と、PBMCと腫瘍細胞とを接触させる際にエンゲージャーを添加しないウェル(陰性対照ウェル)と、PBMCと腫瘍細胞とを接触させる際に過酸化水素等の細胞殺傷能力の高い物質を添加したウェル(陽性対照ウェル)を作成し、測定ウェルの死細胞数と、陰性対照ウェルの死細胞数と、陽性対照ウェルの死細胞数に基づいて、腫瘍細胞傷害活性の値を取得することができる。この場合、腫瘍細胞傷害活性の値は、死細胞に基づく腫瘍細胞傷害活性パーセント(%)として、下式で求めることができる。 Furthermore, the percentage of tumor cytotoxicity (%) may be calculated based on dead cells in place of the above-mentioned number of viable cells, and this value may be used as the value of tumor cytotoxicity. In the case of indirectly contacting PBMC with tumor cells via an engager, when determining the tumor cytotoxic activity percentage (%) based on dead cells, for example, the method described below The percent tumor cytotoxic activity can be calculated. Specifically, a well to which an engager is added when making contact with PBMC and tumor cells (measurement well), and a well to which no engager is added when making contact with PBMC and tumor cells (negative control well), A well (positive control well) to which a substance having a high cell-killing ability such as hydrogen peroxide was added when PBMCs were brought into contact with tumor cells was prepared, and the number of dead cells in the measurement well and the number of dead cells in the negative control well were measured. The value of tumor cytotoxic activity can be obtained based on the number of dead cells in the positive control wells. In this case, the value of the tumor cytotoxic activity can be calculated by the following formula as a percentage (%) of the tumor cytotoxic activity based on dead cells.

[数2]
死細胞に基づく腫瘍細胞傷害活性パーセント(%)
={([測定ウェルの死細胞数]-[陰性対照ウェルの死生細胞数])/[陽性対照ウェルの死生細胞数]-[陰性対照ウェルの死細胞数]}×100
[Equation 2]
Tumor cytotoxic activity based on dead cells (%)
={([number of dead cells in measurement well]-[number of dead cells in negative control well])/[number of dead cells in positive control well]-[number of dead cells in negative control well]} x 100

 例えばCytotoxity LDH assay kit(株式会社同仁化学研究所)等の死細胞測定キットを用いて上記死細胞数を計数する場合には、上記死細胞数に変えて、各ウェルの吸光度に基づいて死細胞に基づく腫瘍細胞傷害活性パーセント(%)を算出してもよい。 For example, when counting the number of dead cells using a dead cell measurement kit such as Cytotoxity LDH assay kit (Dojindo Laboratories Co., Ltd.), change to the number of dead cells and use the dead cells based on the absorbance of each well. The percent (%) of tumor cytotoxic activity based on

 インターフェロン-γの測定値は、例えば、PBMCと腫瘍細胞を接触させた後のウェル内から培養培地を採取し、その培養培地内のインターフェロン-γのタンパク質濃度(あるいは量)を公知のELISA法等を使用して測定することにより取得することができる。 The measurement value of interferon-γ can be obtained by, for example, collecting a culture medium from the well after contacting PBMC with tumor cells, and measuring the protein concentration (or amount) of interferon-γ in the culture medium by a known ELISA method or the like. Can be obtained by measuring using.

 本実施形態では、予め抗原捕捉用の抗インターフェロン-γ抗体をマイクロプレート等の固相上に固定化し、固定化された抗インターフェロン-γ抗体と検体中のインターフェロン-γの複合体を形成させることができる。固相上に固定化された当該複合体又は固相上で形成された複合体を、当該技術において公知の方法で検出することにより、検体に含まれるインターフェロン-γの量又は濃度を測定することができる。 In the present embodiment, an anti-interferon-γ antibody for capturing an antigen is previously immobilized on a solid phase such as a microplate to form a complex of the immobilized anti-interferon-γ antibody and interferon-γ in a sample. You can To measure the amount or concentration of interferon-γ contained in a sample by detecting the complex immobilized on the solid phase or the complex formed on the solid phase by a method known in the art. You can

 抗原捕捉用の抗インターフェロン-γ抗体の固相への固定の方法は、特に限定されない。公知の方法を使用して、直接的に、又は別の物質を介して間接的に行うことができる。直接の結合としては、例えば、物理的吸着などが挙げられる。好ましくは、イムノプレート等を使用して、直接抗インターフェロン-γ抗体をマイクロプレートに物理的に結合させることができる。 The method of immobilizing the anti-interferon-γ antibody for capturing the antigen on the solid phase is not particularly limited. It can be done directly using known methods or indirectly through another substance. Examples of the direct bond include physical adsorption. Preferably, the anti-interferon-γ antibody can be physically bound directly to the microplate using an immunoplate or the like.

 固相の素材は特に限定されず、例えば、ポリスチレン、ポリプロピレンなどが挙げられる。固相の形状は特に限定されず、例えば、マイクロプレート、マイクロチューブ、試験管、ビーズなどが挙げられる。 The solid phase material is not particularly limited, and examples thereof include polystyrene and polypropylene. The shape of the solid phase is not particularly limited, and examples thereof include a microplate, a microtube, a test tube, and beads.

 本方法においては、前記複合体の形成に続いて、固相を洗浄する操作を含んでもよい。洗浄する場合には、界面活性剤等を含むPBS等を使用することができる。 The method may include an operation of washing the solid phase subsequent to the formation of the complex. For washing, PBS containing a surfactant or the like can be used.

 本方法において、前記複合体の検出は、標識物質で標識された検出用抗インターフェロン-γ抗体を使用して行うか、未標識の抗インターフェロン-γ抗体と当該未標識の抗インターフェロン-γ抗体と結合することができる標識物質で標識された抗イムノグロブリン抗体等を使用して行うことができるが、標識された検出用抗インターフェロン-γ抗体を使用することが好ましい。また、検出用抗インターフェロン-γ抗体のインターフェロン-γにおけるエピトープと抗原捕捉用抗インターフェロン-γ抗体のインターフェロン-γにおけるエピトープとは異なることが好ましい。 In this method, the detection of the complex is carried out using an anti-interferon-γ antibody for detection labeled with a labeling substance, or an unlabeled anti-interferon-γ antibody and the unlabeled anti-interferon-γ antibody are used. It can be carried out using an anti-immunoglobulin antibody labeled with a labeling substance capable of binding, but it is preferable to use a labeled anti-interferon-γ antibody for detection. Further, it is preferable that the epitope of interferon-γ of the anti-interferon-γ antibody for detection is different from the epitope of interferon-γ of the anti-interferon-γ antibody for antigen capture.

 検出用抗インターフェロン-γ抗体又は標識抗イムノグロブリン抗体に用いられる標識物質は、検出可能なシグナルが生じる限り、特に限定されない。例えば、蛍光物質、放射性同位元素、及び酵素等が挙げられる。酵素としては、アルカリホスファターゼ、ペルオキシダーゼ等が挙げられる。蛍光物質としては、フルオレセインイソチオシアネート(FITC)、ローダミン、Alexa Fluor(登録商標)などの蛍光色素、GFPなどの蛍光タンパク質などが挙げられる。放射性同位元素としては、125I、14C、32Pなどが挙げられる。それらの中でも、標識物質として、アルカリホスファターゼ、又はペルオキシダーゼが好ましい。 The labeling substance used for the anti-interferon-γ antibody for detection or the labeled anti-immunoglobulin antibody is not particularly limited as long as it produces a detectable signal. For example, fluorescent substances, radioisotopes, enzymes and the like can be mentioned. Examples of the enzyme include alkaline phosphatase and peroxidase. Examples of the fluorescent substance include fluorescent dyes such as fluorescein isothiocyanate (FITC), rhodamine, and Alexa Fluor (registered trademark), and fluorescent proteins such as GFP. Examples of the radioisotope include 125 I, 14 C, 32 P and the like. Among them, alkaline phosphatase or peroxidase is preferable as the labeling substance.

 検出用抗インターフェロン-γ抗体は、当該技術において公知の標識方法により、抗インターフェロン-γ抗体を上記の標識物質で標識して得られる。また、市販のラベリングキットなどを用いて標識してもよい。また、標識イムノグロブリン抗体は、抗インターフェロン-γ抗体の標識と同じ手法を用いてもよいし、市販のものを使用してもよい。 The anti-interferon-γ antibody for detection can be obtained by labeling the anti-interferon-γ antibody with the above-mentioned labeling substance by a labeling method known in the art. Alternatively, labeling may be performed using a commercially available labeling kit or the like. The labeled immunoglobulin antibody may be the same as that used for labeling the anti-interferon-γ antibody, or may be a commercially available one.

 本方法では、複合体に含まれる標識抗インターフェロン-γ抗体の標識物質により生じるシグナルを検出することにより、検体に含まれるインターフェロン-γの測定値を取得できる。ここで、「シグナルを検出する」とは、シグナルの有無を定性的に検出すること、シグナル強度を定量すること、及び、シグナルの強度を半定量的に検出することを含む。半定量的な検出とは、シグナルの強度を、「シグナル発生せず」、「弱」、「中」、「強」などのように段階的に示すことをいう。本工程では、シグナルの強度を定量的又は半定量的に検出することが好ましい。 In this method, the measured value of interferon-γ contained in the sample can be obtained by detecting the signal generated by the labeled substance of the labeled anti-interferon-γ antibody contained in the complex. Here, "detecting a signal" includes qualitatively detecting the presence or absence of a signal, quantifying the signal intensity, and semi-quantitatively detecting the signal intensity. Semi-quantitative detection means indicating the intensity of a signal stepwise such as "no signal is generated", "weak", "medium", "strong". In this step, it is preferable to detect the signal intensity quantitatively or semi-quantitatively.

 シグナルを検出する方法は、公知の方法を使用することができる。本方法では、上記の標識物質に由来するシグナルの種類に応じた測定方法を適宜選択することができる。例えば、標識物質が酵素である場合、該酵素に対する基質を反応させることによって発生する光、色などのシグナルを、ルミノメーター、分光光度計などの公知の装置を用いて測定することにより行うことができる。 As a method for detecting a signal, a known method can be used. In this method, a measuring method can be appropriately selected according to the type of signal derived from the above-mentioned labeling substance. For example, when the labeling substance is an enzyme, light generated by reacting a substrate for the enzyme, a signal such as color, can be measured by using a known device such as a luminometer and a spectrophotometer. it can.

 酵素の基質は、該酵素の種類に応じて公知の基質から適宜選択できる。例えば、酵素としてアルカリホスファターゼを用いる場合、基質としては、CDP-Star(登録商標)(4-クロロ-3-(メトキシスピロ[1, 2-ジオキセタン-3, 2’-(5’-クロロ)トリクシロ[3.3.1.13,7]デカン]-4-イル)フェニルリン酸2ナトリウム)等の化学発光基質、5-ブロモ-4-クロロ-3-インドリルリン酸(BCIP)、5-ブロモ-6-クロロ-インドリルリン酸2ナトリウム、p-ニトロフェニルリン酸等の発色基質が挙げられる。標識物質がペルオキシダーゼである場合には、テトラメチルベンジジン(TMB)等を挙げることができる。 The substrate for the enzyme can be appropriately selected from known substrates according to the type of the enzyme. For example, when alkaline phosphatase is used as the enzyme, the substrate is CDP-Star (registered trademark) (4-chloro-3-(methoxyspiro[1,2-dioxetane-3,2'-(5'-chloro)tricloxyl A chemiluminescent substrate such as [3.3.1.13,7]decane]-4-yl)phenyl sodium phosphate), 5-bromo-4-chloro-3-indolyl phosphate (BCIP), 5- Color forming substrates such as disodium bromo-6-chloro-indolyl phosphate and p-nitrophenyl phosphate can be mentioned. When the labeling substance is peroxidase, tetramethylbenzidine (TMB) and the like can be mentioned.

 標識物質が放射性同位体である場合は、シグナルとしての放射線を、シンチレーションカウンターなどの公知の装置を用いて測定できる。また、標識物質が蛍光物質である場合は、シグナルとしての蛍光を、蛍光マイクロプレートリーダなどの公知の装置を用いて測定できる。なお、励起波長及び蛍光波長は、用いた蛍光物質の種類に応じて適宜決定できる。 When the labeled substance is a radioisotope, radiation as a signal can be measured using a known device such as a scintillation counter. When the labeling substance is a fluorescent substance, the fluorescence as a signal can be measured using a known device such as a fluorescence microplate reader. The excitation wavelength and the fluorescence wavelength can be appropriately determined according to the type of fluorescent substance used.

 シグナルの検出結果は、インターフェロン-γの測定値として用いることができる。例えば、シグナルの強度を定量的に検出する場合は、シグナル強度の測定値自体又は該シグナル強度の測定値から算出される値を、インターフェロン-γのタンパク質の測定値として用いることができる。 The signal detection result can be used as a measurement value for interferon-γ. For example, when the signal intensity is quantitatively detected, the signal intensity measurement value itself or a value calculated from the signal intensity measurement value can be used as the interferon-γ protein measurement value.

 特に言及しない限り、本項における用語の説明は、本明細書における他の実施形態における説明、特許請求の範囲及び図面全体において援用される。
 また、本項に記載される実施形態の別形態として、
Unless otherwise noted, the terms used in this section are incorporated by reference in the description of other embodiments herein, in the claims and throughout the drawings.
In addition, as another form of the embodiment described in this section,

2.悪性腫瘍を治療するための免疫療法剤の有用性の提示方法
 本明細書に開示されるある実施形態は、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法に関する。本実施形態は、ヒトによって実施されても、後述する提示装置10において実施されてもよい。
2. Methods of Presenting Utility of Immunotherapeutic Agents for Treating Malignant Tumors Certain embodiments disclosed herein demonstrate the utility of immunotherapeutic agents for treating malignant tumors in patients with said malignant tumors. Regarding how to present. The present embodiment may be performed by a human or may be performed by the presentation device 10 described later.

 前記提示方法は、前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程(工程1)と、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程(工程2)を含みうる。 The presenting method comprises a step (step 1) of assessing whether tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured (step 1); In the step of presenting that the immunotherapeutic agent is useful to the patient when it is shown that the tumor cells are injured, and/or the tumor cells are not injured in the evaluation. If indicated, the step of presenting to the patient that the immunotherapeutic agent is not useful may be included (step 2).

 工程1は、上記1.で述べた通りである。 Step 1 is the above 1. Is as described in.

 工程2において、「提示する」とはヒトが把握できるように示すことを意図する。 ”In step 2, “present” is intended to be shown so that a person can understand it.

 前記有用性が、前記免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記工程2は、より具体的には、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、を含む。前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示することには、例えば、紙媒体又は表示画面に、前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを示す情報(例えば「なし」、「陰性」、「-(マイナス)」等)を記載又は表示することを含む。前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示することには、例えば、紙媒体又は表示画面に、前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを示す情報(例えば「あり」、「陽性」、「+(プラス)」等)を記載又は表示することを含む。 When the usefulness is the presence or absence of an adverse event that requires treatment interruption due to the immunotherapeutic agent and is incapable of resuming administration, the step 2 more specifically includes the evaluation. In the case where the tumor cells are shown to be injured in, the immunotherapeutic agent does not cause an adverse event in the patient, which requires treatment interruption and makes it impossible to resume administration. If the step of presenting and/or the evaluation shows that the tumor cells are not injured, the immunotherapeutic agent is required to be interrupted to the patient and the administration is not restarted. Presenting that a possible adverse event will occur. To present that the treatment needs to be interrupted and does not cause an adverse event that makes it impossible to resume the administration, for example, on a paper medium or a display screen, the treatment must be interrupted and the administration can be restarted. This includes describing or displaying information (for example, "none", "negative", "-(minus)", etc.) indicating that no adverse adverse events will occur. To present that an adverse event that requires the treatment interruption and makes it impossible to resume the administration, for example, on a paper medium or a display screen, the treatment interruption is required and the administration restart is not possible. This includes describing or displaying information indicating that a possible adverse event will occur (for example, “present”, “positive”, “+ (plus)”, etc.).

 また、前記有用性が、前記免疫療法剤の効果の有無であるとき、前記工程2は、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、を含む。前記免疫療法剤が有効であることを提示することには、例えば、紙媒体又は表示画面に、前記免疫療法剤が有効であることを示す情報(例えば「有効」、「〇」、「+(プラス)」等)を記載又は表示することを含む。前記免疫療法剤が無効であることを提示することには、例えば、紙媒体又は表示画面に、前記免疫療法剤が無効であることを示す情報(例えば「無効」、「×」、「-(マイナス)」等)を記載又は表示することを含む。 Further, when the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the step 2 comprises the step of immunizing the patient when the tumor cells are shown to be injured in the evaluation. Indicating that the immunotherapeutic agent is ineffective to the patient when presenting that the therapeutic agent is effective and/or when the evaluation indicates that the tumor cells are not injured. And presenting. In order to present that the immunotherapeutic agent is effective, for example, information indicating that the immunotherapeutic agent is effective is displayed on a paper medium or a display screen (for example, “effective”, “◯”, “+( Plus)” and the like). To indicate that the immunotherapeutic agent is invalid, for example, information indicating that the immunotherapeutic agent is invalid is displayed on a paper medium or a display screen (for example, "invalid", "x", "-( Minus)” etc.) is included or displayed.

 上記1.で説明された、用語であって、本項においても使用される用語については、上記1.の説明をここに援用する。 Above 1. The terms explained in 1. and also used in this section are described in 1. above. Is incorporated herein by reference.

3.悪性腫瘍を治療するための免疫療法剤の有用性の決定補助方法
 本明細書に開示されるある実施形態は、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助する方法に関する。有用性を補助することとは、医師が有用性を決定することを補助することを含む。本実施形態は、ヒトによって実施されても、後述する補助装置10において実施されてもよい。
 前記補助方法は、前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程(工程i)と、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程(工程ii)を含みうる。
3. Auxiliary Methods for Determining Utility of Immunotherapeutic Agents for Treating Malignant Tumors Certain embodiments disclosed herein provide immunotherapeutic agents for treating malignant tumors useful in patients with said malignant tumors. To assist in the decision of. Aiding utility includes assisting a physician in determining utility. The present embodiment may be carried out by a human or may be carried out in the auxiliary device 10 described later.
The assisting method comprises a step (step i) of assessing whether tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured, and the assessment. In the step of presenting that the immunotherapeutic agent is useful to the patient when it is shown that the tumor cells are injured, and/or the tumor cells are not injured in the evaluation. If indicated, the step of presenting to the patient that the immunotherapeutic agent is not useful may be included (step ii).

 工程iは、上記1.で述べた通りである。 Step i is 1. Is as described in.

 工程iiにおいて、「提示する」とはヒトが把握できるように示すことを意図する。 ”In step ii, “presenting” is intended to be shown so that a person can understand it.

 前記有用性が、前記免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記工程iiは、より具体的には、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、を含む。前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示することには、例えば、紙媒体又は表示画面に、前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを示す情報(例えば「なし」、「陰性」、「-(マイナス)」等)を記載又は表示することを含む。前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示することには、例えば、紙媒体又は表示画面に、前記治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを示す情報(例えば「あり」、「陽性」、「+(プラス)」等)を記載又は表示することを含む。 When the usefulness is the presence or absence of an adverse event resulting from the immunotherapeutic agent requiring treatment interruption and incapable of resuming administration, the step ii, more specifically, the evaluation. In the case where the tumor cells are shown to be injured in, the immunotherapeutic agent does not cause an adverse event in the patient, which requires treatment interruption and makes it impossible to resume administration. If the step of presenting and/or the evaluation shows that the tumor cells are not injured, the immunotherapeutic agent is required to be interrupted to the patient and the administration is not restarted. Presenting that a possible adverse event will occur. To present that the treatment needs to be interrupted and does not cause an adverse event that makes it impossible to resume the administration, for example, on a paper medium or a display screen, the treatment must be interrupted and the administration can be restarted. This includes describing or displaying information (for example, "none", "negative", "-(minus)", etc.) indicating that no adverse adverse events will occur. To present that an adverse event that requires the treatment interruption and makes it impossible to resume the administration, for example, on a paper medium or a display screen, the treatment interruption is required and the administration restart is not possible. This includes describing or displaying information indicating that a possible adverse event will occur (for example, “present”, “positive”, “+ (plus)”, etc.).

 また、前記有用性が、前記免疫療法剤の効果の有無であるとき、前記工程iiは、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、を含む。前記免疫療法剤が有効であることを提示することには、例えば、紙媒体又は表示画面に、前記免疫療法剤が有効であることを示す情報(例えば「有効」、「〇」、「+(プラス)」等)を記載又は表示することを含む。前記免疫療法剤が無効であることを提示することには、例えば、紙媒体又は表示画面に、前記免疫療法剤が無効であることを示す情報(例えば「無効」、「×」、「-(マイナス)」等)を記載又は表示することを含む。 Further, when the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the step ii includes the step of immunizing the patient when the tumor cells are shown to be injured. Indicating that the immunotherapeutic agent is ineffective to the patient when presenting that the therapeutic agent is effective and/or when the evaluation indicates that the tumor cells are not injured. And presenting. In order to present that the immunotherapeutic agent is effective, for example, information indicating that the immunotherapeutic agent is effective is displayed on a paper medium or a display screen (for example, “effective”, “◯”, “+( Plus)” and the like). To indicate that the immunotherapeutic agent is invalid, for example, information indicating that the immunotherapeutic agent is invalid is displayed on a paper medium or a display screen (for example, "invalid", "x", "-( Minus)” etc.) is included or displayed.

 上記1.で説明された、用語であって、本項においても使用される用語については、上記1.の説明をここに援用する。 Above 1. The terms explained in 1. and also used in this section are described in 1. above. Is incorporated herein by reference.

4.悪性腫瘍を治療するための免疫療法剤の有用性の提示装置
 本明細書に開示されるある実施形態は、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する装置10(以下、提示装置10ともいう)に関する。
4. Device for Presenting Utility of Immunotherapeutic Agents for Treating Malignant Tumors Certain embodiments disclosed herein provide immunotherapeutic agents for treating malignant tumors in a patient having the malignant tumor. The present invention relates to a presenting device 10 (hereinafter, also referred to as a presenting device 10).

 図2及び図3に、提示装置10のハードウェアの構成例を示す。提示装置10は、入力部111と、出力部112と、記憶媒体113とに接続されていてもよい。また、マイクロプレートリーダ等により構成される測定部30と接続されていてもよい。すなわち、提示装置10は、測定部30と直接又はネットワーク等を介して接続された、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する提示システム50を構成することもある。 2 and 3 show an example of the hardware configuration of the presentation device 10. The presentation device 10 may be connected to the input unit 111, the output unit 112, and the storage medium 113. Further, it may be connected to the measurement unit 30 configured by a microplate reader or the like. That is, the presentation device 10 includes a presentation system 50 that is connected to the measurement unit 30 directly or via a network or the like, and presents the utility of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor. It may be configured.

 提示装置10において、処理部(CPU)101と、主記憶部102と、ROM(read only memory)103と、補助記憶部104と、通信インタフェース(I/F)105と、入力インタフェース(I/F)106と、出力インタフェース(I/F)107と、メディアインターフェース(I/F)108は、バス109によって互いにデータ通信可能に接続されている。主記憶部102と補助記憶部104とを合わせて、単に記憶部と呼ぶこともある。 In the presentation device 10, a processing unit (CPU) 101, a main storage unit 102, a ROM (read only memory) 103, an auxiliary storage unit 104, a communication interface (I/F) 105, and an input interface (I/F). ) 106, an output interface (I/F) 107, and a media interface (I/F) 108 are connected to each other by a bus 109 so that data communication is possible. The main storage unit 102 and the auxiliary storage unit 104 may be collectively referred to as a storage unit.

 CPU101は、提示装置10の処理部101である。CPU101が、補助記憶部104又はROM103に記憶されているコンピュータプログラムを実行し、取得されるデータの処理を行うことにより、提示装置10が機能する。 The CPU 101 is the processing unit 101 of the presentation device 10. The CPU 101 executes the computer program stored in the auxiliary storage unit 104 or the ROM 103 and processes the acquired data, so that the presentation device 10 functions.

 ROM103は、マスクROM、PROM、EPROM、EEPROMなどによって構成され、CPU101により実行されるコンピュータプログラム及びこれに用いるデータが記録されている。CPU101はMPU101としてもよい。ROM103は、提示装置10の起動時に、CPU101によって実行されるブートプログラムや提示装置10のハードウェアの動作に関連するプログラムや設定を記憶する。 The ROM 103 is composed of a mask ROM, a PROM, an EPROM, an EEPROM, etc., and stores a computer program executed by the CPU 101 and data used for the computer program. The CPU 101 may be the MPU 101. The ROM 103 stores a boot program executed by the CPU 101 when the presentation device 10 is activated and programs and settings related to the operation of the hardware of the presentation device 10.

 主記憶部102は、SRAM又はDRAMなどのRAM(Random access memory)によって構成される。主記憶部102は、ROM103及び補助記憶部104に記録されているコンピュータプログラムの読み出しに用いられる。また、主記憶部102は、CPU101がこれらのコンピュータプログラムを実行するときの作業領域として利用される。さらに主記憶部102は、処理部101が取得した腫瘍細胞が傷害されているか否かの評価結果を一時的に記憶してもよい。さらに、基準値がネットワークから又は補助記憶部104から提供される場合、これらを一時的に記憶してもよい。 The main storage unit 102 is configured by a RAM (Random access memory) such as SRAM or DRAM. The main storage unit 102 is used to read the computer programs recorded in the ROM 103 and the auxiliary storage unit 104. Further, the main storage unit 102 is used as a work area when the CPU 101 executes these computer programs. Furthermore, the main storage unit 102 may temporarily store the evaluation result obtained by the processing unit 101, which indicates whether or not the tumor cells are injured. Further, when the reference values are provided from the network or the auxiliary storage unit 104, these may be temporarily stored.

 補助記憶部104は、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等によって構成される。補助記憶部104には、オペレーティングシステム及びアプリケーションプログラムなどの、CPU101に実行させるための種々のコンピュータプログラム及びコンピュータプログラムの実行に用いる各種設定データが記憶されている。具体的には、基準値等を不揮発性に記憶していてもよい。 The auxiliary storage unit 104 includes a hard disk, a semiconductor memory device such as a flash memory, an optical disk, and the like. The auxiliary storage unit 104 stores various computer programs to be executed by the CPU 101, such as an operating system and application programs, and various setting data used for executing the computer programs. Specifically, the reference value and the like may be stored in a non-volatile manner.

 通信I/F105は、USB、IEEE1394、RS-232Cなどのシリアルインタフェース、SCSI、IDE、IEEE1284などのパラレルインタフェース、及びD/A変換器、A/D変換器などからなるアナログインタフェース、ネットワークインタフェースコントローラ(Network interface controller:NIC)等から構成される。通信I/F105は、CPU101の制御下で、測定部30又は他の外部機器からのデータを受信し、必要に応じて提示装置10が保存又は生成する情報を、測定部30又は外部に送信又は表示する。通信I/F105は、ネットワークを介して測定部30又は他の外部機器と通信を行ってもよい。また、基準値がネットワークから提供される場合には、通信I/F105は、CPU101の制御下で、基準値を受信する。 The communication I/F 105 includes a serial interface such as USB, IEEE1394, RS-232C, a parallel interface such as SCSI, IDE, and IEEE1284, an analog interface including a D/A converter and an A/D converter, a network interface controller ( Network interface controller (NIC), etc. Under the control of the CPU 101, the communication I/F 105 receives data from the measurement unit 30 or another external device, and transmits information stored or generated by the presentation device 10 to the measurement unit 30 or the outside as necessary. indicate. The communication I/F 105 may communicate with the measurement unit 30 or another external device via a network. When the reference value is provided from the network, the communication I/F 105 receives the reference value under the control of the CPU 101.

 入力I/F106は、例えばUSB、IEEE1394、RS-232Cなどのシリアルインタフェース、SCSI、IDE、IEEE1284などのパラレルインタフェース、及びD/A変換器、A/D変換器などからなるアナログインタフェースなどから構成される。入力I/F106は、入力部111から文字入力、クリック、音声入力等を受け付ける。受け付けた入力内容は、主記憶部102又は補助記憶部104に記憶される。 The input I/F 106 is composed of, for example, a serial interface such as USB, IEEE1394, RS-232C, a parallel interface such as SCSI, IDE, IEEE1284, and an analog interface including a D/A converter and an A/D converter. It The input I/F 106 receives character input, click, voice input, and the like from the input unit 111. The received input content is stored in the main storage unit 102 or the auxiliary storage unit 104.

 入力部111は、タッチパネル、キーボード、マウス、ペンタブレット、マイク等から構成され、提示装置10に文字入力又は音声入力を行う。入力部111は、提示装置10の外部から接続されても、提示装置10と一体となっていてもよい。 The input unit 111 includes a touch panel, a keyboard, a mouse, a pen tablet, a microphone, and the like, and performs character input or voice input to the presentation device 10. The input unit 111 may be connected from the outside of the presentation device 10 or integrated with the presentation device 10.

 出力I/F107は、例えば入力I/F106と同様のインタフェースから構成される。出力I/F107は、CPU101が生成した情報を出力部112に出力する。出力I/F107は、CPU101が生成し、補助記憶部104に記憶した情報を、出力部112に出力する。 The output I/F 107 is composed of the same interface as the input I/F 106, for example. The output I/F 107 outputs the information generated by the CPU 101 to the output unit 112. The output I/F 107 outputs the information generated by the CPU 101 and stored in the auxiliary storage unit 104 to the output unit 112.

 出力部112は、例えばディスプレイ、プリンター等で構成され、測定部30から送信される測定結果及び提示装置10における各種操作ウインドウ、腫瘍細胞が傷害されているか否かを評価した評価、有用性の提示内容等を表示する。 The output unit 112 is composed of, for example, a display, a printer, and the like, and evaluates the measurement results transmitted from the measurement unit 30, various operation windows in the presentation device 10, and whether or not tumor cells are injured, and presentation of usefulness. Display contents etc.

 メディアI/F108は、記憶媒体113に記憶された例えばアプリケーションソフト等を読み出す。読み出されたアプリケーションソフト等は、主記憶部102又は補助記憶部104に記憶される。また、メディアI/F108は、CPU101が生成した情報を記憶媒体113に書き込む。メディアI/F108は、CPU101が生成し、補助記憶部104に記憶した情報を、記憶媒体113に書き込む。 The media I/F 108 reads out, for example, application software stored in the storage medium 113. The read application software and the like are stored in the main storage unit 102 or the auxiliary storage unit 104. The media I/F 108 also writes the information generated by the CPU 101 in the storage medium 113. The media I/F 108 writes the information generated by the CPU 101 and stored in the auxiliary storage unit 104 into the storage medium 113.

 記憶媒体113は、フレキシブルディスク、CD-ROM、又はDVD-ROM等で構成される。記憶媒体113は、フレキシブルディスクドライブ、CD-ROMドライブ、又はDVD-ROMドライブ等によってメディアI/F108と接続される。記憶媒体113には、コンピュータがオペレーションを実行するためのアプリケーションプログラム等が格納されていてもよい。 The storage medium 113 is composed of a flexible disk, a CD-ROM, a DVD-ROM, or the like. The storage medium 113 is connected to the media I/F 108 by a flexible disk drive, a CD-ROM drive, a DVD-ROM drive, or the like. The storage medium 113 may store an application program or the like for a computer to execute an operation.

 CPU101は、提示装置10の制御に必要なアプリケーションソフトや各種設定をROM103又は補助記憶部104からの読み出しに代えて、ネットワークを介して取得してもよい。前記アプリケーションプログラムがネットワーク上のサーバコンピュータの補助記憶部内に格納されており、このサーバコンピュータに提示装置10がアクセスして、コンピュータプログラムをダウンロードし、これをROM103又は補助記憶部104に記憶することも可能である。 The CPU 101 may acquire application software and various settings necessary for controlling the presentation device 10 via the network instead of reading from the ROM 103 or the auxiliary storage unit 104. The application program is stored in the auxiliary storage unit of the server computer on the network, and the presentation device 10 accesses the server computer to download the computer program and store the computer program in the ROM 103 or the auxiliary storage unit 104. It is possible.

 また、ROM103又は補助記憶部104には、例えば米国マイクロソフト社が製造販売するWindows(登録商標)などのグラフィカルユーザインタフェース環境を提供するオペレーションシステムがインストールされている。第2の実施形態に係るアプリケーションプログラムは、前記オペレーティングシステム上で動作するものとする。すなわち、提示装置10は、パーソナルコンピュータ等であり得る。 Further, in the ROM 103 or the auxiliary storage unit 104, an operating system that provides a graphical user interface environment such as Windows (registered trademark) manufactured and sold by Microsoft Corporation in the United States is installed. The application program according to the second embodiment operates on the operating system. That is, the presentation device 10 may be a personal computer or the like.

 次に、図4を用いて、提示装置10の動作の概略について説明する。提示装置10の動作は、後述する悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示するためのコンピュータプログラムの指令にしたがって、提示装置10の処理部101が制御する。 Next, an outline of the operation of the presentation device 10 will be described with reference to FIG. The operation of the presentation device 10 is performed by the processing unit 101 of the presentation device 10 according to a command of a computer program for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor described below in a patient having the malignant tumor. Control.

 初めに、処理部101は、検査者によって入力部111から入力される、若しくは処理部101が検査者の制御によって、上記1.で述べた方法にしたがって患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価し、評価結果を取得する(ステップS11)。 First, the processing unit 101 is input by the inspector from the input unit 111, or the processing unit 101 is controlled by the inspector and the above 1. The peripheral blood mononuclear cells collected from the patient according to the method described in 1. above are evaluated as to whether or not the tumor cells contacted directly or indirectly in vitro are injured, and the evaluation result is obtained (step S11). ).

 次に処理部101は、上記1.で述べた方法にしたがって前記評価結果が、腫瘍細胞が傷害されていることを示しているか否かを決定する(ステップ12)。 Next, the processing unit 101 uses the above 1. It is determined whether the evaluation result indicates that the tumor cells are damaged according to the method described in (step 12).

 ステップ12において、評価結果が、腫瘍細胞が傷害されていることを示している場合(YES)には、処理部101は、ステップS13に進み、免疫療法剤が有用であると決定し、その結果を出力部112から出力することにより提示する。ステップS12において、評価結果が、腫瘍細胞が傷害されていないことを示している場合(NO)には、処理部101は、ステップS14に進み、免疫療法剤が有用でないと決定し、その結果を出力部112から出力することにより提示する。 In step 12, when the evaluation result indicates that the tumor cells are injured (YES), the processing unit 101 proceeds to step S13, determines that the immunotherapeutic agent is useful, and the result is Is presented from the output unit 112. In step S12, when the evaluation result indicates that the tumor cells are not injured (NO), the processing unit 101 proceeds to step S14, determines that the immunotherapeutic agent is not useful, and outputs the result. It is presented by outputting from the output unit 112.

 有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS12において、前記評価結果が、前記腫瘍細胞が傷害されていることを示す(YES)場合に、スッテプS13において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS12において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、スッテプS14において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する。 When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted and administration cannot be restarted, the processing unit 101 determines in Step S12 that the evaluation result is If it is indicated that the tumor cells are injured (YES), in step S13, the immunotherapeutic agent causes an adverse event for the patient, in which the treatment needs to be interrupted and the administration cannot be restarted. Show that you don't. When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted and administration cannot be restarted, the processing unit 101 determines in Step S12 that the evaluation result is In the case of (NO) indicating that the tumor cells have not been injured, in step S14, the immunotherapeutic agent causes an adverse event in the patient, which requires treatment interruption and cannot resume administration. Offer to wake up.

 有用性が、免疫療法剤の効果の有無であるとき、前記処理部101は、スッテプS12において、前記評価結果が、前記腫瘍細胞が傷害されていることを示す(YES)場合に、スッテプS13において、患者に対して前記免疫療法剤が有効であることを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS12において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、スッテプS14において、患者に対して前記免疫療法剤が無効であることを提示する。 When the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the processing unit 101, in step S12, in step S13, when the evaluation result indicates that the tumor cells are injured (YES). , Suggest that the immunotherapeutic agent is effective for patients. When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted and administration cannot be restarted, the processing unit 101 determines in Step S12 that the evaluation result is If the tumor cells are not injured (NO), step S14 is presented to the patient that the immunotherapeutic agent is ineffective.

 処理部101は、上記ステップS13、又はステップS14で得られた結果を記録媒体113に記録してもよい。 The processing unit 101 may record the result obtained in step S13 or step S14 in the recording medium 113.

 次に、図5を用いて、提示装置10の動作の別形態について説明する。初めに、処理部101は、検査者によって入力部111から入力される、上記1.で述べた方法により取得された腫瘍細胞傷害活性を反映する値を取得する(ステップS101)。又は、測定部30から腫瘍細胞傷害活性を算出するための情報を取得する。前記情報は、PBMCと接触した腫瘍細胞の生細胞数又は死細胞数を反映する情報であり、例えば生細胞染色試薬又は死細胞測定試薬の吸光度等である。あるいは、前記情報は、インターフェロン-γの測定した際の吸光度等である。 Next, another mode of the operation of the presentation device 10 will be described with reference to FIG. First, the processing unit 101 is input by the inspector from the input unit 111. A value that reflects the tumor cytotoxic activity obtained by the method described in (1) is obtained (step S101). Alternatively, the information for calculating the tumor cytotoxic activity is acquired from the measurement unit 30. The information is information that reflects the number of living cells or dead cells of the tumor cells that have come into contact with PBMC, and is, for example, the absorbance of a living cell staining reagent or a dead cell measurement reagent. Alternatively, the information is the absorbance when interferon-γ is measured.

 次に、処理部101は、ステップS101で取得した腫瘍細胞傷害活性を反映する値を主記憶部102又は補助記憶部104に記憶されている基準値と比較する(ステップS102)。 Next, the processing unit 101 compares the value reflecting the tumor cell cytotoxic activity acquired in step S101 with the reference value stored in the main storage unit 102 or the auxiliary storage unit 104 (step S102).

 続いて処理部101は、ステップS102の比較結果からステップS101で取得した腫瘍細胞傷害活性を反映する値が基準値よりも否かを決定する(ステップS103)。 Subsequently, the processing unit 101 determines whether the value reflecting the tumor cell cytotoxic activity acquired in step S101 is higher than the reference value based on the comparison result in step S102 (step S103).

 処理部101は、ステップS103において腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合には、ステップS104に進み、免疫療法剤が有用であると決定し、その結果を出力部112から出力することにより提示する。ステップS103において、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合には、処理部101は、ステップS105に進み、免疫療法剤が有用でないと決定し、その結果を出力部112から出力することにより提示する。 When the processing unit 101 determines in step S103 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES), the processing unit 101 proceeds to step S104, determines that the immunotherapeutic agent is useful, and Is presented from the output unit 112. When it is determined in step S103 that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), the processing unit 101 proceeds to step S105, determines that the immunotherapeutic agent is not useful, and Is presented from the output unit 112.

 有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS103において、腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合に、スッテプS104において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS103において、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合に、スッテプS105において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する。 When the usefulness is the presence or absence of the adverse event that requires the treatment interruption due to the immunotherapeutic agent and the administration cannot be resumed, the processing unit 101 determines the tumor cytotoxic activity in step S103. When it is determined that the value to be reflected is higher than the reference value (YES), in step S104, the immunotherapeutic agent is administered to the patient for an adverse event that requires treatment interruption and cannot be resumed. Show what you don't. When the usefulness is the presence or absence of the adverse event that requires the treatment interruption due to the immunotherapeutic agent and the administration cannot be resumed, the processing unit 101 determines the tumor cytotoxic activity in step S103. When it is determined that the reflected value is lower than the reference value (NO), in step S105, the immunotherapeutic agent causes an adverse event for the patient, which requires treatment interruption and resumption of administration is impossible. Offer to wake up.

 有用性が、免疫療法剤の効果の有無であるとき、前記処理部101は、スッテプS103において、腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合に、スッテプS104において、患者に対して前記免疫療法剤が有効であることを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部101は、スッテプS103において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合に、スッテプS105において、患者に対して前記免疫療法剤が無効であることを提示する。 When the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the processing unit 101 determines in Step S103 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES), Step S104. In, it is shown to the patient that the immunotherapeutic agent is effective. When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted, and administration cannot be restarted, the processing unit 101, in step S103, the evaluation result is In the case of indicating that the tumor cells are not injured (NO), when it is determined that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), in step S105, the Present that the immunotherapeutic is ineffective.

 処理部101は、上記ステップS104、又はステップS105で得られた結果を記録媒体113に記録してもよい。 The processing unit 101 may record the result obtained in step S104 or step S105 in the recording medium 113.

 上記1.及び2.における用語の説明、各工程の説明は、ここに援用される。 Above 1. And 2. The explanation of terms and explanation of each step are incorporated herein by reference.

5.悪性腫瘍を治療するための免疫療法剤の有用性の提示装置を制御するためのコンピュータプログラム及び前記コンピュータプログラムを記憶した記憶媒体
 本明細書に開示されるある実施形態は、コンピュータに実行させたときに、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示するためのコンピュータプログラムに関する。具体的には、上記4.で述べた悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する提示装置の動作を制御するコンピュータプログラムである。
5. A computer program for controlling a device for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor, and a storage medium storing the computer program according to an embodiment disclosed herein. In particular, the present invention relates to a computer program for presenting the utility of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor. Specifically, the above 4. Regarding the immunotherapeutic agent for treating the malignant tumor described in 1., it is a computer program for controlling the operation of the presentation device that presents the usefulness in the patient having the malignant tumor.

 本実施形態のコンピュータプログラムが実行する各ステップは、上記4.で述べたステップS11~14又はステップS101~105である。上記1.及び2.における用語の説明、各工程の説明は、ここに援用される。 The steps executed by the computer program of this embodiment are the same as those in 4. These are steps S11 to S14 or steps S101 to 105 described above. Above 1. And 2. The explanation of terms and explanation of each step are incorporated herein by reference.

 さらに、本明細書に開示されるある実施形態は、前記コンピュータプログラムを記憶した、記憶媒体に関する。すなわち、前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記憶媒体に記憶される。前記記憶媒体へのプログラムの記憶形式は、前記提示装置が前記プログラムを読み取り可能である限り制限されない。前記記憶媒体への記憶は、不揮発性であることが好ましい。 Furthermore, an embodiment disclosed in the present specification relates to a storage medium storing the computer program. That is, the computer program is stored in a storage medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program in the storage medium is not limited as long as the presentation device can read the program. The storage in the storage medium is preferably non-volatile.

6.悪性腫瘍を治療するための免疫療法剤の有用性の決定補助装置
 本明細書に開示されるある実施形態は、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を決定することを補助する装置20(以下、補助装置20ともいう)に関する。
6. Assistive Device for Determining Utility of Immunotherapeutic Agents for Treating Malignant Tumors Certain embodiments disclosed herein provide immunotherapeutic agents for treating malignant tumors useful in a patient having the malignant tumor. The device 20 (hereinafter, also referred to as the auxiliary device 20) that assists in determining

 補助装置20のハードウェアの構成例は、図2及び図3に示す提示装置10と同様である。補助装置20は、マイクロプレートリーダ等により構成される測定部30と接続されていてもよい。すなわち、補助装置20は、測定部30と直接又はネットワーク等を介して接続された、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助する補助システム50を構成することもある。 An example of the hardware configuration of the auxiliary device 20 is the same as that of the presentation device 10 shown in FIGS. 2 and 3. The auxiliary device 20 may be connected to the measurement unit 30 configured by a microplate reader or the like. That is, the auxiliary device 20 assists the determination of the usefulness of the immunotherapeutic agent for treating a malignant tumor, which is connected to the measurement unit 30 directly or via a network or the like, in a patient having the malignant tumor. 50 may be configured.

 補助装置20のハードウェアの構成の説明は、提示装置10の説明をここに援用する。ここでは、提示装置10の説明における処理部(CPU)101、主記憶部102、ROM103、補助記憶部104、通信インタフェース(I/F)105、入力インタフェース(I/F)106、出力インタフェース(I/F)107、メディアインターフェース(I/F)108、バス109、入力部111、出力部112、及び記憶媒体113は、補助装置20において、それぞれ、処理部(CPU)201、主記憶部202、ROM203、補助記憶部204、通信インタフェース(I/F)205、入力インタフェース(I/F)206、出力インタフェース(I/F)207、メディアインターフェース(I/F)208、バス209、入力部211、出力部212及び記憶媒体213と読み替えるものとする。 For the description of the hardware configuration of the auxiliary device 20, the description of the presentation device 10 is incorporated here. Here, the processing unit (CPU) 101, the main storage unit 102, the ROM 103, the auxiliary storage unit 104, the communication interface (I/F) 105, the input interface (I/F) 106, and the output interface (I) in the description of the presentation device 10 are described. /F) 107, media interface (I/F) 108, bus 109, input unit 111, output unit 112, and storage medium 113 in the auxiliary device 20, respectively, processing unit (CPU) 201, main storage unit 202, ROM 203, auxiliary storage unit 204, communication interface (I/F) 205, input interface (I/F) 206, output interface (I/F) 207, media interface (I/F) 208, bus 209, input unit 211, It should be read as the output unit 212 and the storage medium 213.

 次に、図6を用いて、補助装置20の動作の概略について説明する。補助装置20の動作は、後述する悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示するためのコンピュータプログラムの指令にしたがって、補助装置20の処理部201が制御する。 Next, the outline of the operation of the auxiliary device 20 will be described with reference to FIG. The operation of the assisting device 20 is performed by the processing unit 201 of the assisting device 20 according to a command of a computer program for presenting the usefulness of an immunotherapeutic agent for treating a malignant tumor described below in a patient having the malignant tumor. Control.

 初めに、処理部201は、検査者によって入力部211から入力される、若しくは処理部201が検査者の制御によって、上記1.で述べた方法にしたがって患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価し、評価結果を取得する(ステップS21)。 First, the processing unit 201 is input by the inspector from the input unit 211, or the processing unit 201 is controlled by the inspector and the above 1. The peripheral blood mononuclear cells collected from the patient are directly or indirectly contacted with the tumor cells according to the method described in 1. to evaluate whether the tumor cells are injured, and obtain the evaluation result (step S21). ).

 次に処理部201は、上記1.で述べた方法にしたがって前記評価結果が、腫瘍細胞が傷害されていることを示しているか否かを決定する(ステップ22)。 Next, the processing unit 201 uses the above 1. It is determined whether the evaluation result shows that the tumor cells are injured according to the method described in (22).

 ステップ22において、評価結果が、腫瘍細胞が傷害されていることを示している場合(YES)には、処理部201は、ステップS23に進み、免疫療法剤が有用であると決定し、その結果を出力部212から出力することにより提示する。ステップS22において、評価結果が、腫瘍細胞が傷害されていないことを示している場合(NO)には、処理部201は、ステップS24に進み、免疫療法剤が有用でないと決定し、その結果を出力部212から出力することにより提示する。 In step 22, if the evaluation result indicates that the tumor cells are injured (YES), the processing unit 201 proceeds to step S23, determines that the immunotherapeutic agent is useful, and the result is To be presented by outputting from the output unit 212. In step S22, when the evaluation result indicates that the tumor cells are not injured (NO), the processing unit 201 proceeds to step S24, determines that the immunotherapeutic agent is not useful, and outputs the result. It is presented by outputting from the output unit 212.

 有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS22において、前記評価結果が、前記腫瘍細胞が傷害されていることを示す(YES)場合に、スッテプS23において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないと決定する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS22において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、スッテプS24において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすと決定する。 When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted and administration cannot be restarted, the processing unit 201, in step S22, the evaluation result is If it is indicated that the tumor cells are injured (YES), in step S23, the immunotherapeutic agent causes an adverse event to the patient in which treatment must be interrupted and administration cannot be restarted. Decide not to. When the usefulness is the presence or absence of an adverse event in which treatment due to an immunotherapeutic agent needs to be interrupted and administration cannot be restarted, the processing unit 201, in step S22, the evaluation result is In the case of (NO) indicating that the tumor cells are not injured, in step S24, the immunotherapeutic agent causes an adverse event in the patient, which requires treatment interruption and cannot resume administration. Decide to wake up.

 有用性が、免疫療法剤の効果の有無であるとき、前記処理部201は、スッテプS22において、前記評価結果が、前記腫瘍細胞が傷害されていることを示す(YES)場合に、スッテプS23において、患者に対して前記免疫療法剤が有効であると決定する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS12において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、スッテプS24において、患者に対して前記免疫療法剤が無効であると決定する。 When the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the processing unit 201, in step S22, if the evaluation result indicates that the tumor cells are injured (YES), in step S23. , Determine that the immunotherapeutic agent is effective for the patient. When the usefulness is the presence or absence of an adverse event in which the treatment due to the immunotherapeutic agent needs to be interrupted and the administration cannot be restarted, the processing unit 201, in step S12, the evaluation result is If the tumor cells are not injured (NO), it is determined in step S24 that the immunotherapeutic agent is ineffective for the patient.

 処理部201は、上記ステップS23、又はステップS24で得られた結果を記録媒体213に記録してもよい。 The processing unit 201 may record the result obtained in step S23 or step S24 in the recording medium 213.

 次に、図5を用いて、補助装置20の動作の別形態について説明する。初めに、処理部201は、検査者によって入力部211から入力される、上記1.で述べた方法により取得された腫瘍細胞傷害活性を反映する値を取得する(ステップS201)。又は、測定部30から腫瘍細胞傷害活性を算出するための情報を取得する。前記情報は、PBMCと接触した腫瘍細胞の生細胞数又は死細胞数を反映する情報であり、例えば生細胞染色試薬又は死細胞測定試薬の吸光度等である。あるいは、前記情報は、インターフェロン-γの測定した際の吸光度等である。 Next, another mode of operation of the auxiliary device 20 will be described with reference to FIG. First, the processing unit 201 is input by the inspector from the input unit 211. A value that reflects the tumor cytotoxic activity obtained by the method described in (1) is obtained (step S201). Alternatively, the information for calculating the tumor cytotoxic activity is acquired from the measurement unit 30. The information is information that reflects the number of living cells or dead cells of the tumor cells that have come into contact with PBMC, and is, for example, the absorbance of a living cell staining reagent or a dead cell measurement reagent. Alternatively, the information is the absorbance when interferon-γ is measured.

 次に、処理部201は、ステップS201で取得した腫瘍細胞傷害活性を反映する値を主記憶部202又は補助記憶部204に記憶されている基準値と比較する(ステップS202)。 Next, the processing unit 201 compares the value that reflects the tumor cell cytotoxic activity acquired in step S201 with the reference value stored in the main storage unit 202 or the auxiliary storage unit 204 (step S202).

 続いて処理部201は、ステップS202の比較結果からステップS201で取得した腫瘍細胞傷害活性を反映する値が基準値よりも否かを決定する(ステップS203)。 Subsequently, the processing unit 201 determines from the comparison result of step S202 whether or not the value that reflects the tumor cell cytotoxic activity obtained in step S201 is higher than the reference value (step S203).

 処理部201は、ステップS203において腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合には、ステップS204に進み、免疫療法剤が有用であると決定し、その結果を出力部212から出力することにより提示する。ステップS203において、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合には、処理部201は、ステップS205に進み、免疫療法剤が有用でないと決定し、その結果を出力部212から出力することにより提示する。 When the processing unit 201 determines in step S203 that the value that reflects the tumor cytotoxic activity is higher than the reference value (YES), the processing unit 201 proceeds to step S204, determines that the immunotherapeutic agent is useful, and as a result, To be presented by outputting from the output unit 212. When it is determined in step S203 that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), the processing unit 201 proceeds to step S205, determines that the immunotherapeutic agent is not useful, and To be presented by outputting from the output unit 212.

 有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS203において、腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合に、スッテプS204において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS203において、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合に、スッテプS205において、患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する。 When the usefulness is the presence or absence of the adverse event that requires the treatment interruption due to the immunotherapeutic agent and the administration cannot be resumed, the processing unit 201 determines the tumor cytotoxic activity in step S203. When it is determined that the value to be reflected is higher than the reference value (YES), in step S204, the immunotherapeutic agent is administered to the patient for an adverse event requiring treatment interruption and resumption of administration. Show what you don't. When the usefulness is the presence or absence of the adverse event that requires the treatment interruption due to the immunotherapeutic agent and the administration cannot be resumed, the processing unit 201 determines the tumor cytotoxic activity in step S203. When it is determined that the reflected value is lower than the reference value (NO), in step S205, the immunotherapeutic agent is administered to the patient for an adverse event that requires treatment interruption and cannot be resumed. Offer to wake up.

 有用性が、免疫療法剤の効果の有無であるとき、前記処理部201は、スッテプS203において、腫瘍細胞傷害活性を反映する値が基準値よりも高い(YES)と決定した場合に、スッテプS204において、患者に対して前記免疫療法剤が有効であることを提示する。有用性が、免疫療法剤に起因する治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であるとき、前記処理部201は、スッテプS203において、前記評価結果が、前記腫瘍細胞が傷害されていないことを示す(NO)の場合に、腫瘍細胞傷害活性を反映する値が基準値よりも低い(NO)と決定した場合に、スッテプS205において、患者に対して前記免疫療法剤が無効であることを提示する。 When the usefulness is the presence or absence of the effect of the immunotherapeutic agent, the processing unit 201 determines in Step S203 that the value reflecting the tumor cytotoxic activity is higher than the reference value (YES), Step S204. In, it is shown to the patient that the immunotherapeutic agent is effective. When the usefulness is the presence or absence of an adverse event that requires treatment interruption due to an immunotherapeutic agent and makes it impossible to resume administration, the processing unit 201, in step S203, the evaluation result is In the case of indicating that the tumor cells are not injured (NO), if it is determined that the value reflecting the tumor cytotoxic activity is lower than the reference value (NO), in step S205, the Present that the immunotherapeutic is ineffective.

 処理部201は、上記ステップS204、又はステップS205で得られた結果を記録媒体113に記録してもよい。 The processing unit 201 may record the result obtained in step S204 or step S205 in the recording medium 113.

 上記1.及び2.における用語の説明、各工程の説明は、ここに援用される。 Above 1. And 2. The explanation of terms and explanation of each step are incorporated herein by reference.

7.悪性腫瘍を治療するための免疫療法剤の有用性の決定を補助するためのコンピュータプログラム及び前記コンピュータプログラムを記憶した記憶媒体
 本明細書に開示されるある実施形態は、コンピュータに実行させたときに、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助するためのコンピュータプログラムに関する。具体的には、上記4.で述べた悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助する補助装置20の動作を制御するコンピュータプログラムである。
7. A computer program for assisting the determination of the usefulness of an immunotherapeutic agent for treating a malignant tumor, and a storage medium storing the computer program, according to an embodiment disclosed herein. An immunotherapeutic agent for treating a malignant tumor, the invention relates to a computer program for assisting in determining the usefulness in a patient having the malignant tumor. Specifically, the above 4. The computer program for controlling the operation of the auxiliary device 20 for assisting the determination of the usefulness of the immunotherapeutic agent for treating the malignant tumor described in 1. in the patient having the malignant tumor.

 本実施形態のコンピュータプログラムが実行する各ステップは、上記4.で述べたステップS21~24又はステップS201~205である。上記1.及び2.における用語の説明、各工程の説明は、ここに援用される。 The steps executed by the computer program of this embodiment are the same as those in 4. These are steps S21 to S24 or steps S201 to 205 described above. Above 1. And 2. The explanation of terms and explanation of each step are incorporated herein by reference.

 さらに、本明細書に開示されるある実施形態は、前記コンピュータプログラムを記憶した、記憶媒体に関する。すなわち、前記コンピュータプログラムは、ハードディスク、フラッシュメモリ等の半導体メモリ素子、光ディスク等の記憶媒体に記憶される。前記記憶媒体へのプログラムの記憶形式は、前記提示装置が前記プログラムを読み取り可能である限り制限されない。前記記憶媒体への記憶は、不揮発性であることが好ましい。 Furthermore, an embodiment disclosed in the present specification relates to a storage medium storing the computer program. That is, the computer program is stored in a storage medium such as a hard disk, a semiconductor memory device such as a flash memory, or an optical disk. The storage format of the program in the storage medium is not limited as long as the presentation device can read the program. The storage in the storage medium is preferably non-volatile.

8.免疫療法剤
 本明細書に開示されるある実施形態は、悪性腫瘍を治療するための免疫療法剤に関する。好ましくは、上記2.(上記1.に記載した事項を援用する部分を含む)において述べた提示方法により、免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される免疫療法剤に関する。言い換えると、免疫療法剤は、前記提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者を治療するために使用されるものである。
8. Immunotherapeutic Agents Certain embodiments disclosed herein relate to immunotherapeutic agents for treating malignant tumors. Preferably, the above 2. The present invention relates to an immunotherapeutic agent that is administered to a patient having a malignant tumor for which the immunotherapeutic agent is shown to be useful by the presenting method described in (including the part incorporating the matters described in 1 above). In other words, the immunotherapeutic agent is used for treating a patient having a malignant tumor in which the immunotherapeutic agent is suggested to be useful in the above-mentioned presentation method.

 本開示における免疫療法剤は、悪性腫瘍の免疫療法に使用できる限り制限されない。免疫療法剤には、公知の薬剤、新規の薬剤、及びドラッグリポジショニングにより効能が再発見された薬剤等が含まれ得る。また、免疫療法剤は、所定の機関により医薬品として承認された薬剤であっても、未承認の薬剤であってもよい。好ましくは、本開示における免疫療法剤には、免疫チェックポイント阻害剤として投与される、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、抗LAG3抗体等;養子免疫療法のために投与されるキメラ抗原受容体-T細胞等;サイトカイン療法のために投与されるインターロイキン2等;腫瘍ワクチン療法のために投与される腫瘍抗原由来の蛋白質又はペプチド(例えばWT-1、NY-ESO-1)等;二重特異性T細胞エンゲージャー(BiTE(登録商標)ともいう)、若しくは二重特性抗体(バイスペシフィック抗体)等を有効成分とする薬剤が含まれる。免疫療法剤としては、免疫チェックポイント阻害剤であることが好ましい。言い換えると、前記有効成分は、免疫療法剤を製造するために使用することができる。抗PD1抗体を含む免疫療法剤としては、ニボルマブ、ペムブロリズマブ等を挙げることができる。抗PD-L1抗体を含む免疫療法剤としては、アテゾリズマブ、デュルバルマブ、アベルマブ等を挙げることができる。抗CTLA-4抗体を含む免疫療法剤としては、イピリムマブ等を挙げることができる。 The immunotherapy agent in the present disclosure is not limited as long as it can be used for immunotherapy of malignant tumor. Immunotherapeutic agents may include known agents, new agents, agents whose efficacy has been rediscovered by drug repositioning, and the like. The immunotherapeutic agent may be a drug approved as a drug by a predetermined institution or an unapproved drug. Preferably, the immunotherapeutic agent according to the present disclosure is an anti-PD1 antibody, anti-PD-L1 antibody, anti-CTLA-4 antibody, anti-Tim3 antibody, anti-LAG3 antibody, or the like, which is administered as an immune checkpoint inhibitor; Chimeric antigen receptor-T cells and the like administered for; interleukin-2 and the like administered for cytokine therapy; tumor antigen-derived proteins or peptides (eg WT-1, administered for tumor vaccine therapy) NY-ESO-1) and the like; a drug having an active ingredient such as a bispecific T cell engager (also referred to as BiTE (registered trademark)) or a bispecific antibody (bispecific antibody). The immunotherapeutic agent is preferably an immune checkpoint inhibitor. In other words, the active ingredient can be used to produce an immunotherapeutic agent. Examples of immunotherapeutic agents containing anti-PD1 antibody include nivolumab, pembrolizumab and the like. Examples of immunotherapeutic agents containing anti-PD-L1 antibody include atezolizumab, durvalumab, avelumab and the like. Examples of immunotherapeutic agents containing anti-CTLA-4 antibody include ipilimumab and the like.

 エンゲージャーとしては、上記1.に記載のエンゲージャーを例示することができる。好ましくは、二重特異性T細胞エンゲージャーとしては、CD19とCD3を標的とするエンゲージャー(例えば、ブリナツモマブ等)、EphA2とCD3を標的とするエンゲージャーを挙げることができる。また二重特異性抗体としては、EpCAMとCD3を標的とする抗体を挙げることができる。 As an engager, 1. The engager described in 1. can be exemplified. Preferably, the bispecific T cell engager may include an engager targeting CD19 and CD3 (eg, blinatumomab), and an engager targeting EphA2 and CD3. Examples of bispecific antibodies include antibodies targeting EpCAM and CD3.

 免疫療法剤の有効成分が抗体である場合、前記抗体は、少なくとも前記抗原結合領域を含んでいればよい。前記抗原結合領域は、標的とする抗原と結合する限り制限されない。前記抗原結合領域は、例えば、前記抗原に結合する免疫グロブリンの相補鎖決定領域1(CDR1)、相補鎖決定領域2(CDR2)及び相補鎖決定領域3(CDR3)よりなる群からなる少なくとも一つのCDRを含む。前記抗原結合領域は、好ましくは、CDR1、CDR2及びCDR3よりなる群からなる少なくとも二つのCDRを含み、より好ましくは、CDR1、CDR2及びCDR3を含む。前記CDRは、免疫グロブリンの重鎖由来であっても、免疫グロブリンの軽鎖由来であってもよい。さらに好ましくは、前記抗原結合領域において、各CDRは、それぞれに対応するフレームワーク領域に隣接していてもよい。また、前記抗原結合領域は、免疫グロブリンの可変領域(V領域)、sc-Fv、v-NAR、免疫グロブリンのFab領域又はFab’領域等であってもよい。また、前記抗体は、キメラ抗体、ヒト化抗体等であってもよい。前記抗体には、単一ドメイン抗体、一本鎖抗体、マキシボディ、ミニボディ、イントラボディ、ダイアボディ、トリアボディ、テトラボディ等を含む。 When the active ingredient of the immunotherapeutic agent is an antibody, the antibody may include at least the antigen binding region. The antigen-binding region is not limited as long as it binds to the target antigen. The antigen-binding region is, for example, at least one of the group consisting of complementary chain determining region 1 (CDR1), complementary chain determining region 2 (CDR2) and complementary chain determining region 3 (CDR3) of immunoglobulin that binds to the antigen. Includes CDR. The antigen-binding region preferably comprises at least two CDRs consisting of CDR1, CDR2 and CDR3, more preferably CDR1, CDR2 and CDR3. The CDR may be derived from an immunoglobulin heavy chain or an immunoglobulin light chain. More preferably, in the antigen-binding region, each CDR may be adjacent to the corresponding framework region. The antigen-binding region may be an immunoglobulin variable region (V region), sc-Fv, v-NAR, immunoglobulin Fab region or Fab' region, or the like. Further, the antibody may be a chimeric antibody, a humanized antibody or the like. The antibodies include single domain antibodies, single chain antibodies, maxibodies, minibodies, intrabodies, diabodies, triabodies, tetrabodies and the like.

 免疫療法剤が、公知の薬剤である場合、又はドラッグリポジショニングにより効能が再発見された薬剤等であり、用法及び用量が決定されている場合には、前記免疫療法剤の用法及び用量は既知の用法及び用量にしたがって決定することができる。 If the immunotherapeutic agent is a known drug, or if the efficacy and the like have been rediscovered by drug repositioning and the usage and dose have been determined, the usage and dose of the immunotherapeutic agent are known. Can be determined according to the usage and dose.

 免疫療法剤は、有効成分100質量%からなるものであってもよいが、通常は有効成分に加えて、薬学的に許容される担体又は添加剤を含有するものである。後者の場合の免疫療法剤の有効成分の割合は、制限はされないものの、通常5~95質量%の範囲から選択することができる。好ましくは30~80質量%の割合である。 The immunotherapeutic agent may consist of 100% by mass of the active ingredient, but usually it contains a pharmaceutically acceptable carrier or additive in addition to the active ingredient. In the latter case, the ratio of the active ingredient of the immunotherapeutic agent is not particularly limited, but it can usually be selected from the range of 5 to 95% by mass. The proportion is preferably 30 to 80% by mass.

 免疫療法剤の投与形態としては、経口投与;並びに静脈内投与、筋肉内投与、皮下投与、経粘膜投与、経皮投与、及び直腸内投与等の非経口投与を挙げることができる。好ましくは経口投与及び静脈内投与であり、より好ましくは静脈内投与である。免疫療法剤は、かかる投与方法に応じて、種々の形態の製剤(剤型)に調製することができる。 The dosage form of the immunotherapeutic agent includes oral administration; parenteral administration such as intravenous administration, intramuscular administration, subcutaneous administration, transmucosal administration, transdermal administration, and rectal administration. Oral administration and intravenous administration are preferred, and intravenous administration is more preferred. The immunotherapeutic agent can be prepared into various forms of preparations (forms) depending on the administration method.

 以下に、各剤型について説明するが、本明細書において用いられる剤型はこれらに限定されるものではなく、医薬製剤分野において通常用いられる各種剤型を用いることができる。 Each dosage form will be described below, but the dosage form used in the present specification is not limited to these, and various dosage forms normally used in the field of pharmaceutical formulation can be used.

 経口投与を行う場合の剤型として、散剤、顆粒剤、カプセル剤、丸剤、錠剤、エリキシル剤、懸濁剤、乳剤及びシロップ剤を挙げることができ、これらの中から適宜選択することができる。また、それらの製剤について徐放化、安定化、易崩壊化、難崩壊化、腸溶性化、易吸収化等の修飾を施すことができる。 Examples of dosage forms for oral administration include powders, granules, capsules, pills, tablets, elixirs, suspensions, emulsions and syrups, which can be appropriately selected from these. .. Further, these preparations can be modified such as sustained release, stabilization, easy disintegration, difficult disintegration, enteric coating, and easy absorption.

 また、静脈内投与、筋肉内投与、又は皮下投与を行う場合の剤型として、注射剤又は点滴剤(用時調製の乾燥品を含む)等があり、適宜選択することができる。 In addition, the dosage form for intravenous administration, intramuscular administration, or subcutaneous administration includes injections and infusions (including dry products prepared at the time of use), which can be appropriately selected.

 経粘膜投与、経皮投与、又は直腸内投与を行う場合の剤型として、咀嚼剤、舌下剤、パッカル剤、トローチ剤、軟膏剤、貼布剤、液剤等があり、適用場所に応じて適宜選択するここができる。また、それらの製剤について徐放化、安定化、易崩壊化、難崩壊化、易吸収化等の修飾を施すことができる。 Dosage forms for transmucosal administration, transdermal administration, or rectal administration include chewing agents, sublingual agents, puckers, troches, ointments, patches, liquids, etc. You can choose here. Further, these preparations can be modified such as sustained release, stabilization, easy disintegration, difficult disintegration, and easy absorption.

 免疫療法剤にはその剤形(経口投与または各種の非経口投与の剤形)に応じて、薬学的に許容される担体及び添加剤を配合することができる。薬学的に許容される担体及び添加剤としては、溶剤、賦形剤、コーティング剤、基剤、結合剤、滑沢剤、崩壊剤、溶解補助剤、懸濁化剤、粘稠剤、乳化剤、安定剤、緩衝剤、等張化剤、無痛化剤、保存剤、矯味剤、芳香剤、着色剤が挙げられる。以下に、医薬上許容される担体及び添加剤の具体例を列挙するが、これらに制限されるものではない。 The immunotherapeutic agent can be mixed with pharmaceutically acceptable carriers and additives depending on its dosage form (oral administration or various parenteral administration dosage forms). Examples of pharmaceutically acceptable carriers and additives include solvents, excipients, coating agents, bases, binders, lubricants, disintegrating agents, solubilizing agents, suspending agents, thickening agents, emulsifying agents, Stabilizers, buffers, isotonic agents, soothing agents, preservatives, corrigents, fragrances, and colorants are mentioned. Specific examples of pharmaceutically acceptable carriers and additives are listed below, but the invention is not limited thereto.

 溶剤としては、精製水、滅菌精製水、注射用水、生理食塩液、ラッカセイ油、エタノール、グリセリン等を挙げることができる。賦形剤としては、デンプン類(例えばバレイショデンプン、コムギデンプン、トウモロコシデンプン)、乳糖、ブドウ糖、白糖、結晶セルロース、硫酸カルシウム、炭酸カルシウム、炭酸水素ナトリウム、塩化ナトリウム、タルク、酸化チタン、トレハロース、キシリトール等を挙げることができる。 As the solvent, purified water, sterilized purified water, water for injection, physiological saline, peanut oil, ethanol, glycerin and the like can be mentioned. Excipients include starches (eg potato starch, wheat starch, corn starch), lactose, glucose, sucrose, crystalline cellulose, calcium sulfate, calcium carbonate, sodium hydrogen carbonate, sodium chloride, talc, titanium oxide, trehalose, xylitol. Etc. can be mentioned.

 結合剤としては、デンプン及びその誘導体、セルロース及びその誘導体(例えばメチルセルロース、エチルセルロース、ヒドロキシプロピルセルロース、カルボキシメチルセルロース)、ゼラチン、アルギン酸ナトリウム、トラガント、アラビアゴム等の天然高分子化合物、ポリビニルピロリドン、ポリビニルアルコール等の合成高分子化合物、デキストリン、ヒドロキシプロピルスターチ等を挙げることができる。 As the binder, starch and its derivatives, cellulose and its derivatives (for example, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, carboxymethyl cellulose), gelatin, sodium alginate, tragacanth, natural polymer compounds such as gum arabic, polyvinylpyrrolidone, polyvinyl alcohol, etc. Examples thereof include synthetic polymer compounds, dextrin, hydroxypropyl starch and the like.

 滑沢剤としては、軽質無水ケイ酸、ステアリン酸及びその塩類(たとえばステアリン酸マグネシウム)、タルク、ワックス類、コムギデンブン、マクロゴール、水素添加植物油、ショ糖脂肪酸エステル、ポリエチレングリコール、シリコン油等を挙げることができる。 Lubricants include light anhydrous silicic acid, stearic acid and salts thereof (eg magnesium stearate), talc, waxes, wheat den bun, macrogol, hydrogenated vegetable oil, sucrose fatty acid ester, polyethylene glycol, silicone oil and the like. be able to.

 崩壊剤としては、デンプン及びその誘導体、寒天、ゼラチン末、炭酸水素ナトリウム、炭酸カルシウム、セルロース及びその誘導体、ヒドロキシプロピルスターチ、カルボキシメチルセルロース及びその塩類並びにその架橋体、低置換型ヒドロキシプロピルセルロース等を挙げることができる。 Examples of the disintegrant include starch and its derivatives, agar, gelatin powder, sodium hydrogen carbonate, calcium carbonate, cellulose and its derivatives, hydroxypropyl starch, carboxymethyl cellulose and its salts and cross-linked products thereof, low-substituted hydroxypropyl cellulose and the like. be able to.

 溶解補助剤としては、シクロデキストリン、エタノール、プロピレングリコール、ポリエチレングリコール等を挙げることができる。懸濁化剤としては、カルボキシメチルセルロースナトリウム、ポリピニルピロリドン、アラビアゴム、トラガント、アルギン酸ナトリウム、モノステアリン酸アルミニウム、クエン酸、各種界面活性剤等を挙げることができる。 Examples of the solubilizing agent include cyclodextrin, ethanol, propylene glycol, polyethylene glycol and the like. Examples of the suspending agent include sodium carboxymethyl cellulose, polypinylpyrrolidone, gum arabic, tragacanth, sodium alginate, aluminum monostearate, citric acid and various surfactants.

 粘稠剤としては、カルボキシメチルセルロースナトリウム、ポリピニルピロリドン、メチルセルロース、ヒドロキシプロピルメチルセルロース、ポリビニルアルコール、トラガント、アラビアゴム、アルギン酸ナトリウム等を挙げることができる。 Examples of the thickener include sodium carboxymethylcellulose, polypinylpyrrolidone, methylcellulose, hydroxypropylmethylcellulose, polyvinyl alcohol, tragacanth, gum arabic, sodium alginate and the like.

 乳化剤は、アラビアゴム、コレステロール、トラガント、メチルセルロース、レシチン、各種界面活性剤(例えば、ステアリン酸ポリオキシル40、セスキオレイン酸ソルビタン、ポリソルベート80、ラウリル硫酸ナトリウム)等を挙げることができる。 Examples of the emulsifier include gum arabic, cholesterol, tragacanth, methyl cellulose, lecithin, various surfactants (for example, polyoxyl 40 stearate, sorbitan sesquioleate, polysorbate 80, sodium lauryl sulfate).

 安定剤としては、トコフェロール、キレート剤(たとえばEDTA、チオグリコール酸)、不活性ガス(たとえば窒素、二酸化炭素)、還元性物質(例えば亜硫酸水素ナトリウム、チオ硫酸ナトリウム、アスコルビン酸、ロンガリット)等を挙げることができる。 Examples of the stabilizer include tocopherol, chelating agents (eg EDTA, thioglycolic acid), inert gases (eg nitrogen, carbon dioxide), reducing substances (eg sodium bisulfite, sodium thiosulfate, ascorbic acid, rongalite) and the like. be able to.

 緩衝剤としては、リン酸水素ナトリウム、酢酸ナトリウム、クエン酸ナトリウム、ホウ酸等を挙げることができる。 Examples of the buffer include sodium hydrogen phosphate, sodium acetate, sodium citrate, boric acid and the like.

 等張化剤としては、塩化ナトリウム、ブドウ糖等を挙げることができる。無痛化剤こしては、局所麻酔剤(塩酸プロカイン、リドカイン)、ペンジルアルコール、ブドウ糖、ソルビトール、アミノ酸等を挙げることができる。 As the tonicity agent, sodium chloride, glucose and the like can be mentioned. Examples of soothing agents include local anesthetics (procaine hydrochloride, lidocaine), penzyl alcohol, glucose, sorbitol, amino acids and the like.

 矯味剤としては、白糖、サッカリン、カンゾウエキス、ソルビトール、キシリトール、グリセリン等を挙げることができる。芳香剤としては、トウヒチンキ、ローズ油等を挙げることができる。着色剤としては、水溶性食用色素、レーキ色素等を挙げることができる。 As the corrigent, sucrose, saccharin, licorice extract, sorbitol, xylitol, glycerin and the like can be mentioned. Examples of the aromatic agents include spruce tincture and rose oil. Examples of colorants include water-soluble food dyes and lake dyes.

 保存剤としては、安息香酸及びその塩類、パラオキシ安息香酸エステル類、クロロブタノール、逆性石けん、ベンジルアルコール、フェノール、チロメサール、デヒドロ酢酸、ホウ酸、等を挙げることができる。 Examples of preservatives include benzoic acid and its salts, paraoxybenzoic acid esters, chlorobutanol, inverted soap, benzyl alcohol, phenol, tiromesal, dehydroacetic acid, boric acid, and the like.

 コーティング剤としては、白糖、ヒドロキシプロピルセルロース(HPC)、セラック、ゼラチン、グリセリン、ソルビトール、ヒドロキシプロピルメチルセルロース(HPMC)、エチルセルロース、ポリビニルピロリドン(PVP)、ヒドロキシプロピルメチルセルロースフタレート(HPMCP)、セルロースアセテートフタレート(CAP)、メチルメタアクリレート-メタアクリル酸共重合体及び上記記載した高分子等を挙げることができる。 As the coating agent, sucrose, hydroxypropylcellulose (HPC), shellac, gelatin, glycerin, sorbitol, hydroxypropylmethylcellulose (HPMC), ethylcellulose, polyvinylpyrrolidone (PVP), hydroxypropylmethylcellulose phthalate (HPMCP), cellulose acetate phthalate (CAP). ), a methylmethacrylate-methacrylic acid copolymer, and the polymers described above.

 基剤としては、ワセリン、流動パラフィン、カルナウバロウ、牛脂、硬化油、パラフィン、ミツロウ、植物油、マクロゴール、マクロゴール脂肪酸エステル、ステアリン酸、カルボキシメチルセルロースナトリウム、ベントナイト、カカオ脂、ウイテップゾール、ゼラチン、ステアリルアルコール、加水ラノリン、セタノール、軽質流動パラフィン、親水ワセリン、単軟膏、白色軟膏、親水軟膏、マクロゴール軟膏、ハードファット、水中油型乳剤性基剤、油中水型乳剤性碁剤等を挙げることができる。 As a base, petrolatum, liquid paraffin, carnauba wax, beef tallow, hydrogenated oil, paraffin, beeswax, vegetable oil, macrogol, macrogol fatty acid ester, stearic acid, sodium carboxymethyl cellulose, bentonite, cacao butter, witepsol, gelatin, stearyl. Alcohol, lanolin hydrous, cetanol, light liquid paraffin, hydrophilic petrolatum, simple ointment, white ointment, hydrophilic ointment, macrogol ointment, hard fat, oil-in-water emulsion base, water-in-oil emulsion go You can

 なお、上記の各剤型について、公知のドラッグデリバリーシステム(DDS)の技術を採用することができる。本明細書にいうDDS製剤とは、徐放化製剤、局所適用製剤(トローチ、バッカル錠、舌下錠等)、薬物放出制御製剤、腸溶性製剤及び胃溶性製剤等、投与経路、バイオアベイラビリティー、副作用等を勘案した上で、最適の製剤形態にした製剤である。 Note that well-known drug delivery system (DDS) technology can be adopted for each of the above dosage forms. As used herein, the DDS preparations include sustained-release preparations, topical preparations (troches, buccal tablets, sublingual tablets, etc.), controlled-release preparations, enteric-coated preparations and gastric-soluble preparations, administration routes, bioavailability. In consideration of side effects, etc., it is a formulation in an optimal formulation form.

 また、免疫療法剤が、新規の薬剤のように用法及び用量が決定されていない場合、その投与量は、治療対象とするがんの種類や場所、がんのステージ(進行度)、患者の病態、年齢、性別、体重、併用する抗がん剤の種類などに応じて、変動し得、これらの要因に応じて適宜設定することができる。免疫療法剤をヒトに経口投与する場合、その投与量は、有効成分の量に換算して0.03~300mg/kg/日の範囲から適宜設定することができる。免疫療法剤が静脈内投与剤である場合、の有効血中濃度が0.1~3000μg/mL、より好ましくは1~1000μg/mLの範囲となるような投与量で、1日当たりを0.5~50mg/kgとなるように投与することができる。 In addition, when the usage and dose of an immunotherapeutic agent are not determined like a new drug, the dose depends on the type and location of the cancer to be treated, the stage (progression) of the cancer, and the patient. It may vary depending on the disease state, age, sex, body weight, type of anticancer drug used in combination, etc., and can be appropriately set according to these factors. When the immunotherapeutic agent is orally administered to humans, the dose can be appropriately set within the range of 0.03 to 300 mg/kg/day in terms of the amount of the active ingredient. When the immunotherapeutic agent is an intravenous administration agent, the effective blood concentration of is 0.1 to 3000 μg/mL, more preferably 1 to 1000 μg/mL. It can be administered at ˜50 mg/kg.

 本実施形態において、免疫療法剤は、好ましくは、前記提示方法により、免疫療法剤が有用であることが提示された悪性腫瘍を有する患者(以下、「投与対象患者」ともいう)に対して、一次治療のために投与される。ここで一次治療とは、悪性腫瘍を有する患者に初めて投与される化学療法のことである。投与方法は静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably, for the patient having a malignant tumor for which the immunotherapeutic agent is suggested to be useful by the presenting method (hereinafter, also referred to as “administration target patient”), Administered for first line treatment. Here, the first-line treatment is chemotherapy that is first administered to a patient having a malignant tumor. The method of administration is intravenous administration at intervals of 2 to 4 weeks.

 本実施形態において、免疫療法剤は、好ましくは対象患者に対して、外科的な手術の前の術前化学療法のために投与される。ここで、手術前とは、好ましくは、手術施行日の90日前~1日前、より好ましくは60日前~7日前をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the target patient for preoperative chemotherapy before surgical operation. Here, the term “before surgery” refers to preferably 90 days to 1 day before, and more preferably 60 days to 7 days before, the day of surgery. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 本実施形態において、免疫療法剤は、好ましくは対象患者に対して、外科的な手術の後の術後補助化学療法のために投与される。ここで、手術後とは、好ましくは、手術施行日から1日~180日間、より好ましくは7日間~90日間をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the target patient for postoperative adjuvant chemotherapy after surgical operation. Here, the term “after surgery” means preferably 1 to 180 days, more preferably 7 to 90 days from the date of operation. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 本実施形態において、免疫療法剤は、好ましくは放射線治療の施行前の対象患者に投与される。ここで、投与方法は、好ましくは、放射線治療の施行日から42日~1日前、より好ましくは30日間~7日前をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the target patient before the radiotherapy is performed. Here, the administration method is preferably 42 days to 1 day before, more preferably 30 days to 7 days before the date of radiotherapy. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 本実施形態において、免疫療法剤は、好ましくは放射線治療を受けた対象患者に投与される。ここで、投与方法は、好ましくは、放射線治療の施行日から1日~180日間、より好ましくは3日間~90日間をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the subject patient who has undergone radiotherapy. Here, the administration method is preferably 1 to 180 days, more preferably 3 to 90 days from the date of radiotherapy. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 前記放射線治療は、患部に、好ましくはリニア・アクセラレータを用いてエックス線照射若しくは電子線照射を行うことによって実施することができる。ここでエックス線照射の条件は、腫瘍の進行度や大きさなどによって異なるが、通常1回線量1.5~3Gy,好ましくは2Gy程度の照射を、週に2~5回、好ましくは週4~5回程度、1~5週間にわたって行う方法を挙げることができる。総線量としては20~70Gy,好ましくは40~70Gy程度、より好ましくは50~60Gy程度を挙げることができる。また電子線照射の条件は、やはり腫瘍の進行度や大きさなどによって異なるが、通常1回線量2~5Gy、好ましくは4Gy程度の照射を、週1~5回、好ましくは週2~3回程度、1~5週間にわたって行う方法を挙げることができる。総線量としては30~70Gy、好ましくは40~60Gy程度を挙げることができる。 The radiation treatment can be performed by irradiating the affected area with X-rays or electron rays, preferably using a linear accelerator. Here, the conditions of X-ray irradiation vary depending on the degree of progression and size of the tumor, but usually one dose of 1.5 to 3 Gy, preferably about 2 Gy, is applied 2 to 5 times a week, preferably 4 to a week. The method may be performed about 5 times over 1 to 5 weeks. The total dose may be 20 to 70 Gy, preferably about 40 to 70 Gy, and more preferably about 50 to 60 Gy. The electron beam irradiation conditions also depend on the degree of tumor progression and size, but usually one dose of 2 to 5 Gy, preferably about 4 Gy, is irradiated 1 to 5 times a week, preferably 2 to 3 times a week. The method may be carried out for about 1 to 5 weeks. The total dose may be about 30 to 70 Gy, preferably about 40 to 60 Gy.

 本実施形態において、免疫療法剤は、好ましくは化学放射線治療の施行前の対象患者に投与される。ここで、投与方法は、好ましくは、放射線治療の施行日から42日~1日前、より好ましくは30日間~3日前をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the target patient before the chemoradiotherapy is performed. Here, the administration method is preferably 42 days to 1 day before, more preferably 30 days to 3 days before the date of radiotherapy. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 本実施形態において、免疫療法剤は、好ましくは化学放射線治療を受けた対象患者に投与される。ここで、投与方法は、好ましくは、放射線治療の施行日から1日~12か月間、より好ましくは3日間~240日間をいう。投与方法は、静脈内投与で2~4週間間隔で投与を行う。 In the present embodiment, the immunotherapeutic agent is preferably administered to the target patient who has undergone chemoradiotherapy. Here, the administration method is preferably 1 day to 12 months, more preferably 3 days to 240 days from the date of radiotherapy. The administration method is intravenous administration at intervals of 2 to 4 weeks.

 化学放射線療法とは、放射線治療と抗がん剤の併用療法である。放射線治療は1回線量2Gy、週5回、6週間、総線量としては60Gy、抗がん剤治療は白金系抗がん剤(例えば、シスプラチンまたはカルボプラチン)を含む2種類の抗がん剤治療を少なくとも2サイクル行う。 Chemoradiotherapy is a combination therapy of radiation therapy and anticancer drug. Radiation therapy is 2 Gy for one dose, 5 times a week for 6 weeks, total dose is 60 Gy, and anticancer drug treatment is two types of anticancer drug treatment including platinum anticancer drug (eg, cisplatin or carboplatin) For at least 2 cycles.

 ここで、免疫療法剤によっては、国ごとに適用例が限定されている場合がある。例えば本願出願の時点での適用例を以下に示すが、本明細書に開示される実施形態は以下の形態に限定して解釈される必要はない:
 イピリムマブ:悪性黒色腫、好ましくは根治切除不能な悪性黒色腫;及び腎細胞癌、根治切除不能又は転移性の腎細胞癌、
 ニボルマブ:悪性黒色腫;非小細胞肺癌、好ましくは切除不能な進行又は再発の非小細胞肺癌;腎細胞癌、好ましくは根治切除不能又は転移性の腎細胞癌;ホジキンリンパ腫、好ましくは再発又は難治性の古典的ホジキンリンパ腫;頭頸部癌、好ましくは再発又は遠隔転移を有する頭頸部癌;胃癌、好ましくはがん化学療法後に増悪した治癒切除不能な進行又は再発の胃癌;悪性胸膜中皮腫、好ましくはがん化学療法後に増悪した切除不能な進行又は再発の悪性胸膜中皮腫、
 ペムブロリズマブ:悪性黒色腫;非小細胞肺癌、好ましくは切除不能な進行又は再発の非小細胞肺癌;ホジキンリンパ腫、好ましくは再発又は難治性の古典的ホジキンリンパ腫;尿路上皮癌、好ましくはがん化学療法後に増悪した根治切除不能な尿路上皮癌、
 アテゾリズマブ:非小細胞肺癌、好ましくは切除不能な進行又は再発の非小細胞肺癌、
 デュルバルマブ:非小細胞肺癌、好ましくは切除不能な局所進行の非小細胞肺癌における根治的化学放射線療法後の維持療法、
 アベルマブ:メルケル細胞癌、好ましくは根治切除不能なメルケル細胞癌。
Here, depending on the immunotherapeutic agent, the application example may be limited in each country. For example, application examples at the time of filing of the present application are shown below, but the embodiments disclosed herein need not be limited to the following forms:
Ipilimumab: malignant melanoma, preferably unresectable malignant melanoma; and renal cell carcinoma, unresectable or metastatic renal cell carcinoma,
Nivolumab: malignant melanoma; non-small cell lung cancer, preferably unresectable advanced or recurrent non-small cell lung cancer; renal cell carcinoma, preferably unresectable or metastatic renal cell carcinoma; Hodgkin lymphoma, preferably relapsed or refractory Classic Hodgkin lymphoma; head and neck cancer, preferably head and neck cancer with recurrent or distant metastases; gastric cancer, preferably unresectable advanced or recurrent gastric cancer exacerbated after cancer chemotherapy; malignant pleural mesothelioma, Unresectable advanced or recurrent malignant pleural mesothelioma, which is preferably aggravated after cancer chemotherapy,
Pembrolizumab: malignant melanoma; non-small cell lung cancer, preferably unresectable advanced or recurrent non-small cell lung cancer; Hodgkin lymphoma, preferably relapsed or refractory classic Hodgkin lymphoma; urothelial cancer, preferably cancer chemistry Unresectable urothelial cancer that worsened after therapy,
Atezolizumab: non-small cell lung cancer, preferably unresectable advanced or recurrent non-small cell lung cancer,
Durvalumab: maintenance therapy after definitive chemoradiotherapy in non-small cell lung cancer, preferably unresectable locally advanced non-small cell lung cancer,
Avelumab: Merkel cell carcinoma, preferably unresectable Merkel cell carcinoma.

 本実施形態において、免疫療法剤は、異なる2種以上を組み合わせて投与することができる。好ましくは、免疫チェックポイント阻害剤と、他の1種以上の免疫チェックポイント阻害剤以外の免疫療法剤とを組み合わせて投与することができる。また、より好ましくは、免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤を組み合わせて投与することができる。免疫チェックポイント阻害剤と、他の1種以上の免疫チェックポイント阻害剤とを組み合わせる例として、好ましくは、抗PD1抗体を含む免疫チェックポイント阻害剤と、他の抗体(例えば抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む免疫チェックポイント阻害剤)との組み合わせることができる。より好ましくは、抗PD1抗体を含む免疫チェックポイント阻害剤と、抗CTLA-4抗体とを組み合わせることができる。1種以上とは、好ましくは2種以上、より好ましくは3種以上である。 In the present embodiment, two or more different immunotherapeutic agents can be administered in combination. Preferably, the immune checkpoint inhibitor can be administered in combination with an immunotherapeutic agent other than one or more other immune checkpoint inhibitors. Also, more preferably, the immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor can be administered in combination. As an example of combining an immune checkpoint inhibitor and one or more other immune checkpoint inhibitors, preferably, an immune checkpoint inhibitor containing an anti-PD1 antibody and another antibody (eg, an anti-PD-L1 antibody, Anti-CTLA-4 antibody, anti-Tim3 antibody, or anti-LAG3 antibody-containing immune checkpoint inhibitor). More preferably, an immune checkpoint inhibitor containing an anti-PD1 antibody can be combined with an anti-CTLA-4 antibody. One or more is preferably two or more, more preferably three or more.

 本実施形態において「組み合わせ」又は「組み合わせる」とは、
(i)最初から免疫チェックポイント阻害剤の有効成分と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)の有効成分の両方が含まれている状態(配合剤)である場合、
(ii)免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とがそれぞれ別個の包装形態で存在し、組み合わせ物(キット)として販売される場合、又は
(iii)免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とがそれぞれ別個の包装形態で、また別個の流通経路で市場に存在し、使用時に組み合わせて使用される場合を包含する意味で用いられる。
In the present embodiment, “combination” or “combination” means
(I) Includes both an active ingredient of an immune checkpoint inhibitor from the beginning and an active ingredient of one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors). If it is in a state (combination),
(Ii) A combination product in which the immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) exist in separate packaging forms, respectively. Or (iii) an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors), respectively. It is used in the meaning of including a case where they are present in the market in separate packaging forms and in separate distribution channels and used in combination when used.

 また「免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とを組み合わせる」との表現には、「免疫チェックポイント阻害剤の有効成分と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)の有効成分とを含む」、「実質的に免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とからなる」、「免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とからなる」のすべての意味が含まれるものとする。 In addition, the expression "combining an immune checkpoint inhibitor with one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors)" means "immunity checkpoint inhibitor". Including an active ingredient of an inhibitor and an active ingredient of one or more immunotherapeutic agents (or one or more other immune checkpoint inhibitors) other than the immune checkpoint inhibitor", "substantially immune checkpoint inhibition" Agent and one or more immunotherapeutic agents other than immune checkpoint inhibitors (or other one or more immune checkpoint inhibitors)", "Immune checkpoint inhibitors and 1 other than immune checkpoint inhibitors" It consists of one or more immunotherapeutic agents (or one or more other immune checkpoint inhibitors)".

 例えば、免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)との投与のタイミングは、免疫チェックポイント阻害剤と免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とが別々に投与される形態を有するものとして、それぞれ別個の包装形態からなる免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)を、同時又は時間をずらして患者に投与する形態のものを挙げることができる。 For example, the timing of administration of an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) is different from that of the immune checkpoint inhibitor. Immune check consisting of separate packaging forms, each having a form in which it is separately administered with one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) A form in which a point inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or other one or more immune checkpoint inhibitors) are administered to the patient at the same time or at staggered times be able to.

 同時に投与する形態としては、経口投与形態を有する免疫チェックポイント阻害剤と、経口投与形態を有する免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とを同時に経口投与する形態;非経口投与形態を有する免疫チェックポイント阻害剤と、非経口投与形態を有する免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)とを同時に非経口投与する形態を挙げることができる。 As the mode of simultaneous administration, an immune checkpoint inhibitor having an oral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral dosage form (or one or more other immune checkpoint inhibitors) Agent) and an immune checkpoint inhibitor having a parenteral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having a parenteral dosage form (or one or more other immunotherapeutic agents) (Immune checkpoint inhibitor) and a form of parenteral administration at the same time.

 時間をずらして投与する形態としては、経口投与形態を有する免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)をそれぞれ時間をずらして経口投与する形態;非経口投与形態を有する免疫チェックポイント阻害剤と、非経口投与形態を有する免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)をそれぞれ時間をずらして非経口投与する形態を挙げることができる。 As a form of staggered administration, an immune checkpoint inhibitor having an oral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) Orally administered at different times; an immune checkpoint inhibitor having a parenteral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having a parenteral dosage form (or another one) The above-mentioned immune checkpoint inhibitor) may be parenterally administered at different times.

 一方が経口投与形態で、他方が非経口投与形態である場合に、両者を同時にまた並行して投与する形態を挙げることができる。両者を時間をずらして投与する形態としては、経口投与形態を有する免疫チェックポイント阻害剤と、非経口投与形態を有する免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)をそれぞれ時間をずらして投与する形態;非経口投与形態を有する免疫チェックポイント阻害剤と、経口投与形態を有する免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)をそれぞれ時間をずらして投与する形態を挙げることができる。 When one is an oral administration form and the other is a parenteral administration form, a form in which both are administered simultaneously and in parallel can be mentioned. As a form of administering both of them with a staggered time, one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral administration form and the immune checkpoint inhibitor having a parenteral administration form (or another one type) The above-mentioned immune checkpoint inhibitors) are administered at different times; an immune checkpoint inhibitor having a parenteral dosage form, and one or more immunotherapeutic agents other than the immune checkpoint inhibitor having an oral dosage form (Or one or more other immune checkpoint inhibitors) may be administered at staggered times.

 時間をずらして投与する場合、免疫チェックポイント阻害剤に先立って免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)を投与しても、また免疫チェックポイント阻害剤投与後に免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)を投与しても、いずれの順番でもよい。 When administered at staggered times, even if one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) are administered prior to the immune checkpoint inhibitor, In addition, one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) may be administered after the administration of the immune checkpoint inhibitor, or in any order.

 具体的には、免疫チェックポイント阻害剤と、他の免疫療法剤とを組み合わせた治療剤が経口投与剤である場合、免疫チェックポイント阻害剤の投与量は、0.003~3000mg/kg/日の範囲から適宜設定することができる。また組み合わせる他の免疫療法剤の投与量も0.1~5000mg/kg/日、0.1~5000mg/kg/回、0.1~5000mg/m2/回又は10万~500万JRU/回から適宜設定することができる。また、投薬回数は1~8日間投与し、5日~9週間の休薬期間を設けることができる。 Specifically, when the therapeutic agent that is a combination of an immune checkpoint inhibitor and another immunotherapeutic agent is an oral administration agent, the dose of the immune checkpoint inhibitor is 0.003 to 3000 mg/kg/day. It can be set appropriately from the range. The dose of other immunotherapeutic agents to be combined is also 0.1 to 5000 mg/kg/day, 0.1 to 5000 mg/kg/dose, 0.1 to 5000 mg/m2/d or 100,000 to 5 million JRU/dose. It can be set appropriately. In addition, the frequency of administration may be 1 to 8 days, and a drug holiday of 5 days to 9 weeks may be provided.

 免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)を組み合わせた治療剤が静脈内投与剤である場合、免疫チェックポイント阻害剤の有効成分の有効血中濃度が0.1~3000μg/mL、より好ましくは1~1000μg/mLの範囲となるような投与量で、1日当たりを0.5~50mg/kgとなるように投与することがでる。また免疫チェックポイント阻害剤以外の1種以上の免疫療法剤(又は他の1種以上の免疫チェックポイント阻害剤)の投与量も通常投与する量の10~100%、好ましくは10~95%の範囲において適宜設定することができる。 When the therapeutic agent that combines an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor (or one or more other immune checkpoint inhibitors) is an intravenous administration agent, The dose in which the effective blood concentration of the active ingredient of the checkpoint inhibitor is 0.1 to 3000 μg/mL, more preferably 1 to 1000 μg/mL, and the daily dose is 0.5 to 50 mg/kg. Can be administered as Further, the dose of one or more immunotherapeutic agents (or other one or more immune checkpoint inhibitors) other than the immune checkpoint inhibitor is also 10 to 100%, preferably 10 to 95% of the normally administered dose. It can be appropriately set within the range.

 本実施形態において、1種以上の免疫療法剤(免疫チェックポイント阻害剤と、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と投与する場合、又は免疫チェックポイント阻害剤と、他の1種以上の免疫チェックポイント阻害剤を投与する場合を含む)は、免疫療法剤以外の免疫療法剤以外の1種以上の抗がん剤(以下、単に「他の抗がん剤」ともいう)と組み合わせて投与されてもよい。本実施形態において、好ましくは、1種以上の免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とを組み合わせた治療剤である。1種以上とは、好ましくは2種以上、より好ましくは3種以上である。 In the present embodiment, when one or more immunotherapeutic agents (an immune checkpoint inhibitor and one or more immunotherapeutic agents other than the immune checkpoint inhibitor are administered, or an immune checkpoint inhibitor and another 1 Including one or more immune checkpoint inhibitors) is one or more anti-cancer agents other than immuno-therapeutic agents other than immuno-therapeutic agents (hereinafter also simply referred to as "other anti-cancer agents") May be administered in combination with. In the present embodiment, the therapeutic agent is preferably a combination of one or more immunotherapeutic agents and one or more anti-cancer agents other than the immunotherapeutic agents. One or more is preferably two or more, more preferably three or more.

 本実施形態において「組み合わせる」又は「組み合わせる」とは、
(i)最初から免疫療法剤の有効成分と免疫療法剤以外の1種以上の抗がん剤の有効成分の両方が含まれている状態(配合剤)である場合、
(ii)免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とがそれぞれ別個の包装形態で存在し、組み合わせ物(キット)として販売される場合、又は
(iii)免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とがそれぞれ別個の包装形態で、また別個の流通経路で市場に存在し、使用時に組み合わせて使用される場合を包含する意味で用いられる。
In the present embodiment, “combining” or “combining” means
(I) In the case where the active ingredient of the immunotherapeutic agent and the active ingredient of one or more anti-cancer agents other than the immunotherapeutic agent are contained from the beginning (combination drug),
(Ii) When the immunotherapeutic agent and one or more anti-cancer agents other than the immunotherapeutic agent exist in separate packaging forms and are sold as a combination (kit), or (iii) the immunotherapeutic agent One or more anti-cancer agents other than immunotherapeutic agents are used in the meaning of including the case where they are present in the market in separate packaging forms and in separate distribution channels and used in combination at the time of use.

 また「免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とを組み合わせる」との表現には、「免疫療法剤の有効成分と免疫療法剤以外の1種以上の抗がん剤の有効成分とを含む」、「実質的に免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とからなる」、「免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とからなる」のすべての意味が含まれるものとする。 In addition, the expression "combining an immunotherapeutic agent with one or more anticancer agents other than the immunotherapeutic agent" means "the active ingredient of the immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent". Active ingredient of "," consisting essentially of an immunotherapeutic agent and one or more anti-cancer agents other than the immunotherapeutic agent, ""the immunotherapeutic agent and one or more anti-cancer agents other than the immunotherapeutic agent. It is meant to include all meanings of "comprising a drug".

 例えば、免疫療法剤と、免疫療法剤以外の1種以上の抗がん剤との投与のタイミングは、免疫療法剤と免疫療法剤以外の1種以上の抗がん剤とが別々に投与される形態を有するものとして、それぞれ別個の包装形態から免疫療法剤と、免疫療法剤以外の1種以上の抗がん剤を、同時又は時間をずらして患者に投与する形態のものを挙げることができる。 For example, the timing of administration of the immunotherapeutic agent and the one or more anti-cancer agents other than the immuno-therapeutic agent is such that the immuno-therapeutic agent and the one or more anti-cancer agents other than the immunotherapeutic agent are separately administered. Examples of those having different forms include those in which the immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent are administered to the patient at the same time or at different times from separate packaging forms. it can.

 同時に投与する形態としては、経口投与形態を有する免疫療法剤と、経口投与形態を有する免疫療法剤以外の1種以上の抗がん剤とを同時に経口投与する形態;非経口投与形態を有する免疫療法剤と、非経口形態を有する免疫療法剤以外の1種以上の抗がん剤とを同時に非経口投与する形態を挙げることができる。 As a form of simultaneous administration, an immunotherapeutic agent having an oral administration form and one or more anti-cancer agents other than the immunotherapeutic agent having an oral administration form are orally administered simultaneously; immunity having a parenteral administration form An example is a form in which a therapeutic agent and one or more anticancer agents other than the immunotherapeutic agent having a parenteral form are administered parenterally at the same time.

 時間をずらして投与する形態としては、経口投与形態を有する免疫療法剤と、経口投与形態を有する免疫療法剤以外の1種以上の抗がん剤をそれぞれ時間をずらして経口投与する形態;非経口投与形態を有する免疫療法剤と、非経口投与形態を有する免疫療法剤以外の1種以上の抗がん剤をそれぞれ時間をずらして非経口投与する形態を挙げることができる。 As a form of staggered administration, an immunotherapeutic agent having an oral dosage form and one or more anticancer agents other than the immunotherapeutic agent having an oral dosage form are orally administered at staggered times; The immunotherapy agent having an oral dosage form and one or more anti-cancer agents other than the immunotherapy agent having a parenteral dosage form may be parenterally administered at different times.

 一方が経口投与形態で、他方が非経口投与形態である場合に、両者を同時にまた並行して投与する形態を挙げることができる。両者を時間をずらして投与する形態としては、経口投与形態を有する免疫療法剤と、非経口投与する免疫療法剤以外の1種以上の抗がん剤をそれぞれ時間をずらしてする形態;非経口投与形態を有する免疫療法剤と、経口投与する免疫療法剤以外の1種以上の抗がん剤をそれぞれ時間をずらして投与する形態が含まれる。 When one is an oral administration form and the other is a parenteral administration form, a form in which both are administered simultaneously and in parallel can be mentioned. As a mode to administer both of them with a staggered time, a mode in which an immunotherapy agent having an oral dosage form and one or more anti-cancer agents other than the immunotherapy agent to be parenterally administered are staggered respectively; parenteral An immunotherapy agent having a dosage form and a form in which one or more anticancer agents other than the immunotherapy agent to be orally administered are administered at staggered times, respectively.

 時間をずらして投与する場合、免疫療法剤に先立って免疫療法剤以外の1種以上の抗がん剤を投与しても、また免疫療法剤投与後に免疫療法剤以外の1種以上の抗がん剤を投与しても、いずれの順番でもよい。 When administered at staggered times, one or more anti-cancer agents other than the immunotherapeutic agent may be administered prior to the immunotherapeutic agent, and one or more anti-cancer agents other than the immunotherapeutic agent may be administered after the immunotherapy agent is administered. The drug may be administered in any order.

 具体的には、免疫療法剤と、免疫療法剤以外の1種以上の抗がん剤とを組み合わせた治療剤が経口投与剤である場合、免疫療法剤の投与量は、0.003~3000mg/kg/日の範囲から適宜設定することができる。また組み合わせる1種以上の抗がん剤の投与量も0.1~5000mg/kg/日、0.1~5000mg/kg/回、0.1~5000mg/m2/回又は10万~500万JRU/回から適宜設定することができる。また、投薬回数は1~8日間投与し、5日~9週間の休薬期間を設けることができる。 Specifically, when the therapeutic agent that is a combination of an immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent is an oral administration agent, the dose of the immunotherapeutic agent is 0.003 to 3000 mg. It can be appropriately set within the range of /kg/day. The dose of one or more anti-cancer agents to be combined is also 0.1 to 5000 mg/kg/day, 0.1 to 5000 mg/kg/dose, 0.1 to 5000 mg/m2/dose or 100,000 to 5 million JRU. It can be appropriately set from / times. In addition, the frequency of administration may be 1 to 8 days, and a drug holiday of 5 days to 9 weeks may be provided.

 免疫療法剤と、免疫療法剤以外の1種以上の抗がん剤とを組み合わせた治療剤が静脈内投与剤である場合、免疫療法剤の有効成分の有効血中濃度が0.1~3000μg/mL、より好ましくは1~1000μg/mLの範囲となるような投与量で、1日当たりを0.5~50mg/kgとなるように投与することがでる。また1種以上の抗がん剤の投与量も通常投与する量の10~100%、好ましくは10~95%の範囲において適宜設定することができる。 When the therapeutic agent in which the immunotherapeutic agent and one or more anticancer agents other than the immunotherapeutic agent are combined is an intravenously administered agent, the effective blood concentration of the active ingredient of the immunotherapeutic agent is 0.1 to 3000 μg. /ML, more preferably 1 to 1000 μg/mL, and the daily dose may be 0.5 to 50 mg/kg. Further, the dose of one or more anti-cancer agents can also be appropriately set within the range of 10 to 100%, preferably 10 to 95% of the normally administered dose.

 前記免疫療法剤以外の1種以上の抗がん剤は、腫瘍を治療することを目的とする薬剤である限り制限されない。例えば、前記他の抗がん剤として、アルキル化薬、代謝拮抗薬、抗腫瘍性抗生物質、微小血管阻害薬、ホルモン又はホルモン類似薬、白金製剤、トポイソメラーゼ阻害薬、サイトカイン、抗体薬、分子標的薬、ステロイド薬、非特異的免疫賦活薬、及びその他の抗がん剤の中から、対象とするがんに応じて適宜選択することができる。 The one or more anticancer agents other than the immunotherapeutic agents are not limited as long as they are agents intended to treat tumors. For example, other anticancer agents include alkylating agents, antimetabolites, antitumor antibiotics, microvascular inhibitors, hormones or hormone analogs, platinum preparations, topoisomerase inhibitors, cytokines, antibody drugs, molecular targets. It can be appropriately selected from a drug, a steroid drug, a non-specific immunostimulant, and other anti-cancer agents according to the target cancer.

 アルキル化薬としては、例えば、シクロホスファミド、イホスファミド、ブスルファン、メルファラン、ベンダムスチン塩酸塩、ニムスチン塩酸塩、ラニムスチン、カルムスチン、ストレプトゾシン、ダガルバジン、プロカルバジン塩酸塩、テモゾロミド等;代謝拮抗薬としては、メトトレキサート、ペメトレキセドナトリウム、プララトレキサート、フルオロウラシル、ドキシフルリジン、カペシタビン、テガフール、シタラビン、シタラビンオクホスファート水和物、エノシタビン、ゲムシタビン塩酸塩、メルカプトプリン水和物、フルダラビンリン酸エステル、ネララビン、ペントスタチン、クラドリビン、クロファラビン、フォロデシン塩酸塩、レボホリナートカルシウム、ホリナートカルシウム、ヒドロキシカルバミド、アナグレリド塩酸塩、クリサンタスパーゼ、L-アスパラギナーゼ、アザシチジン、トリフリルジン・チピラシル塩酸塩、ペメトレキセド等;抗腫瘍性抗生物質としては、マイトマイシンC、アクチノマイシンD、ブレオマイシン、ペプロマイシン硫酸塩、ジノスタチンスチラマー等;微小血管阻害薬としては、ビンクリスチン硫酸塩、ビンブラスチン硫酸塩、ビンデシン硫酸塩、ビノレルビン酒石酸塩、パクリタキセル、ドセタキセル水和物、カバジタキセル、エリブリンメシル酸塩等;ホルモン又はホルモン類似薬(ホルモン剤)としては、アナストロゾール、エキセメスタン、レトロゾール、タモキシフェンクエン酸塩、トレミフェンクエン酸塩、フルベストラント、フルタミド、ビカルタミド、エンザルタミド、クロルマジノン酢酸エステル、アビラテロン酢酸エステル、メドロキシプロゲステロン酢酸エステル、エストラムスチンリン酸エステルナトリウム水和物、ゴセレリン酢酸塩、リュープロレリン酢酸塩、デガレリクス酢酸塩等;白金製剤としては、シスプラチン、ミリプラチン水和物、カルボプラチン、ネダプラチン、オキサリプラチン等;トポイソメラーゼ阻害薬としては、イリノテカン塩酸塩水和物、及びノギテカン塩酸塩等のトポイソメラーゼI阻害薬、並びにドキソルビシン塩酸塩、ダウノルビシン塩酸塩、ピラルビシン、エピルビシン塩酸塩、イダルビシン塩酸塩、アクラルビシン塩酸塩、アムルビシン塩酸塩、ミトキサントロン塩酸塩、エトポシド、及びソブゾキサン等のトポイソメラーゼII阻害薬;サイトカインとしては、インターフェロンガンマ-1a、テセロイキン、セルモロイキン等;抗体薬としては、トラスツズマブ、リツキシマブ、ゲムツズマブオゾガマイシン、ベバシズマブ、セツキシマブ等;分子標的薬としては、セツキシマブ、バニツムマブ、トランスツズマブ、ペルツズマブ、ペバシズマブ、ラムシルマブ、リツキシマブ、オファツムマブ、ダラツムマブ、アレムツズマブ、エロツズマブ、トランスツズマブ エムタンシン、ゲムツズマブオジガマイシン、イノツズマブオゾガマイシン、イブリツモマブ、ゲフィチニブ、エルロチニブ塩酸塩、アファチニブマレイン酸塩、オチメルチニブメシル酸塩、イマチニブメシル酸塩、ボルテゾミブ、ボスチニブ、ボナチニブ、アキシチニブ、ブレンツキシマブベドチン、ソラフェニブトシル酸塩、スニチニブリンゴ酸塩、バゾパニプ塩酸塩、レゴラフェニブ、バンデタニブ、レンバチニブメシル酸塩、カルフィルゾミブ、イキサゾミブクエン酸塩、クチゾチニブ、アレクチニブ塩酸塩、セリチニブ、イブルチニブ、モガムリズマブ、ルキソリチニブリン酸塩、ベムラフェニブ、ダムラフェニブメシル酸塩、トラメチニブ、エベロリムス、テムシロリムス、シロリムス、アフリベルセプトベータ、パルボシクリブ、トレチノイン、タミバロテン、ベキサロテン、ボリノスタット、バノビノスタット乳酸塩、ロミデプシン、サリドマイド、ニロチニブ塩酸塩水和物、ポマリミド、ダサチニブ水和物、ラパチニブトシル酸塩水和物、エベロリムス、レナリドミド水和物、テムシロリムス、ボリノスタット、トレチノイン、及びタミバロテン等;ステロイド薬としては、デキサメタゾン等;非特異的免疫賦活薬としては、OK-432、乾燥BCG、かわらたけ多糖体製剤、レンチナン、ウベニメクス等;その他、トラベクテジン、アセグラトン、ポルフィマーナトリウム、タラポルフィンナトリウム、エタノール、三酸化ヒ素、タルク、デクスラゾキサン、塩化ストロンチウム、メトロニダゾール、塩化ラジウム223Ra、ヒトチロトビンアルファ等を例示することができる。 Examples of alkylating agents include cyclophosphamide, ifosfamide, busulfan, melphalan, bendamustine hydrochloride, nimustine hydrochloride, ranimustine, carmustine, streptozocin, dagalbazine, procarbazine hydrochloride, temozolomide; as antimetabolites, Methotrexate, pemetrexed sodium, pralatrexate, fluorouracil, doxyfluridine, capecitabine, tegafur, cytarabine, cytarabine ocfosfate hydrate, enocitabine, gemcitabine hydrochloride, mercaptopurine hydrate, fludarabine phosphate, nelarabine, pentostatin, pentostatin. , Clofarabine, forodecin hydrochloride, levofolinate calcium, holinate calcium, hydroxycarbamide, anagrelide hydrochloride, chrysantaspase, L-asparaginase, azacitidine, trifurildin tipiracil hydrochloride, pemetrexed, etc.; as an antitumor antibiotic, mitomycin C , Actinomycin D, bleomycin, peplomycin sulfate, dinostatin styramer, etc.; microvessel inhibitors include vincristine sulfate, vinblastine sulfate, vindesine sulfate, vinorelbine tartrate, paclitaxel, docetaxel hydrate, cabazitaxel, eribulin. Mesylate, etc.; Hormones or hormone analogs (hormonal agents) include anastrozole, exemestane, letrozole, tamoxifen citrate, toremifene citrate, fulvestrant, flutamide, bicalutamide, enzalutamide, chlormadinone acetate, Abiraterone acetate, medroxyprogesterone acetate, estramustine phosphate sodium hydrate, goserelin acetate, leuprorelin acetate, degarelix acetate, etc.; platinum preparations include cisplatin, miriplatin hydrate, carboplatin, Nedaplatin, oxaliplatin, etc.; topoisomerase inhibitors include topoisomerase I inhibitors such as irinotecan hydrochloride hydrate and nogitecan hydrochloride, and doxorubicin hydrochloride, daunorubicin hydrochloride, pirarubicin, epirubicin hydrochloride, idarubicin hydrochloride, aclarubicin hydrochloride. Topoisomerase II inhibitors such as salts, amrubicin hydrochloride, mitoxantrone hydrochloride, etoposide, and sobuzoxane; as cytokines, interferon gamma-1a , Teseleukin, sermoleukin, etc.; antibody drugs include trastuzumab, rituximab, gemtuzumab ozogamicin, bevacizumab, cetuximab, etc.; , Dalatumumab, alemtuzumab, erotuzumab, transtuzumab emtansine, gemtuzumab odigamicin, inotuzumab ozogamicin, ibritumomab, gefitinib, erlotinib hydrochloride, afatinib maleate, otimertinib mesilibate, imate , Bortezomib, bosutinib, bonatinib, axitinib, brentuximab vedotin, sorafenib tosylate, sunitinib malate, vazopanip hydrochloride, regorafenib, vandetanib, lenbatinib mesilate, carfilzomib, ixazomib citrate, , Alectinib hydrochloride, ceritinib, ibrutinib, mogamulizumab, ruxolitinib phosphate, vemurafenib, damrafenib mesylate, trametinib, everolimus, temsirolimus, sirolimus, aflibercepto, parvocyclib, tretinoin, tretinoin, tacitinoin, tretinoin, tacitinoin, tretinoin, tatinoline, tretinoin, tacitinoin Vanobinostat lactate, romidepsin, thalidomide, nilotinib hydrochloride hydrate, pomalimid, dasatinib hydrate, lapatinib tosylate hydrate, everolimus, lenalidomide hydrate, temsirolimus, vorinostat, tretinoin, and tamibarotene; as steroid drugs, Dexamethasone and the like; non-specific immunostimulants such as OK-432, dried BCG, Kawatake mushroom polysaccharide preparation, lentinan, ubenimex and the like; Examples thereof include talc, dexrazoxane, strontium chloride, metronidazole, radium chloride 223 Ra, human tyrotobin alfa and the like.

 悪性腫瘍を治療するための免疫療法剤と、免疫療法剤以外の1種以上の抗がん剤を組み合わせる場合、例えば、免疫チェックポイント阻害剤(例えば、抗PD1抗体を含む)と、白金製剤と、代謝拮抗剤(例えば、ペメトレキセド)等を挙げることができる。このような組み合わせは、非扁平上皮癌の治療に好ましい。また、別の例として、免疫チェックポイント阻害剤(例えば、抗PD1抗体を含む)と、白金製剤(例えば、カルボプラチン)と、微小血管阻害薬(例えば、パクリタキセル又はnab-パクリタキセル)との組み合わせを挙げることができる。このような組み合わせは、扁平上皮癌の治療に好ましい。また、別の例として、免疫チェックポイント阻害剤(例えば、抗PD-L1抗体を含む)と、白金製剤(例えば、カルボプラチン)と、微小血管阻害薬(例えば、パクリタキセル又はnab-パクリタキセル)と、分子標的治療薬(例えば、血管新生阻害薬である、ベバシズマブ等)との組み合わせを挙げることができる。このような組み合わせは、非扁平上皮癌の治療に好ましい。 When combining an immunotherapeutic agent for treating a malignant tumor and one or more anti-cancer agents other than the immunotherapeutic agent, for example, an immune checkpoint inhibitor (including, for example, an anti-PD1 antibody) and a platinum preparation , Antimetabolites (eg, pemetrexed), and the like. Such combinations are preferred for the treatment of non-squamous cell carcinoma. As another example, a combination of an immune checkpoint inhibitor (eg, including anti-PD1 antibody), a platinum agent (eg, carboplatin), and a microvascular inhibitor (eg, paclitaxel or nab-paclitaxel) can be mentioned. be able to. Such combinations are preferred for the treatment of squamous cell carcinoma. As another example, an immune checkpoint inhibitor (eg, including anti-PD-L1 antibody), a platinum agent (eg, carboplatin), a microvascular inhibitor (eg, paclitaxel or nab-paclitaxel), and a molecule A combination with a targeted therapeutic agent (for example, an angiogenesis inhibitor, bevacizumab, etc.) can be mentioned. Such combinations are preferred for the treatment of non-squamous cell carcinoma.

9.腫瘍細胞が傷害されているか否かを評価するための検査試薬及びキット
(1)エンゲージャーを含む検査試薬及びキット
 本開示のある実施形態は、エンゲージャーを含む、腫瘍細胞が傷害されているか否かを評価するための検査試薬81に関する。前記検査試薬81は、上記2.に記載の提示方法及び上記3.に記載の補助方法を実施するためにも使用することができる。図8Aに検査試薬81の構成を示す。図8Bにキット8の構成を示す。検査試薬81は、エンゲージャーを溶解するための緩衝液等を含んでいてもよい。また緩衝液は、エンゲージャーを安定化させるための還元剤(β-メルカプトエタノール、ジチオスレイトール等)、界面活性剤、アルブミン等を含んでいてもよい。また、アジ化ナトリウム等の防腐剤を含んでいても良い。エンゲージャーは、乾燥状態であっても、前記緩衝液に溶解されていてもよい。
9. Test Reagent and Kit for Evaluating Whether Tumor Cells are Damaged (1) Test Reagent and Kit Containing Engager In one embodiment of the present disclosure, whether tumor cells are damaged, which includes an engager, are disclosed. It relates to a test reagent 81 for evaluating whether or not. The test reagent 81 is the same as in 2. above. And the presentation method described in 3. above. It can also be used to carry out the auxiliary methods described in. FIG. 8A shows the structure of the test reagent 81. The structure of the kit 8 is shown in FIG. 8B. The test reagent 81 may include a buffer solution or the like for dissolving the engager. Further, the buffer solution may contain a reducing agent (β-mercaptoethanol, dithiothreitol, etc.) for stabilizing the engager, a surfactant, albumin and the like. It may also contain a preservative such as sodium azide. The engager may be in a dry state or may be dissolved in the buffer solution.

 本開示のある実施形態は、検査試薬81を含む、腫瘍細胞が傷害されているか否かを評価するためのキット8に関する。前記キット8は、上記2に記載の提示方法を実施するためにも使用することができる。本実施形態のキット8には、少なくとも検査試薬81と、前記検査試薬を使って行う測定方法を記載したプロトコール、又は前記プロトコールにアクセスするためのURL、Qコード等の情報を記載した添付書類82等を含む。 An embodiment of the present disclosure relates to a kit 8 including a test reagent 81 for evaluating whether tumor cells are injured. The kit 8 can also be used to carry out the presentation method described in 2 above. The kit 8 of the present embodiment includes at least a test reagent 81 and a protocol describing a measurement method performed using the test reagent, or an attached document 82 describing information such as a URL and a Q code for accessing the protocol. Including etc.

(2)抗インターフェロン-γ抗体を含む検査試薬及びキット
 本開示のある実施形態は、抗インターフェロン-γ抗体を含む、腫瘍細胞が傷害されているか否かを評価するための検査試薬91a及びキット9に関する。前記検査試薬91aは、上記2.に記載の提示方法及び上記3.に記載の補助方法を実施するためにも使用することができる。図8Cにキット9の構成を示す。キット9はELISAキットの例であるが、これに制限されない。キット9には、抗インターフェロン-γ抗体を含む検査試薬91aと、マイクロプレート92(検査試薬91aに含まれる抗インターフェロン抗体とはエピトープが異なる抗体が固定されていてもよい)と、抗インターフェロン-γ抗体を検出するための酵素標識抗体、又は酵素標識検出体(検査試薬91aに含まれる抗インターフェロン抗体にビオチンが標識されている場合には、酵素標識アビジン、又は酵素標識ストレプトアビジン)等を含む検出試薬91bと、キット9を使って行う測定方法を記載したプロトコール、又は前記プロトコールにアクセスするためのURL、Qコード等の情報を記載した添付書類93等を含む。なお、図示しないが、キット9には、前記酵素と反応する基質が含まれていてもよい。
(2) Test Reagent and Kit Containing Anti-Interferon-γ Antibody According to an embodiment of the present disclosure, a test reagent 91a and a kit 9 for evaluating whether or not a tumor cell is damaged, the anti-interferon-γ antibody is included. Regarding The inspection reagent 91a is the same as in 2. above. And the presentation method described in 3. above. It can also be used to carry out the auxiliary methods described in. The structure of the kit 9 is shown in FIG. 8C. Kit 9 is an example of an ELISA kit, but is not limited to this. The kit 9 includes a test reagent 91a containing an anti-interferon-γ antibody, a microplate 92 (an antibody having an epitope different from that of the anti-interferon antibody contained in the test reagent 91a may be fixed), and an anti-interferon-γ. Detection including an enzyme-labeled antibody for detecting an antibody, or an enzyme-labeled detector (enzyme-labeled avidin, or enzyme-labeled streptavidin when the anti-interferon antibody contained in the test reagent 91a is labeled with biotin) It includes a reagent 91b and a protocol that describes a measurement method using the kit 9, or an attachment 93 that describes information such as a URL and a Q code for accessing the protocol. Although not shown, the kit 9 may include a substrate that reacts with the enzyme.

 検査試薬91aに含まれる抗インターフェロン-γ抗体は、上記1.に記載したものを使用することができる。検査試薬91aには、少なくとも抗インターフェロン-γ抗体が1種以上含まれていればよい。抗インターフェロン-γ抗体がポリクローナル抗体である場合には、1種の抗原で免役して得られたポリクローナル抗体であってもよく、また2種以上の抗原で並行して同一個体に免役して得られたポリクローナル抗体であってもよい。さらに、2種以上の抗原をそれぞれ別の動物に接種して得られたそれぞれのポリクローナル抗体を混合してもよい。抗インターフェロン-γ抗体がモノクローナル抗体である場合には、1種のハイブリドーマから産生されるモノクローナル抗体であってもよいが、2種以上のハイブリドーマから産生されたモノクローナル抗体であって、それぞれのモノクローナル抗体が同一又は異なるエピトープを認識する複数のモノクローナル抗体が2種以上含まれていてもよい。また、ポリクローナル抗体とモノクローナル抗体を1種以上ずつ混合して含んでいてもよい。 The anti-interferon-γ antibody contained in the test reagent 91a is 1. Those described in can be used. The test reagent 91a may contain at least one anti-interferon-γ antibody. When the anti-interferon-γ antibody is a polyclonal antibody, it may be a polyclonal antibody obtained by immunizing with one kind of antigen, or may be obtained by immunizing the same individual with two or more kinds of antigens in parallel. It may be a prepared polyclonal antibody. Furthermore, the respective polyclonal antibodies obtained by inoculating two or more antigens into different animals may be mixed. When the anti-interferon-γ antibody is a monoclonal antibody, it may be a monoclonal antibody produced by one kind of hybridoma, but it is a monoclonal antibody produced by two or more kinds of hybridoma, and each monoclonal antibody Two or more kinds of monoclonal antibodies that recognize the same or different epitopes may be contained. In addition, one or more polyclonal antibodies and one or more monoclonal antibodies may be mixed and included.

 当該検査試薬91aに含まれる抗インターフェロン-γ抗体の形態は、特に制限されず、抗インターフェロン-γ抗体を含む抗血清若しくは腹水等の乾燥状態又は液体状態であってもよい。また、抗インターフェロン-γ抗体の形態は、精製抗インターフェロン-γ抗体、抗インターフェロン-γ抗体を含む免疫グロブリン画分若しくは抗インターフェロン-γ抗体を含むIgG画分の乾燥状態又は水溶液であってもよい。 The form of the anti-interferon-γ antibody contained in the test reagent 91a is not particularly limited, and may be a dry state or a liquid state of antiserum or ascites fluid containing the anti-interferon-γ antibody. Further, the form of the anti-interferon-γ antibody may be a purified anti-interferon-γ antibody, an immunoglobulin fraction containing the anti-interferon-γ antibody or an IgG fraction containing the anti-interferon-γ antibody in a dry state or an aqueous solution. ..

 前記形態が、抗インターフェロン-γ抗体を含む抗血清若しくは腹水の乾燥状態又は液体状態である場合、さらにβ-メルカプトエタノール、DTT等の安定化剤;アルブミン等の保護剤;ポリオキシエチレン(20)ソルビタンモノラウレート、ポリオキシエチレン(10)オクチルフェニルエーテル等の界面活性剤、アジ化ナトリウム等の防腐剤等の少なくとも一つを含んでいてもよい。また、抗インターフェロン-γ抗体の形態が、精製抗インターフェロン-γ抗体、抗インターフェロン-γ抗体を含む免疫グロブリン画分若しくは抗インターフェロン-γ抗体を含むIgG画分の乾燥状態又は水溶液である場合、さらに、リン酸緩衝液等のバッファー成分;β-メルカプトエタノール、DTT等の安定化剤;アルブミン等の保護剤;塩化ナトリウム等の塩;ポリオキシエチレン(20)ソルビタンモノラウレート、ポリオキシエチレン(10)オクチルフェニルエーテル等の界面活性剤等、アジ化ナトリウム等の防腐剤の少なくとも一つを含んでいてもよい。 When the form is a dry state or a liquid state of antiserum or ascites containing anti-interferon-γ antibody, a stabilizer such as β-mercaptoethanol or DTT; a protective agent such as albumin; polyoxyethylene (20) It may contain at least one of a surfactant such as sorbitan monolaurate and polyoxyethylene (10) octyl phenyl ether, and a preservative such as sodium azide. Further, when the form of the anti-interferon-γ antibody is a purified anti-interferon-γ antibody, an immunoglobulin fraction containing the anti-interferon-γ antibody or an IgG fraction containing the anti-interferon-γ antibody in a dry state or an aqueous solution, , Buffer components such as phosphate buffer, stabilizers such as β-mercaptoethanol and DTT, protective agents such as albumin, salts such as sodium chloride, polyoxyethylene (20) sorbitan monolaurate, polyoxyethylene (10 ) It may contain at least one of preservatives such as sodium azide and the like, such as surfactants such as octylphenyl ether and the like.

 本実施形態において、検査試薬91aが単体で供給される場合には、当該抗インターフェロン-γ抗体は未標識であっても、上述の標識物質で標識されていてもよいが、上記1.で述べた標識物質で標識されていることが好ましい。また、抗原捕捉用の抗インターフェロン-γ抗体が、固相表面等に固定化されたマイクロプレート92が検査試薬として提供されてもよい。 In the present embodiment, when the test reagent 91a is supplied alone, the anti-interferon-γ antibody may be unlabeled or labeled with the above-mentioned labeling substance. It is preferably labeled with the labeling substance described in 1 above. Alternatively, a microplate 92 having an anti-interferon-γ antibody for capturing an antigen immobilized on a solid phase surface or the like may be provided as a test reagent.

 以下に実施例を示して本明細書の開示内容を具体的に説明するが、本発明は実施例に限定し解釈されるものではない。 The present invention will be specifically described below with reference to examples, but the present invention is not construed as being limited to the examples.

I.実施例1:末梢血単核細胞を用いた腫瘍細胞傷害活性パーセントの測定
1.方法
(1)U251細胞株を、1×105個/mLとなるように10%FBSを添加したRPMI1640培地(10%FBS加RPMI1640培地)に懸濁した細胞懸濁液を調製した。前記細胞懸濁液を100μL/ウェルとなるように96ウェルプレートに播種し、5%炭酸ガスが存在する湿式インキュベータ内で、24時間、37℃でインキュベーションした。
I. Example 1: Measurement of percent tumor cytotoxic activity using peripheral blood mononuclear cells 1. Method (1) A cell suspension was prepared by suspending the U251 cell line in RPMI1640 medium (RPMI1640 medium containing 10% FBS) supplemented with 10% FBS at 1×10 5 cells/mL. The cell suspension was seeded on a 96-well plate at 100 μL/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.

(2)肺癌患者から末梢血(ヘパリン採血)を採取し、LymphoprepTM (Alere Technologies AS)を使って、添付のプロトコールにしたがって末梢血単核細胞を含む細胞群(「PBMC」とする)を回収した。前記PBMCの回収は、後述する(3)の工程の直前に行った。 (2) Peripheral blood (heparin blood sampling) was collected from a lung cancer patient, and a cell group containing peripheral blood mononuclear cells (referred to as "PBMC") was collected using Lymphoprep (Alere Technologies AS) according to the attached protocol. did. The PBMCs were collected immediately before the step (3) described later.

(3)(1)でインキュベーションが終わった96ウェルプレートに、1ウェルあたり、前記PBMCの細胞数が5×104個、EphA2-CD3 BiTE(登録商標)が終濃度で100 ng/mLとなるように添加した。コントロールとして、(1)でインキュベーションが終わった96ウェルプレート内に前記PBMCのみを添加し、EphA2-CD3 BiTE(登録商標)を添加しないウェルを準備した。前記PBMCとEphA2-CD3 BiTE(登録商標)を添加した後の1ウェルあたりの10%FBS加RPMI培地の量は、総量が200μLとなるようにした。PBMCとEphA2-CD3 BiTE(登録商標)を添加したあと、48時間インキュベーションした。 (3) The number of cells of the PBMC is 5×10 4 , and the final concentration of EphA2-CD3 BiTE (registered trademark) is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added. As a control, a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1). After adding the PBMC and EphA2-CD3 BiTE (registered trademark), the total volume of RPMI medium containing 10% FBS was 200 μL per well. After adding PBMC and EphA2-CD3 BiTE (registered trademark), the mixture was incubated for 48 hours.

(4)インキュベーション終了後、10%FBS加RPMI培地で各ウェルを4回洗浄した。洗浄後培地が除去された各ウェルに、100μLの10%FBS加RPMI培地と20μLのCellTiter 96(登録商標) AQueous One Solution Reagent (MTS reagent:プロメガ)を添加した。その後、96ウェルマイクロプレートを5%炭酸ガスが存在する湿式インキュベータ内で、20分、37℃でインキュベーションした。 (4) After completion of the incubation, each well was washed 4 times with RPMI medium containing 10% FBS. After washing, 100 μL of RPMI medium containing 10% FBS and 20 μL of CellTiter 96 (registered trademark) A Queous One Solution Reagent (MTS reagent: Promega) were added to each well from which the medium had been removed. Then, the 96-well microplate was incubated at 37° C. for 20 minutes in a wet incubator in the presence of 5% carbon dioxide.

(5)(4)でインキュベーションが終了した96ウェルマイクロプレートについて、マイクロプレートリーダを使って各ウェルの490 nmにおける吸光度を測定した。続いて、下式にしたがって、腫瘍細胞傷害活性パーセントを算出した。
  U251細胞株とPBMCを含むウェルの吸光度=A
  U251細胞株とPBMCとEphA2-CD3 BiTEを含むウェルの吸光度=B
(5) Regarding the 96-well microplate in which the incubation was completed in (4), the absorbance at 490 nm of each well was measured using a microplate reader. Then, the percent tumor cytotoxic activity was calculated according to the following formula.
Absorbance of well containing U251 cell line and PBMC = A
Absorbance of well containing U251 cell line, PBMC and EphA2-CD3 BiTE = B

  腫瘍細胞傷害活性パーセント (%)=[(A-B)/A]×100 Tumor cytotoxic activity percentage (%) = [(A-B)/A] x 100

2.結果
 図9に各患者のPBMCの腫瘍細胞傷害活性パーセントを示す。BiTEとしてCD3に結合するものを使用していることから、このPBMCの腫瘍細胞傷害活性は、末梢血T細胞の腫瘍細胞傷害活性であると考えられる。図9からわかるように、肺癌患者の末梢血T細胞の腫瘍細胞傷害活性は、患者によって異なっており、腫瘍細胞傷害活性が数%しかない患者も存在する一方で、80%以上と高い値を示す患者も存在した。このことから、各患者の末梢血T細胞の腫瘍細胞傷害活性には、個人差があることがわかった。
2. Results FIG. 9 shows the percent tumor cytotoxic activity of PBMC in each patient. Since BiTE that binds to CD3 is used, the tumor cytotoxic activity of this PBMC is considered to be the tumor cytotoxic activity of peripheral blood T cells. As can be seen from FIG. 9, the tumor cytotoxic activity of peripheral blood T cells in lung cancer patients varies depending on the patient, and while some patients have tumor cytotoxic activity of only a few %, a high value of 80% or more is obtained. There were also patients presenting. From this, it was found that the tumor cytotoxic activity of peripheral blood T cells of each patient varies among individuals.

II.実験例1:肺癌組織内のT細胞の腫瘍細胞傷害活性
 肺癌組織内のT細胞の腫瘍細胞傷害活性パーセントは、以下の方法にしたがって肺癌組織から細胞を回収し、その細胞群の細胞傷害活性として測定した。
 癌組織を6 cm Dishに移して細断した後、組織攪拌溶液(HBSS(-) + 2% FBS + 10mM HEPESにTumor Dissociation Kit (Miltenyi Biotec)を添加)に入れてgentleMACSTM Dissociator (Miltenyi Biotec)で攪拌後、37℃で維持したインキュベータ内で30分回転させながらインキュベーションした。その後、細胞を含む攪拌液を70 μmメッシュを通して、組織残渣を取り除き、濾液を600xg、10分遠心して沈殿物を回収した。細胞を含む沈殿物にBD Pharm lyse (BD Biosciences)を添加して2分間静置した。静置後の細胞を含む溶解液にHBSS Bufferを加えて600xg、10分遠心し、再度沈殿物を回収した。再回収された沈殿物に30% パーコール液を加えて12000xg、30秒遠心し、再度沈殿物を回収した。再回収された沈殿物をHBSS Bufferで洗浄し、12000xg、30秒遠心して組織内の細胞を回収した。回収された細胞の腫瘍細胞傷害活性パーセントは、上記I.と同様の方法で測定した。BiTEとしてCD3に結合するものを使用していることから、この肺癌組織内から回収された細胞の腫瘍細胞傷害活性は、肺癌組織内T細胞の腫瘍細胞傷害活性であると考えられる。
II. Experimental Example 1: Tumor cytotoxic activity of T cells in lung cancer tissue The percent tumor cytotoxic activity of T cells in lung cancer tissue was determined by collecting cells from lung cancer tissue according to the following method and determining the cytotoxic activity of the cell group. It was measured.
The cancer tissue was transferred to a 6 cm Dish and shredded, then placed in a tissue agitation solution (HBSS(-) + 2% FBS + 10 mM HEPES with Tumor Dissociation Kit (Miltenyi Biotec) added) and gentleMACS TM Dissociator (Miltenyi Biotec) After stirring at 37° C., the cells were incubated for 30 minutes while rotating in an incubator maintained at 37° C. Then, the stirred solution containing cells was passed through a 70 μm mesh to remove the tissue residue, and the filtrate was centrifuged at 600×g for 10 minutes to recover the precipitate. BD Pharm lyse (BD Biosciences) was added to the cell-containing pellet and left standing for 2 minutes. HBSS Buffer was added to the lysate containing cells after standing, and the mixture was centrifuged at 600 xg for 10 minutes, and the precipitate was collected again. A 30% Percoll solution was added to the recollected precipitate, and the mixture was centrifuged at 12000 xg for 30 seconds to collect the precipitate again. The recollected precipitate was washed with HBSS Buffer and centrifuged at 12000 xg for 30 seconds to collect cells in the tissue. The percent tumor cytotoxic activity of the recovered cells was determined according to I. It measured by the method similar to. Since BiTE that binds to CD3 is used, the tumor cytotoxic activity of the cells recovered from the lung cancer tissue is considered to be the tumor cytotoxic activity of T cells in the lung cancer tissue.

III.実施例2:T細胞レセプターレパトア解析
 同一患者において腫瘍内と末梢血に共通するT細胞クローンが存在するか否かを確認するため、T細胞レセプター(TCR)のレパトア解析を行った。
 肺癌患者末梢血PBMC及び肺癌組織内の細胞からFACSAria(Becton Dickinson)を用いてCD8陽性T細胞を採取した。採取したCD8陽性T細胞からRNAを抽出し、Adaptor-Ligation PCR法を用いてTCR遺伝子を増幅した後に、次世代シーケンサーを用いて塩基配列を決定した。TCRレパトア解析はRepertoire Genesis株式会社(http://www.repertoire.co.jp/detailed_repertoire.html)に外注した。
 レパトア解析は、TCRαとTCRβそれぞれについて行った。
 患者1~4について、PBMCと肺癌組織内T細胞について出現頻度の高い上位30クローンのレパートリーを比較した。
III. Example 2: T cell receptor repertoire analysis In order to confirm whether or not a T cell clone common to tumor and peripheral blood exists in the same patient, repertoire analysis of T cell receptor (TCR) was performed.
CD8-positive T cells were collected from cells in peripheral blood PBMCs of lung cancer patients and lung cancer tissues using FACSAria (Becton Dickinson). RNA was extracted from the collected CD8-positive T cells, the TCR gene was amplified using the Adapter-Ligation PCR method, and then the nucleotide sequence was determined using a next-generation sequencer. The TCR repertoire analysis was outsourced to Repertoire Genesis Co., Ltd. (http://www.repertoire.co.jp/detailed_repertoire.html).
The repertoire analysis was performed for each of TCRα and TCRβ.
For patients 1-4, we compared the repertoires of the top 30 most frequently occurring clones of PBMC and T cells in lung cancer tissues.

 患者1は及び患者2は、肺癌組織内CD8陽性T細胞の腫瘍細胞傷害活性パーセントが高かった例であり、患者3は、前記腫瘍細胞傷害活性パーセントが中程度であった例であり、患者4は前記腫瘍細胞傷害活性パーセントが低かった例である。
 結果を図10に示す。
Patient 1 and patient 2 were cases in which the percent tumor cytotoxic activity of CD8-positive T cells in lung cancer tissue was high, and patient 3 was an example in which the percent tumor cytotoxic activity was moderate, and patient 4 Is an example where the tumor cell cytotoxic activity percentage was low.
The results are shown in Fig. 10.

 出現頻度の高い上位30クローンのレパートリーを比較した結果、肺癌組織内T細胞の腫瘍細胞傷害活性パーセントが高かった又は中程度であった患者1~3は、末梢血と肺癌組織内CD8陽性T細胞で複数種の共通クローンが認められた。これに対して、肺癌組織内T細胞の腫瘍細胞傷害活性パーセントが低かった患者4では、末梢血と肺癌組織内CD8陽性T細胞で共通するクローンが1クローンのみ確認された。このことから、肺癌組織内T細胞の腫瘍細胞傷害活性が高い程、肺癌組織内で腫瘍細胞の抗原特異的に増殖した細胞障害性T細胞が末梢血にも多く出現することが示唆された。 As a result of comparing the repertoires of the 30 most frequently occurring clones, patients 1 to 3 who had a high or moderate percent tumor cytotoxic activity of T cells in lung cancer tissues were found to have peripheral blood and lung cancer tissue CD8-positive T cells. Multiple common clones were found in. On the other hand, in patient 4 in which the percentage of tumor cell cytotoxicity of T cells in lung cancer tissues was low, only one clone was confirmed that was common to peripheral blood and CD8-positive T cells in lung cancer tissues. From this, it was suggested that the higher the tumor cytotoxic activity of the T cells in the lung cancer tissue, the more the cytotoxic T cells that proliferated in the lung cancer tissue in an antigen-specific manner appeared in the peripheral blood.

IV.実施例3:肺癌組織内T細胞の腫瘍細胞傷害活性と末梢血T細胞の腫瘍細胞傷害活性の相関
 次に、肺癌組織内T細胞の腫瘍細胞傷害活性パーセントとPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントの相関を求めた。結果を図11に示す。相関係数Rとp値は統計ソフトJMPを用いて求めた。p値は、相関の有無の検定値を示す。
IV. Example 3: Correlation between tumor cytotoxic activity of T cells in lung cancer tissue and tumor cytotoxic activity of peripheral blood T cells Next, the percentage of tumor cytotoxic activity of T cells in lung cancer tissue and the peripheral blood T cells derived from PBMC were examined. The correlation of percent tumor cytotoxic activity was determined. The results are shown in Fig. 11. The correlation coefficient R 2 and the p value were obtained using statistical software JMP. The p-value shows a test value for the presence or absence of correlation.

 また、比較例として、患者から採取したPBMCとU251細胞株にEphA2-CD3 BiTEを加えて2日間培養した培養上清について、抗腫瘍作用を示すインターフェロンγ(IFNγ)の濃度を求め、この結果と肺癌組織内T細胞の腫瘍細胞傷害活性パーセントの相関を求めた。IFNγの濃度は、Bio-Plex Proヒトサイトカイン GI 27-plexパネル(Bio-Rad)を使用して測定した。 In addition, as a comparative example, the concentration of interferon γ (IFNγ) showing an antitumor effect was determined for the culture supernatant obtained by adding EphA2-CD3BiTE to PBMC and U251 cell lines collected from patients for 2 days, and The correlation of percent tumor cytotoxic activity of T cells in lung cancer tissue was determined. The IFNγ concentration was measured using the Bio-Plex Pro human cytokine GI27-plex panel (Bio-Rad).

 その結果、肺癌組織内T細胞の腫瘍細胞傷害活性パーセントと末梢血T細胞の腫瘍細胞傷害活性パーセントの相関はR2=0.36であり、p=0.009となり、末梢血T細胞の細胞傷害活性は、肺癌組織内T細胞の腫瘍細胞傷害活性を反映すると考えられた。これに対して、IFNγは、R2=0.22であり、p=0.048となり、IFNγも肺癌組織内T細胞の腫瘍細胞傷害活性と相関することが示された。しかし、IFNγに比べ末梢血T細胞の腫瘍細胞傷害活性の方が、肺癌組織内T細胞の腫瘍細胞傷害活性とより強く相関することが示された。 As a result, the correlation between the percent tumor cytotoxic activity of T cells in lung cancer tissue and the percent tumor cytotoxic activity of peripheral blood T cells was R 2 =0.36, p=0.009, and the cytotoxic activity of peripheral blood T cells was It was considered to reflect the tumor cytotoxic activity of T cells in lung cancer tissues. In contrast, IFNγ was R 2 =0.22 and p=0.048, indicating that IFNγ also correlates with the tumor cytotoxic activity of T cells in lung cancer tissues. However, it was shown that the tumor cytotoxic activity of peripheral blood T cells was more strongly correlated with the tumor cytotoxic activity of T cells in lung cancer tissues than that of IFNγ.

 以上の結果から、末梢血T細胞の腫瘍細胞傷害活性を測定することにより、腫瘍組織内のT細胞の腫瘍細胞傷害活性を予測することができ、ひいては、末梢血中のPBMCの腫瘍細胞傷害活性を測定することにより、腫瘍免疫療法の効果を予測できると考えられた。 From the above results, the tumor cytotoxic activity of T cells in tumor tissue can be predicted by measuring the tumor cytotoxic activity of peripheral blood T cells, which in turn leads to the tumor cytotoxic activity of PBMC in peripheral blood. It was considered that the effect of tumor immunotherapy could be predicted by measuring

V.実施例4:ニボルマブの効果と末梢血T細胞の腫瘍細胞傷害活性との相関
1.方法
 18人の患者について、実施例1にしたがって測定したPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントと、ニボルマブの効果とを比較した。ニボルマブの効果は以下の方法により評価した。実施例1に記載の方法にしたがって、U251細胞株を96ウェルプレートに播種し、5%炭酸ガスが存在する湿式インキュベータ内で、24時間、37℃でインキュベーションした。実験例1に記載の方法にしたがって回収した各患者の肺癌組織内の細胞(細胞数は5×104個/ウェル)と、1μg/mlのニボルマブ(小野薬品工業株式会社より分与を受けた)とを100 ng/mlのEphA2-CD3 BiTE(登録商標)ともに各ウェルに添加した。陰性コントロールとしてニボルマブに替えて1μg/mlのヒトIgG4(アブカム)を100 ng/mlのEphA2-CD3 BiTE(登録商標)ともに培地に添加した。実施例1に記載の方法にしたがって、CellTiter 96(登録商標) AQueous One Solution Reagent (MTS reagent:プロメガ)を用いて吸光度を測定し、下式にしたがって腫瘍細胞傷害活性パーセントを算出した。
V. Example 4: Correlation between the effect of nivolumab and tumor cytotoxic activity of peripheral blood T cells 1. Method The effect of nivolumab was compared with the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC measured according to Example 1 in 18 patients. The effect of nivolumab was evaluated by the following method. According to the method described in Example 1, the U251 cell line was seeded on a 96-well plate and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas. Cells in the lung cancer tissue of each patient collected according to the method described in Experimental Example 1 (cell number is 5×10 4 cells/well) and 1 μg/ml nivolumab (distributed by Ono Pharmaceutical Co., Ltd.) And 100 ng/ml of EphA2-CD3 BiTE (registered trademark) were added to each well. As a negative control, 1 μg/ml of human IgG4 (abcam) was added to the medium together with 100 ng/ml of EphA2-CD3 BiTE (registered trademark) in place of nivolumab. According to the method described in Example 1, the absorbance was measured using CellTiter 96 (registered trademark) AQueous One Solution Reagent (MTS reagent: Promega), and the percent tumor cytotoxic activity was calculated according to the following formula.

  U251細胞株と肺癌組織内の細胞と陰性コントロールとEphA2-CD3 BiTEを含むウェルの吸光度=A
  U251細胞株と肺癌組織内の細胞とニボルマブとEphA2-CD3 BiTEを含むウェルの吸光度=B
Absorbance of well containing U251 cell line, cells in lung cancer tissue, negative control and EphA2-CD3 BiTE = A
Absorbance of wells containing U251 cell line, cells in lung cancer tissue, nivolumab and EphA2-CD3 BiTE = B

  腫瘍細胞傷害活性パーセント (%) =[(A-B)/A]×100
 相関係数Rとp値は統計ソフトJMPを用いて求めた。p値は、相関の有無の検定値を示す。
Tumor cytotoxic activity percentage (%) = [(A-B)/A] x 100
The correlation coefficient R 2 and the p value were obtained using statistical software JMP. The p-value shows a test value for the presence or absence of correlation.

2.結果
 ニボルマブの効果と末梢血T細胞の腫瘍細胞傷害活性パーセントの相関を図12Aに示す。R2=0.27、p=0.028であり、ニボルマブの効果とPBMCの腫瘍細胞傷害活性パーセントが相関することが示された。また、患者を末梢血T細胞の腫瘍細胞傷害活性パーセントが高い方から9名と、残りの9名との2群に分け、ROC曲線から暫定的なカットオフ値を求めたところ、28%となった。この2群間で、ニボルマブの効果について有意差検定を行ったところ、p=0.0005となり、末梢血T細胞の腫瘍細胞傷害活性パーセントの高い群では有意にニボルマブの効果が高かった(図12B)。
2. Results The correlation between the effect of nivolumab and the percent tumor cytotoxic activity of peripheral blood T cells is shown in Figure 12A. R 2 =0.27, p=0.028, indicating that the effect of nivolumab correlates with the percent tumor cytotoxic activity of PBMC. In addition, patients were divided into two groups, 9 from the highest tumor cytotoxic activity of peripheral blood T cells, and the remaining 9 and the tentative cutoff value was calculated from the ROC curve to be 28%. became. When a significant difference test was performed on the effect of nivolumab between the two groups, p=0.005, and the effect of nivolumab was significantly high in the group having a high percent tumor cytotoxic activity of peripheral blood T cells (FIG. 12B).

 このことから、PBMCを用いて測定される末梢血T細胞の腫瘍細胞傷害活性は腫瘍免疫療法の効果との相関が高いことが示された。また、末梢血T細胞の腫瘍細胞傷害活性は腫瘍免疫療法の効果の評価マーカーとして使用できることが示された。 From this, it was shown that the tumor cytotoxic activity of peripheral blood T cells measured using PBMC has a high correlation with the effect of tumor immunotherapy. Further, it was shown that the tumor cytotoxic activity of peripheral blood T cells can be used as a marker for evaluating the effect of tumor immunotherapy.

VI.実施例5:末梢血T細胞の腫瘍細胞傷害活性と腫瘍免疫療法の効果との相関
 次に、肺癌患者のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントと、腫瘍免疫療法の効果との相関を検討した。患者から採取したPBMCについて実施例1にしたがって腫瘍細胞傷害活性パーセントを測定し、腫瘍細胞傷害活性パーセントが25%以上の患者3名を抽出した。前記3名の患者において、抗PD-1抗体を用いた腫瘍免疫療法により腫瘍の縮小が認められ、その治療継続期間は140日以上であり、長期間の治療が可能であった。このように腫瘍の縮小が認められ、治療を継続できたことは、腫瘍免疫療法が有効であることを意味する。
VI. Example 5 Correlation between Tumor Cytotoxic Activity of Peripheral Blood T Cells and Effect of Tumor Immunotherapy Next, the percentage of tumor cytotoxic activity of peripheral blood T cells derived from PBMC of lung cancer patients and the effect of tumor immunotherapy were compared. The correlation of The percent tumor cytotoxic activity of PBMC collected from the patients was measured according to Example 1, and three patients having a percent tumor cytotoxic activity of 25% or more were extracted. In the above-mentioned 3 patients, tumor shrinkage was observed by tumor immunotherapy using anti-PD-1 antibody, and the duration of treatment was 140 days or longer, which enabled long-term treatment. The reduction of the tumor and the ability to continue the treatment in this way indicate that tumor immunotherapy is effective.

 現在、腫瘍免疫療法を適用するか否かを判断するにあたり、腫瘍組織におけるPD-L1発現細胞の陽性率が1つの指標とされている。前記3名の患者について、腫瘍組織におけるPD-L1発現肺癌細胞の陽性率を計測したところ、2名の患者については、陽性率が50%以上であったが、1名の患者は陽性率が2%と非常に低値であった。PD-L1発現細胞の陽性率は、PD-L1タンパク質の免疫染色(免疫抗体法)を行うことにより評価されている。PD-L1発現細胞が2%しかない患者はPD-L1タンパク質の免疫染色では、腫瘍免疫療法が有効な患者であるとは予測することができない。 Currently, the positive rate of PD-L1-expressing cells in tumor tissue is used as an index when determining whether to apply tumor immunotherapy. When the positive rate of PD-L1-expressing lung cancer cells in the tumor tissue was measured for the 3 patients, the positive rate was 50% or more for the 2 patients, but the positive rate for 1 patient was It was a very low value of 2%. The positive rate of PD-L1 expressing cells is evaluated by performing immunostaining (immunoantibody method) of PD-L1 protein. Patients with only 2% PD-L1 expressing cells cannot be predicted by PD-L1 protein immunostaining to be effective for tumor immunotherapy.

 一方で、前記PD-L1発現細胞が2%しかない患者のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性を測定することにより、前記患者の末梢血T細胞が腫瘍細胞を傷害すると決定することができ、実際にそのような患者において腫瘍免疫療法の効果が認められた。従って、本開示における腫瘍免疫療法の効果を予測する方法によれば、PD-L1発現細胞の陽性率が低い患者についても、腫瘍免疫療法の適用できるか否かの予測があると考えられた。さらには、腫瘍免疫療法が有効な患者をPD-L1タンパク質の免疫染色よりも高い検出率で検出できると考えられた。 On the other hand, by measuring the tumor cytotoxic activity of peripheral blood T cells derived from PBMC of a patient whose PD-L1 expressing cells are only 2%, it is determined that the peripheral blood T cells of the patient injure the tumor cells. In fact, the effect of tumor immunotherapy was observed in such patients. Therefore, according to the method of predicting the effect of tumor immunotherapy according to the present disclosure, it is considered that there is prediction of whether tumor immunotherapy can be applied to patients with a low PD-L1 expressing cell positive rate. Furthermore, it was considered that the tumor immunotherapy-effective patients could be detected with a higher detection rate than the PD-L1 protein immunostaining.

VII.実施例6:腫瘍細胞傷害活性とPD-L1発現細胞の陽性率との治療効果の予測精度の比較
1.方法
 30人の非小細胞肺癌患者について、ペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントを測定し、その後の治療効果、無増悪生存期間及び治療中断の必要があり、かつ投与再開が不可能となる有害事象(重篤な有害事象)の出現との関係を調べた。PBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントは以下の方法により評価した。現在、腫瘍免疫療法を適用するか否かを判断するにあたり、腫瘍組織におけるPD-L1発現細胞の陽性率(Tumor Proportion Score : TPS)が1つの指標とされており、これを比較対象とした。
VII. Example 6: Comparison of prediction accuracy of therapeutic effect between tumor cytotoxic activity and positive rate of PD-L1 expressing cells 1. METHODS: Peripheral blood T cells derived from PBMCs prior to treatment with pembrolizumab or nivolumab were assayed for percent tumor cytotoxic activity in 30 patients with non-small cell lung cancer, followed by treatment efficacy, progression-free survival and the need for treatment discontinuation. , And the relationship with the occurrence of adverse events (serious adverse events) that made it impossible to resume administration. Percentage of tumor cytotoxic activity of peripheral blood T cells derived from PBMC was evaluated by the following method. Currently, in determining whether to apply tumor immunotherapy, the positive rate of PD-L1 expressing cells (Tumor Proportion Score: TPS) in tumor tissues is used as one index, and this was used as a comparison target.

末梢血単核細胞を用いた腫瘍細胞傷害活性パーセントの測定
(1)U251細胞株を、1×105個/mLとなるように10%FBSを添加したRPMI1640培地(10%FBS加RPMI1640培地)に懸濁した細胞懸濁液を調製した。前記細胞懸濁液を100μL/ウェルとなるように96ウェルプレートに播種し、5%炭酸ガスが存在する湿式インキュベータ内で、24時間、37℃でインキュベーションした。
(2)肺癌患者から末梢血(ヘパリン採血)を採取し、LymphoprepTM (Alere Technologies AS)を使って、添付のプロトコールにしたがって末梢血単核細胞を含む細胞群(「PBMC」とする)を回収した。前記PBMCの回収は、後述する(3)の工程の直前に行った。
(3)(1)でインキュベーションが終わった96ウェルプレートに、1ウェルあたり、前記PBMCの細胞数が5×104個、EphA2-CD3 BiTE(登録商標)が終濃度で100 ng/mLとなるように添加した。コントロールとして、(1)でインキュベーションが終わった96ウェルプレート内に前記PBMCのみを添加し、EphA2-CD3 BiTE(登録商標)を添加しないウェルを準備した。前記PBMCとEphA2-CD3 BiTE(登録商標)を添加した後の1ウェルあたりの10%FBS加RPMI培地の量は、総量が200μLとなるようにした。PBMCとEphA2-CD3 BiTE(登録商標)を添加したあと、48時間インキュベーションした。
(4)インキュベーション終了後、10%FBS加RPMI培地で各ウェルを4回洗浄した。洗浄後培地が除去された各ウェルに、100μLの10%FBS加RPMI培地と20μLのCellTiter 96(登録商標) AQueous One Solution Reagent (MTS reagent:プロメガ)を添加した。その後、96ウェルマイクロプレートを5%炭酸ガスが存在する湿式インキュベータ内で、20分、37℃でインキュベーションした。
(5)(4)でインキュベーションが終了した96ウェルマイクロプレートについて、マイクロプレートリーダを使って各ウェルの490 nmにおける吸光度を測定した。続いて、下式にしたがって、腫瘍細胞傷害活性パーセントを算出した。
  U251細胞株とPBMCを含むウェルの吸光度=A
  U251細胞株とPBMCとEphA2-CD3 BiTEを含むウェルの吸光度=B
  腫瘍細胞傷害活性パーセント (%)=[(A-B)/A]×100
Measurement of Percentage Tumor Cytotoxicity Using Peripheral Blood Mononuclear Cells (1) RPMI1640 medium (10% FBS-containing RPMI1640 medium) containing 10% FBS of U251 cell line at 1×10 5 cells/mL A cell suspension suspended in was prepared. The cell suspension was seeded on a 96-well plate at 100 μL/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
(2) Peripheral blood (heparin blood sampling) was collected from a lung cancer patient, and a cell group containing peripheral blood mononuclear cells (referred to as "PBMC") was collected using Lymphoprep (Alere Technologies AS) according to the attached protocol. did. The PBMCs were collected immediately before the step (3) described later.
(3) The number of cells of the PBMC is 5×10 4 , and the final concentration of EphA2-CD3 BiTE (registered trademark) is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added. As a control, a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1). After adding the PBMC and EphA2-CD3 BiTE (registered trademark), the total volume of RPMI medium containing 10% FBS was 200 μL per well. After adding PBMC and EphA2-CD3 BiTE (registered trademark), the mixture was incubated for 48 hours.
(4) After completion of the incubation, each well was washed 4 times with RPMI medium containing 10% FBS. After washing, 100 μL of RPMI medium supplemented with 10% FBS and 20 μL of CellTiter 96 (registered trademark) AQueous One Solution Reagent (MTS reagent: Promega) were added to each well from which the medium was removed. Then, the 96-well microplate was incubated at 37° C. for 20 minutes in a wet incubator in the presence of 5% carbon dioxide.
(5) Regarding the 96-well microplate in which the incubation was completed in (4), the absorbance at 490 nm of each well was measured using a microplate reader. Then, the percent tumor cytotoxic activity was calculated according to the following formula.
Absorbance of well containing U251 cell line and PBMC = A
Absorbance of well containing U251 cell line, PBMC and EphA2-CD3 BiTE = B
Tumor cytotoxic activity percentage (%) = [(A-B)/A] x 100

2.結果
 ニボルマブ又はペムブロリズマブの治療効果について、治療前の大きさの30%以下となった場合(部分奏功)及び病変の30%未満の縮小から20%未満の増大(安定)を陽性とし、20%以上の増大(進行)を陰性として、カットオフ値をROC曲線(Receiver Operator Characteristic Curve)を用いて算出して、感度及び特異度、ROC曲線の下の面積(Area Under Curve : AUC)を比較した。カットオフ値について、「真陽性感度-(1-特異度)」の値が最大になる腫瘍細胞傷害活性パーセント値を最適なカットオフ値とした。その結果、本実施例においてはペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントのカットオフ値は18.98%と算出され、感度78.26%、特異度60.00%となり、AUCは0.65217であった(図13A)。これに対して、ペムブロリズマブ又はニボルマブの治療前の腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値は55.00%と算出され、感度69.57%、特異度60.00%となり、AUCは0.60870であった(図13B)。これらの結果から腫瘍細胞傷害活性がニボルマブ及びペムブロリズマブの治療効果予測に優れていることが示された。
2. Results Regarding the therapeutic effect of nivolumab or pembrolizumab, when the size was 30% or less of the size before treatment (partial response) and the lesion was reduced by less than 30% to less than 20% (stable), the positive result was 20% or more. The increase (progression) was regarded as negative, and the cutoff value was calculated using a ROC curve (Receiver Operator Characteristic Curve) to compare the sensitivity and specificity with the area under the ROC curve (Area Under Curve: AUC). Regarding the cutoff value, the percent value of the tumor cytotoxicity that maximizes the value of "true positive sensitivity-(1-specificity)" was taken as the optimum cutoff value. As a result, in this example, the cutoff value of the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC before treatment with pembrolizumab or nivolumab was calculated to be 18.98%, the sensitivity was 78.26%, and the specificity was 60.00%, The AUC was 0.65217 (Figure 13A). On the other hand, the cut-off value for the positive rate of PD-L1 expressing cells in the tumor tissue before treatment with pembrolizumab or nivolumab was calculated to be 55.00%, the sensitivity was 69.57%, the specificity was 60.00%, and the AUC was 0.60870. (FIG. 13B). From these results, it was shown that the tumor cytotoxic activity was excellent in predicting the therapeutic effects of nivolumab and pembrolizumab.

 なお、ペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントと腫瘍組織におけるPD-L1発現細胞の陽性率を比較した結果、ピアソンの積率相関係数R2=0.088、p=0.1111であり、両者に相関は認められなかった(図14)。 As a result of comparing the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMC before treatment with pembrolizumab or nivolumab with the positive rate of PD-L1 expressing cells in the tumor tissue, the Pearson product moment correlation coefficient R 2 = The values were 0.088 and p=0.1111, and no correlation was found between them (Fig. 14).

 次に、図13で算出された、ペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントのカットオフ値18.98%について、18.98%以上の患者群(21名)と18.98%未満の患者群(9名)の無増悪生存期間を比較した結果、Log-rank testでp=0.0006となり、18.98%以上の患者群が有意に無増悪生存期間の延長を示した(図15A)。これに対して、図13で算出された、ペムブロリズマブ又はニボルマブの治療前の腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値55.00%について、55.00%以上の患者群(19名)と55.00%未満の患者群(11名)の無増悪生存期間を比較した結果、Log-rank testでp=0.6019となり、有意差は認められなかった(図15B)。ペムブロリズマブ又はニボルマブの治療前の腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値50.00%について、50.00%以上の患者群(22名)と50.00%未満の患者群(8名)の無増悪生存期間を比較した結果、Log-rank testでp=0.8935となり、有意差は認められなかった(図15C)。 Next, regarding the cutoff value 18.98% of the tumor cytotoxic activity percentage of peripheral blood T cells derived from PBMC before the treatment of pembrolizumab or nivolumab calculated in FIG. 13, 18.98% or more patient group (21 people) As a result of comparing the progression-free survival of the patient group less than 18.98% (nine), p = 0.0006 in the Log-rank test, and the patient group of 18.98% or more showed significantly prolonged progression-free survival (Fig. 15A). On the other hand, the cut-off value of the positive rate of PD-L1 expressing cells in the tumor tissue before the treatment of pembrolizumab or nivolumab of 55.00% calculated in FIG. 13 was 55.00% or more, and the patient group (19 persons) was 55.00% or more. As a result of comparing the progression-free survival time of the patient group (11 patients) of less than %, p = 0.6019 in the Log-rank test, and no significant difference was observed (Fig. 15B). With a cut-off value of 50.00% for PD-L1-expressing cells in the tumor tissue before treatment with pembrolizumab or nivolumab, progression-free in 50.00% or more patient groups (22 patients) and 50.00% or less patient groups (8 patients) As a result of comparing the survival periods, p=0.8935 was found in the Log-rank test, and no significant difference was observed (Fig. 15C).

 また、ペムブロリズマブ又はニボルマブの治療を受けた患者を別々に解析した結果、ペムブロリズマブ及びニボルマブいずれについても、治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントが18.98%以上の患者群が有意に無増悪生存期間の延長を示した(図16A及び図17A)。これに対して、PD-L1発現細胞の陽性率のカットオフ値55.00%以上、又は50.00%以上の患者は、ペムブロリズマブ及びニボルマブいずれについても、有意な無増悪生存期間の延長は認められなかった(図16B、C及び図17B、C)。 In addition, as a result of separately analyzing patients treated with pembrolizumab or nivolumab, for both pembrolizumab and nivolumab, the patient cytotoxic activity percentage of peripheral blood T cells derived from PBMC before treatment was 18.98% or more. It showed significantly prolonged progression-free survival (Figs. 16A and 17A). In contrast, patients with a PD-L1-expressing cell positive cut-off value of 55.00% or higher, or 50.00% or higher, did not have a significant prolongation of progression-free survival for both pembrolizumab and nivolumab ( 16B, C and 17B, C).

 ニボルマブ又はペムブロリズマブの治療について、治療効果及び無増悪生存期間の他に、重篤な有害事象を発症する患者を治療前に予測することも重要であることから、ペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性と重篤な有害事象の出現との関係を調べた。その結果、ペムブロリズマブ又はニボルマブの重篤な有害事象が出現した患者群(7名:肺炎3名、白質脳症1名、筋炎1名、肝機能障害1名、腸閉塞1名)は重篤な有害事象が出現しなかった患者群(23名)に比べて、有意に治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性が低かった(t検定にてp=0.0179)(図18A)。これに対して、ペムブロリズマブ又はニボルマブの治療前の腫瘍組織におけるPD-L1発現細胞の陽性率については、重篤な有害事象が出現した患者群(7名)と重篤な有害事象が出現しなかった患者群(23名)の間に有意差は認められなかった(t検定にてp=0.2601)(図18B)。 Regarding the treatment of nivolumab or pembrolizumab, it is important to predict the patient who develops serious adverse events before treatment, in addition to the therapeutic effect and progression-free survival, so PBMC before treatment with pembrolizumab or nivolumab is important. The relationship between the tumor cytotoxic activity of the peripheral blood T cells derived and the appearance of serious adverse events was investigated. As a result, patients with serious adverse events of pembrolizumab or nivolumab (7: 3 pneumonia, 1 leukoencephalopathy, 1 myositis, 1 liver dysfunction, 1 intestinal obstruction) were serious adverse events. The tumor cytotoxic activity of peripheral blood T cells derived from PBMCs before treatment was significantly lower than that in the patient group (23 patients) in which was not observed (p=0.0179 by t-test) (FIG. 18A). On the other hand, regarding the positive rate of PD-L1 expressing cells in the tumor tissue before treatment with pembrolizumab or nivolumab, there were no serious adverse events in the patient group (7 patients) in which serious adverse events occurred. No significant difference was observed between the patient groups (23 patients) (p=0.2601 by t-test) (FIG. 18B).

 次に、ペムブロリズマブ又はニボルマブの重篤な有害事象が出現しなかった場合を陽性とし、重篤な有害事象が出現した場合を陰性として、本実施例におけるカットオフ値をROC曲線(Receiver Operator Characteristic Curve)を用いて算出して、感度及び特異度、ROC曲線の下の面積(Area Under Curve : AUC)を比較した。カットオフ値について、「真陽性感度-(1-特異度)」の値が最大になる腫瘍細胞傷害活性パーセント値を最適なカットオフ値とした。その結果、ペムブロリズマブ又はニボルマブの治療前のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントのカットオフ値は35.60%と算出され、感度69.57%、特異度100.00%となり、AUCは0.77640であった(図19A)。これに対して、ペムブロリズマブ又はニボルマブの治療前の腫瘍組織におけるPD-L1発現細胞の陽性率のカットオフ値は65.00%と算出され、感度73.91%、特異度71.43%となり、AUCは0.65528であった(図19B)。これらの結果から腫瘍細胞傷害活性がニボルマブ及びペムブロリズマブの治療中止となる重篤な有害事象出現の予測に優れていることが示された。 Next, the case where serious adverse events of pembrolizumab or nivolumab did not occur was defined as positive, and the case where serious adverse events occurred was defined as negative, and the cutoff value in this example was calculated using the ROC curve (Receiver Operator Characteristic Curve). ) Was used to compare sensitivity and specificity, and the area under the ROC curve (Area Under Curve: AUC). Regarding the cutoff value, the percent value of the tumor cytotoxicity that maximizes the value of "true positive sensitivity-(1-specificity)" was taken as the optimum cutoff value. As a result, the cut-off value for the percent tumor cytotoxic activity of peripheral blood T cells derived from PBMCs before treatment with pembrolizumab or nivolumab was calculated to be 35.60%, sensitivity was 69.57%, specificity was 100.00%, and AUC was 0.77640. (Fig. 19A). On the other hand, the cut-off value of the positive rate of PD-L1 expressing cells in the tumor tissue before the treatment of pembrolizumab or nivolumab was calculated to be 65.00%, the sensitivity was 73.91%, the specificity was 71.43%, and the AUC was 0.65528. (FIG. 19B). These results indicate that the tumor cytotoxicity is excellent in predicting the occurrence of serious adverse events resulting in treatment discontinuation of nivolumab and pembrolizumab.

VIII.実施例7:インターフェロン-γの濃度による免疫療法剤の有用性の予測
1.方法
 非小細胞肺癌患者について、PBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントを測定し、培養上清のインターフェロン-γ(IFNγ)の濃度との相関を求めた。PBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセント及び培養上清のインターフェロンγの濃度は以下の方法により評価した。
VIII. Example 7: Prediction of usefulness of immunotherapeutic agents by the concentration of interferon-γ Method For non-small cell lung cancer patients, the percentage of tumor cytotoxic activity of peripheral blood T cells derived from PBMC was measured, and the correlation with the concentration of interferon-γ (IFNγ) in the culture supernatant was determined. The tumor cytotoxic activity percentage of peripheral blood T cells derived from PBMC and the concentration of interferon γ in the culture supernatant were evaluated by the following methods.

末梢血単核細胞を用いた腫瘍細胞傷害活性パーセントと培養上清インターフェロンγ濃度の測定
(1)U251細胞株を、1×105個/mLとなるように10%FBSを添加したRPMI1640培地(10%FBS加RPMI1640培地)に懸濁した細胞懸濁液を調製した。前記細胞懸濁液を100μL/ウェルとなるように96ウェルプレートに播種し、5%炭酸ガスが存在する湿式インキュベータ内で、24時間、37℃でインキュベーションした。
(2)肺癌患者から末梢血(ヘパリン採血)を採取し、LymphoprepTM (Alere Technologies AS)を使って、添付のプロトコールにしたがって末梢血単核細胞を含む細胞群(「PBMC」とする)を回収した。前記PBMCの回収は、後述する(3)の工程の直前に行った。
(3)(1)でインキュベーションが終わった96ウェルプレートに、1ウェルあたり、前記PBMCの細胞数が5×104個、EphA2-CD3 BiTE(登録商標)が終濃度で100 ng/mLとなるように添加した。コントロールとして、(1)でインキュベーションが終わった96ウェルプレート内に前記PBMCのみを添加し、EphA2-CD3 BiTE(登録商標)を添加しないウェルを準備した。前記PBMCとEphA2-CD3 BiTE(登録商標)を添加した後の1ウェルあたりの10%FBS加RPMI培地の量は、総量が200μLとなるようにした。PBMCとEphA2-CD3 BiTE(登録商標)を添加したあと、48時間インキュベーションした。
(4)インキュベーション終了後、培養上清を採取保存(-20℃)したのちに、10%FBS加RPMI培地で各ウェルを4回洗浄した。洗浄後培地が除去された各ウェルに、100μLの10%FBS加RPMI培地と20μLのCellTiter 96(登録商標) AQueous One Solution Reagent (MTS reagent:プロメガ)を添加した。その後、96ウェルマイクロプレートを5%炭酸ガスが存在する湿式インキュベータ内で、20分、37℃でインキュベーションした。
(5)(4)でインキュベーションが終了した96ウェルマイクロプレートについて、マイクロプレートリーダを使って各ウェルの490 nmにおける吸光度を測定した。続いて、下式にしたがって、腫瘍細胞傷害活性パーセントを算出した。
  U251細胞株とPBMCを含むウェルの吸光度=A
  U251細胞株とPBMCとEphA2-CD3 BiTEを含むウェルの吸光度=B
  腫瘍細胞傷害活性パーセント (%)=[(A-B)/A]×100
(6)(4)で-20℃で保存した培養上清について、抗腫瘍作用を示すインターフェロンγ(IFNγ)の濃度を求めた。IFNγの濃度は、Bio-Plex Proヒトサイトカイン GI 27-plexパネル(Bio-Rad)を使用して測定した。
Percentage of tumor cytotoxicity using peripheral blood mononuclear cells and measurement of culture supernatant interferon γ concentration (1) RPMI1640 medium containing 10% FBS of U251 cell line (1×10 5 cells/mL) ( A cell suspension suspended in RPMI1640 medium containing 10% FBS) was prepared. The cell suspension was seeded on a 96-well plate at 100 μL/well and incubated at 37° C. for 24 hours in a wet incubator in the presence of 5% carbon dioxide gas.
(2) Peripheral blood (heparin blood sampling) was collected from a lung cancer patient, and a cell group containing peripheral blood mononuclear cells (referred to as "PBMC") was collected using Lymphoprep (Alere Technologies AS) according to the attached protocol. did. The PBMCs were collected immediately before the step (3) described later.
(3) The number of cells of the PBMC is 5×10 4 , and the final concentration of EphA2-CD3 BiTE (registered trademark) is 100 ng/mL per well in the 96-well plate after the incubation in (1). So added. As a control, a well in which only the PBMC was added and EphA2-CD3 BiTE (registered trademark) was not added was prepared in a 96-well plate after the incubation in (1). After adding the PBMC and EphA2-CD3 BiTE (registered trademark), the total volume of RPMI medium containing 10% FBS was 200 μL per well. After adding PBMC and EphA2-CD3 BiTE (registered trademark), the mixture was incubated for 48 hours.
(4) After the incubation, the culture supernatant was collected and stored (-20°C), and then each well was washed 4 times with RPMI medium containing 10% FBS. After washing, 100 μL of RPMI medium supplemented with 10% FBS and 20 μL of CellTiter 96 (registered trademark) AQueous One Solution Reagent (MTS reagent: Promega) were added to each well from which the medium was removed. Then, the 96-well microplate was incubated at 37° C. for 20 minutes in a wet incubator in the presence of 5% carbon dioxide.
(5) Regarding the 96-well microplate in which the incubation was completed in (4), the absorbance at 490 nm of each well was measured using a microplate reader. Then, the percent tumor cytotoxic activity was calculated according to the following formula.
Absorbance of well containing U251 cell line and PBMC = A
Absorbance of well containing U251 cell line, PBMC and EphA2-CD3 BiTE = B
Tumor cytotoxic activity percentage (%) = [(A-B)/A] x 100
(6) The concentration of interferon γ (IFNγ) showing an antitumor effect was determined for the culture supernatant stored at -20°C in (4). The concentration of IFNγ was measured using the Bio-Plex Pro human cytokine GI 27-plex panel (Bio-Rad).

2.結果
 非小細胞肺癌患者のPBMCに由来する末梢血T細胞の腫瘍細胞傷害活性パーセントと培養上清のインターフェロン-γ濃度を比較した結果、ピアソンの積率相関係数R2=0.44、p=0.0021であり、両者に相関が認められた(図20)。このことから、末梢血T細胞の腫瘍細胞傷害活性パーセントに代えて、培養上清のインターフェロン-γ濃度を測定しても免疫療法剤の有用性を評価できると考えられた。
2. Results As a result of comparing the tumor cytotoxic activity percentage of peripheral blood T cells derived from PBMCs of patients with non-small cell lung cancer with the interferon-γ concentration of the culture supernatant, the Pearson product moment correlation coefficient R 2 =0.44, p=0.0021 And a correlation was observed between the two (FIG. 20). From this, it was considered that the usefulness of the immunotherapeutic agent could be evaluated by measuring the interferon-γ concentration of the culture supernatant instead of the percent of tumor cytotoxic activity of peripheral blood T cells.

101 処理部
10 提示装置
201 処理部
20 補助装置
101 Processing Unit 10 Presentation Device 201 Processing Unit 20 Auxiliary Device

Claims (39)

下記工程1、及び工程2を含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性を提示する方法:
工程1:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程2:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
A method for presenting usefulness of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor, comprising the following Step 1 and Step 2:
Step 1: Evaluating whether or not the tumor cells directly or indirectly contacted with the peripheral blood mononuclear cells collected from the patient in vitro are injured, and Step 2: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
前記工程2が、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は
前記工程2が、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、
を含む、請求項1に記載の提示方法。
The usefulness is the presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and incapable of resuming administration,
When the step 2 shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent for the patient needs to be interrupted and the administration cannot be restarted. If the step of presenting that no adverse event occurs, and/or the step 2 is shown in the evaluation that the tumor cells are not injured, the patient is treated with the immunotherapeutic agent. Presenting an adverse event that requires discontinuation and that makes it impossible to resume administration,
The presentation method according to claim 1, comprising:
前記有用性が、前記免疫療法剤の効果の有無であり、
前記工程2が、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は
前記工程2が、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、
を含む、請求項1又は請求項2に記載の提示方法。
The usefulness is the presence or absence of the effect of the immunotherapeutic agent,
Said step 2 presents to said patient that said immunotherapeutic agent is effective when said tumor cells are shown to be injured, and/or said step 2 Presenting to the patient that the immunotherapeutic agent is ineffective if the evaluation indicates that the tumor cells are not injured,
The presentation method according to claim 1 or 2, which includes:
前記工程1の腫瘍細胞が傷害されているか否かを評価する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程、及び
前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高い場合に前記腫瘍細胞が傷害されていることを示す工程、及び/又は前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より低い場合に前記腫瘍細胞が傷害されていないことを示す工程、
を含む、請求項1から請求項3のいずれか一項に記載の提示方法。
The step of evaluating whether or not the tumor cells in the step 1 is injured,
A step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells contacted with the tumor cells, and comparing the value reflecting the tumor cytotoxic activity with a corresponding reference value, and the reference corresponding to the value If the value is lower than the corresponding reference value, the step of indicating that the tumor cells are injured when the value is higher than the value, and/or the value reflecting the tumor cytotoxic activity is compared with the corresponding reference value. A step showing that the tumor cells are not injured,
The presentation method according to any one of claims 1 to 3, further comprising:
腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、請求項4に記載の提示方法。 The presentation method according to claim 4, wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、請求項4に記載の提示方法。 The presentation method according to claim 4, wherein the in vitro contact is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is a measurement value of interferon-γ in the culture medium. 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、請求項1から請求項6のいずれか一項に記載の提示方法。 7. The in vitro contact is an indirect contact between a peripheral blood mononuclear cell collected from the patient and a tumor cell via an engager, according to any one of claims 1 to 6. How to present the description. 前記エンゲージャーが、二重特異性分子である、請求項7に記載の提示方法。 The presentation method according to claim 7, wherein the engager is a bispecific molecule. 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、請求項8に記載の提示方法。 9. The bispecific molecule is a molecule comprising an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. How to present the description. 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、請求項1から請求項9に記載のいずれか一項に提示方法。 The method according to any one of claims 1 to 9, wherein the tumor cells are cultured cell lines derived from malignant tumors. 請求項7から請求項10のいずれか一項に記載の提示方法において、前記免疫療法剤について、前記悪性腫瘍を有する患者における有用性を評価するための、エンゲージャーを含む、
検査試薬。
The presentation method according to any one of claims 7 to 10, comprising an engager for evaluating the usefulness of the immunotherapeutic agent in a patient having the malignant tumor.
Testing reagent.
下記工程iから工程iiを含む、悪性腫瘍を治療するための免疫療法剤について、前記悪性腫瘍を有する患者における有用性の決定を補助する方法:
工程i:前記患者から採取された末梢血単核細胞と、直接、又は間接的にin vitroで接触した腫瘍細胞が傷害されているか否かを評価する工程、及び
工程ii:前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が有用であることを提示する工程、及び/又は
前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が有用でないことを提示する工程。
A method for assisting in determining the usefulness of an immunotherapeutic agent for treating a malignant tumor in a patient having the malignant tumor, comprising the following steps i to ii:
Step i: a step of evaluating whether or not tumor cells directly or indirectly contacted with peripheral blood mononuclear cells collected from the patient in vitro are injured, and step ii: the tumor in the evaluation Presenting the immunotherapeutic agent to the patient as being useful if the cells are shown to be injured, and/or indicating that the tumor cells are not injured in the assessment. If so, presenting to the patient that the immunotherapeutic agent is not useful.
前記有用性が、前記免疫療法剤に起因する、治療中断の必要があり、かつ投与再開が不可能となる有害事象の発生の有無であり、
工程iiが、前記評価において前記腫瘍細胞が傷害されていることが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こさないことを提示する工程、及び/又は
工程iiが、前記評価において前記腫瘍細胞が傷害されていないことが示された場合に、前記患者に対して前記免疫療法剤が、治療中断の必要があり、かつ投与再開が不可能となる有害事象を起こすことを提示する工程、
を含む、請求項12に記載の補助方法。
The usefulness is the presence or absence of an adverse event resulting from the immunotherapeutic agent, which requires treatment interruption and incapable of resuming administration,
If the step ii shows that the tumor cells are injured in the evaluation, the immunotherapeutic agent is harmful to the patient because the treatment needs to be interrupted and the administration cannot be restarted. If the step of presenting no event and/or step ii is shown in the assessment that the tumor cells are not injured, the immunotherapeutic agent is directed to the patient for treatment interruption. The need to present an adverse event that makes it impossible to resume administration,
13. The method of claim 12, including:
前記有用性が、前記免疫療法剤の効果の有無であり、
工程iiが、前記評価において前記腫瘍細胞が傷害されていると決定された場合に、前記患者に対して前記免疫療法剤が有効であることを提示する工程、及び/又は
工程iiが、前記評価において前記腫瘍細胞が傷害されていないと決定された場合に、前記患者に対して前記免疫療法剤が無効であることを提示する工程、
を含む、請求項12又は請求項13に記載の補助方法。
The usefulness is the presence or absence of the effect of the immunotherapeutic agent,
Step ii presenting to said patient that said immunotherapeutic agent is effective if said tumor cells are determined to be injured in said evaluation, and/or step ii comprises said evaluation Presenting to the patient that the immunotherapeutic agent is ineffective if it is determined that the tumor cells are not injured in
The auxiliary method according to claim 12 or claim 13, which comprises:
前記工程iの腫瘍細胞が傷害されているか否かを評価する工程が、
前記腫瘍細胞と接触した末梢血単核細胞の腫瘍細胞傷害活性を反映する値を取得する工程、及び
前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より高い場合に腫瘍細胞が傷害されていることを示す工程、及び/又は前記腫瘍細胞傷害活性を反映する値を対応する基準値と比較し、前記値が対応する基準値より低い場合に前記腫瘍細胞が傷害されていないことを示す工程、
を含む、請求項12から請求項14のいずれか一項に記載の補助方法。
The step of evaluating whether the tumor cells in step i are injured,
A step of obtaining a value reflecting the tumor cytotoxic activity of peripheral blood mononuclear cells in contact with the tumor cell, and comparing the value reflecting the tumor cytotoxic activity with a corresponding reference value, and the value corresponds to the criterion A step indicating that the tumor cells are injured when higher than the value, and/or a value reflecting the tumor cytotoxic activity is compared with a corresponding reference value, and when the value is lower than the corresponding reference value, A step showing that the tumor cells are not injured,
The assistance method according to any one of claims 12 to 14, which comprises:
腫瘍細胞傷害活性を反映する値が、前記腫瘍細胞の生細胞数又は死細胞数に基づいて算出された腫瘍細胞傷害活性の値である、請求項15に記載の補助方法。 The method according to claim 15, wherein the value reflecting the tumor cytotoxic activity is the value of the tumor cytotoxic activity calculated based on the number of living cells or dead cells of the tumor cells. 前記in vitroにおける接触が培養培地内での接触であり、腫瘍細胞傷害活性を反映する値が、前記培養培地中のインターフェロン-γの測定値である、請求項15に記載の補助方法。 The assisting method according to claim 15, wherein the in vitro contact is contact in the culture medium, and the value reflecting the tumor cytotoxic activity is a measurement value of interferon-γ in the culture medium. 前記in vitroにおける接触が、前記患者から採取された末梢血単核細胞と、腫瘍細胞との、エンゲージャーを介した間接的な接触である、請求項12から請求項17のいずれか一項に記載の補助方法。 18. The in vitro contact is an indirect contact between peripheral blood mononuclear cells collected from the patient and tumor cells via an engager, according to any one of claims 12 to 17. Described assistance method. 前記エンゲージャーが、二重特異性分子である、請求項18に記載の補助方法。 19. The method of claim 18, wherein the engager is a bispecific molecule. 前記二重特異性分子が、T細胞の表面抗原の少なくとも一種と結合する抗原結合領域と、前記腫瘍細胞の表面抗原の少なくとも一種と結合する抗原結合領域とを含む分子である、請求項19に記載の補助方法。 20. The bispecific molecule is a molecule comprising an antigen-binding region that binds to at least one surface antigen of T cells and an antigen-binding region that binds to at least one surface antigen of tumor cells. Described assistance method. 前記腫瘍細胞が、悪性腫瘍由来の培養細胞株である、請求項12から請求項20に記載のいずれか一項に補助方法。 The method according to any one of claims 12 to 20, wherein the tumor cells are cultured cell lines derived from malignant tumors. 請求項18から請求項21のいずれか一項に記載の補助方法において、前記免疫療法剤について、前記悪性腫瘍を有する患者における有用性を評価するための、エンゲージャーを含む、
検査試薬。
The adjuvant method according to any one of claims 18 to 21, further comprising an engager for evaluating the usefulness of the immunotherapeutic agent in a patient having the malignant tumor.
Testing reagent.
請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、悪性腫瘍を治療するための免疫療法剤。 Immunity for treating a malignant tumor, which is administered to a patient having a malignant tumor for which the immunotherapeutic agent is suggested to be useful in the method for presentation according to any one of claims 1 to 10. Therapeutic agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して1次治療のために投与される、免疫療法剤。 An immunotherapy, which is administered for a first-line treatment to a patient having a malignant tumor in which the immunotherapeutic agent is shown to be useful in the presentation method according to any one of claims 1 to 10. Agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して術前化学療法のために投与される、免疫療法剤。 Immune, which is administered for preoperative chemotherapy to a patient having a malignant tumor in which the immunotherapeutic agent is shown to be useful in the presentation method according to any one of claims 1 to 10. Therapeutic agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して術後補助化学療法のために投与される、免疫療法剤。 The immunotherapy agent is administered for postoperative adjuvant chemotherapy to a patient having a malignant tumor which is suggested to be useful in the presentation method according to any one of claims 1 to 10. Immunotherapeutic agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、放射線治療後の前記患者に投与される免疫療法剤。 An immunity to be administered to a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of claims 1 to 10, which is administered to the patient after radiation treatment. Therapeutic agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者であって、化学放射線治療後の前記患者に投与される免疫療法剤。 A patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of claims 1 to 10, and the patient is administered to the patient after chemoradiotherapy. Immunotherapeutic agent. 免疫療法剤が、免疫チェックポイント阻害剤である、請求項23から請求項28のいずれか一項に記載の免疫療法剤。 The immunotherapy agent according to any one of claims 23 to 28, wherein the immunotherapy agent is an immune checkpoint inhibitor. 前記免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、請求項29に記載の免疫療法剤。 The immunotherapy agent according to claim 29, wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、請求項23から請求項30のいずれか一項に記載の免疫療法剤。 31. The method according to any one of claims 23 to 30, wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. Immunotherapeutic agent. 前記肺癌が、前記免疫療法剤の投与開始時に間質性肺炎を合併している、請求項31に記載の免疫療法剤。 The immunotherapy agent according to claim 31, wherein the lung cancer is associated with interstitial pneumonia at the start of administration of the immunotherapy agent. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、免疫チェックポイント阻害剤であって、他の1種以上の免疫チェックポイント阻害剤と組み合わせて投与される、免疫チェックポイント阻害剤。 An immunological checkpoint inhibitor, which is administered to a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of claims 1 to 10. , An immune checkpoint inhibitor administered in combination with one or more other immune checkpoint inhibitors. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して投与される、免疫チェックポイント阻害剤であって、免疫チェックポイント阻害剤以外の1種以上の免疫療法剤と組み合わせて投与される、免疫チェックポイント阻害剤。 An immunological checkpoint inhibitor, which is administered to a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful in the presentation method according to any one of claims 1 to 10. , An immune checkpoint inhibitor, which is administered in combination with one or more immunotherapeutic agents other than the immune checkpoint inhibitor. 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、請求項33又は請求項34に記載の免疫チェックポイント阻害剤。 35. The immune checkpoint inhibitor according to claim 33 or 34, wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. .. 免疫チェックポイント阻害剤が、抗PD1抗体、抗PD-L1抗体、抗CTLA-4抗体、抗Tim3抗体、又は抗LAG3抗体を含む、請求項33から請求項35のいずれか一項に記載の免疫チェックポイント阻害剤。 The immunity according to any one of claims 33 to 35, wherein the immune checkpoint inhibitor comprises an anti-PD1 antibody, an anti-PD-L1 antibody, an anti-CTLA-4 antibody, an anti-Tim3 antibody, or an anti-LAG3 antibody. Checkpoint inhibitor. 請求項1から請求項10のいずれか一項に記載の提示方法において免疫療法剤が有用であることが提示された悪性腫瘍を有する患者に対して、免疫療法剤以外の1種以上の抗がん剤と組み合わせて投与される免疫療法剤。 The method for presenting an immunotherapeutic agent according to any one of claims 1 to 10, wherein at least one anti-immunotherapeutic agent other than the immunotherapeutic agent is administered to a patient having a malignant tumor for whom the immunotherapeutic agent is suggested to be useful. An immunotherapeutic agent that is administered in combination with a cancer drug. 前記悪性腫瘍が、肺癌、頭頚部癌、悪性黒色腫、腎細胞癌、尿路上皮癌、胃癌、又は悪性胸膜中皮腫である、請求項37に記載の免疫療法剤。 38. The immunotherapeutic agent according to claim 37, wherein the malignant tumor is lung cancer, head and neck cancer, malignant melanoma, renal cell carcinoma, urothelial cancer, gastric cancer, or malignant pleural mesothelioma. 免疫療法剤が、免疫チェックポイント阻害剤である、請求項37又は請求項38に記載の免疫療法剤。 39. The immunotherapeutic agent according to claim 37 or 38, wherein the immunotherapeutic agent is an immune checkpoint inhibitor.
PCT/JP2018/048494 2018-12-28 2018-12-28 Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor Ceased WO2020136879A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/JP2018/048494 WO2020136879A1 (en) 2018-12-28 2018-12-28 Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/JP2018/048494 WO2020136879A1 (en) 2018-12-28 2018-12-28 Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor

Publications (1)

Publication Number Publication Date
WO2020136879A1 true WO2020136879A1 (en) 2020-07-02

Family

ID=71126269

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/048494 Ceased WO2020136879A1 (en) 2018-12-28 2018-12-28 Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor

Country Status (1)

Country Link
WO (1) WO2020136879A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117987297A (en) * 2023-08-11 2024-05-07 合肥瀚微生物科技有限公司 A strain of intestinal bacteria and its application in anti-tumor immunotherapy
CN119530396A (en) * 2025-01-22 2025-02-28 中国科学院杭州医学研究所 Markers and systems for predicting the efficacy of tumor immunotherapy

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016531907A (en) * 2013-08-02 2016-10-13 アデュロ・バイオテック・ホールディングス・ヨーロッパ・ベスローテン・フエンノートシャップAduro Biotech Holdings, Europe B.V. Combination of CD27 agonist and immune checkpoint inhibition for immune stimulation

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016531907A (en) * 2013-08-02 2016-10-13 アデュロ・バイオテック・ホールディングス・ヨーロッパ・ベスローテン・フエンノートシャップAduro Biotech Holdings, Europe B.V. Combination of CD27 agonist and immune checkpoint inhibition for immune stimulation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHAMOTO, KENJI: ""Predictive biomarkers for anti-PD-l/PD-Ll antibody therapy", JOURNAL OF MOLECULAR TARGETED THERAPY FOR CANCER, vol. 13, no. 4, 2016, pages 485 (71) - 489 (75), XP009522295, ISSN: 1347-6955 *
KRIJN K DIJKSTRA; CHIARA M CATTANEO; FLEUR WEEBER; MYRIAM CHALABI; JORIS VAN DE HAAR; LORENZO F FANCHI; MAARTEN SLAGTER; DAPHNE L : "Generation of Tumor- Reactive T Cells by Co-culture of Peripheral Blood Lymphocytes and Tumor Organoids", CELL, vol. 174, no. 6, 1 September 2018 (2018-09-01), pages 1 - 26, XP055722242, ISSN: 0092-8674, DOI: 10.1016/j.cell.2018.07.009 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117987297A (en) * 2023-08-11 2024-05-07 合肥瀚微生物科技有限公司 A strain of intestinal bacteria and its application in anti-tumor immunotherapy
CN119530396A (en) * 2025-01-22 2025-02-28 中国科学院杭州医学研究所 Markers and systems for predicting the efficacy of tumor immunotherapy

Similar Documents

Publication Publication Date Title
JP6508785B2 (en) Treatment Using Bruton Tyrosine Kinase Inhibitors and Immunotherapy
JP2023118967A (en) Tumor mutational burden
AU2017278220C1 (en) Receptor selective retinoid and rexinoid compounds and immune modulators for cancer immunotherapy
JP2025029025A (en) Methods for treating lung cancer using a combination of anti-PD-1 and anti-CTLA-4 antibodies
ES2877757T3 (en) Therapeutic treatment of breast cancer based on c-MAF status
BR112017008165B1 (en) SINGLE-DOMAIN ANTIBODY TARGETED AGAINST AN INTRACELLULAR COMPONENT AND ITS USE, IN VITRO METHODS FOR MEASURING THE LEVELS OF THE ABOVE ANTIBODY AND TO DIAGNOSTIC A DISTRIBUTION MEASURED BY AN INTRACELLULAR COMPONENT, POLYEPHYDROPOLYOID, ISOLATED COMPOSITE, ISOLATED AND COMPOSITE
CN117771364A (en) Ovarian cancer therapy based on anti-CD 47 agents
JP2022527133A (en) How to regulate macrophage activity
JP2016521280A (en) How to treat cancer with combination therapy
US20180246113A1 (en) Cxcl11 and smica as predictive biomarkers for efficacy of anti-ctla4 immunotherapy
US20210315909A1 (en) Polymorphic compounds and uses thereof
CA3087779A1 (en) Agents and methods for predicting response to therapy
AU2017347822A1 (en) Use of beta-catenin as a biomarker for treating cancers using anti-Dkk-1 antibody
TW202021617A (en) Biomarker for predicting efficacy of immune checkpoint inhibitor
US10220091B2 (en) Combination treatments with sonic hedgehog inhibitors
DK2984108T3 (en) Anti-s100a7 antibodies for the treatment and diagnosis of cancer
WO2020136879A1 (en) Method for presenting effectiveness of immunotherapeutic agent for treating malignant tumor and immunotherapeutic agent for treating malignant tumor
MX2014013931A (en) Methods for treatment of gastric cancer.
JP2023553247A (en) Compositions and methods for cancer diagnosis
CN113677994A (en) Methods and agents for assessing T cell function and predicting response to therapy
JP2021517135A (en) Combination treatment of chemotherapy-resistant cancer
BR112021000340A2 (en) SPECIFIC EPITHEPHONE FOR SMO PROTEIN, ANTIBODY OR FRAGMENT OF THE SAME, NUCLEIC ACID MOLECULE, VECTOR, HOST CELL, COMPOSITION OF VACCINE, PHARMACEUTICAL COMPOSITION, USE OF THE ANTIBODY FOR THE QUANTUM PROTECTION, TO QUANTIFY SMO PROTEIN
CN111867598A (en) tumor immune activator
CN119923258A (en) Treatment of cancers with mutations in WNT pathway tumor suppressor genes
EP3487490A1 (en) Compounds for targeting cancer stem cells

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: 18944936

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18944936

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP