WO2013168550A1 - Cancer immunotherapy evaluation method, cancer immunotherapy evaluation device, cancer immunotherapy evaluation program, cancer immunotherapy evaluation system, and information communication terminal device - Google Patents

Cancer immunotherapy evaluation method, cancer immunotherapy evaluation device, cancer immunotherapy evaluation program, cancer immunotherapy evaluation system, and information communication terminal device Download PDF

Info

Publication number
WO2013168550A1
WO2013168550A1 PCT/JP2013/061807 JP2013061807W WO2013168550A1 WO 2013168550 A1 WO2013168550 A1 WO 2013168550A1 JP 2013061807 W JP2013061807 W JP 2013061807W WO 2013168550 A1 WO2013168550 A1 WO 2013168550A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
treatment
evaluation
discriminant
value
Prior art date
Application number
PCT/JP2013/061807
Other languages
French (fr)
Japanese (ja)
Inventor
信矢 菊池
純也 米田
祐子 道端
小林 幹
Original Assignee
味の素株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 味の素株式会社 filed Critical 味の素株式会社
Priority to JP2014514434A priority Critical patent/JP6375947B2/en
Publication of WO2013168550A1 publication Critical patent/WO2013168550A1/en

Links

Images

Classifications

    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6806Determination of free amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/70Mechanisms involved in disease identification
    • G01N2800/7023(Hyper)proliferation
    • G01N2800/7028Cancer

Definitions

  • the present invention relates to a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation device, a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a cancer immunotherapy evaluation system, and an information communication terminal device.
  • Immunotherapy is a treatment that uses the substance that activates immunocompetent cells, cytokines, antibodies, etc. to direct the immune function of the living body in the intended direction, and immunotherapy for various cancers It is known.
  • cancer immunotherapy can exert certain effects, there are responders and non-responders to treatment, and the effects vary among individuals.
  • the responder includes an individual whose cancer has been eradicated, reduced or improved (mixed responder or partial responder), an individual whose cancer has not progressed, and the like.
  • an individual in which cancer has not progressed is an increase in cancer, quality of life (QOL) is improved or maintained, and / or average compared to an individual who has not been treated. An individual whose life expectancy has increased.
  • QOL quality of life
  • a method of evaluating the effectiveness of a treatment method for cancer As a method of evaluating the effectiveness of a treatment method for cancer, a method of evaluating by image is basically proposed, and a method of evaluating by extending the survival period is usually used.
  • many of the conventional methods for evaluating the effectiveness of therapeutic methods for cancer are used in the evaluation and re-evaluation of new drugs. For this reason, it is difficult to apply the conventional method to the evaluation of normal cancer treatment.
  • Non-Patent Documents 1 and 2 There are also attempts to directly measure immune responses in tumor tissues and regional lymph nodes. For example, it has been reported that detection of antigen-specific activated T cells in a local cancer region is highly involved in clinical outcome (Non-patent Document 3). However, the burden on the patient side is large, labor is required, and clinical specimens are often not obtained.
  • Non-patent Document 4 blood serum serine and glutamic acid concentrations are normalized by surgery in breast cancer patients.
  • Non-Patent Documents 5 and 6 a decrease in non-essential amino acids in blood immediately after surgery for cancer patients.
  • Non-patent Document 7 a decrease in blood proline, taurine, and glutamic acid concentrations increase and alanine concentration decreases by Ukrain treatment of breast cancer patients.
  • Non-patent Document 8 17 types of blood amino acid concentrations were increased in the first cycle in patients who were effective with chemotherapy including cisplatin compared to patients who were not effective.
  • Patent Literature 1, Patent Literature 2, and Patent Literature 3 relating to a method for associating an amino acid concentration with a biological state are disclosed as prior patents.
  • Patent Document 4 relating to a method for evaluating lung cancer status using amino acid concentration
  • Patent Literature 5 relating to a method for evaluating breast cancer status using amino acid concentration
  • Patent Document 6 relating to a method for evaluating colon cancer status using amino acid concentration
  • Patent Document 7 related to a method for evaluating cancer status using amino acid concentration
  • Patent Document 8 related to a method for evaluating gastric cancer status using amino acid concentration
  • Patent Document 9 Patent Document 9
  • Patent Document 10 that evaluates the state of female genital cancer using amino acid concentration
  • Patent Document 11 that evaluates the state of prostate disease including prostate cancer using amino acid concentration are disclosed.
  • Non-Patent Documents 4 to 8 are reports on surgical operation and chemotherapy, and there are no reports related to changes in blood amino acid concentration in cancer immunotherapy. Further, even if the index formula groups disclosed in Patent Document 1 to Patent Document 11 are used for evaluating the therapeutic effect of cancer immunotherapy, the evaluation target is different, so that sufficient accuracy can be obtained for evaluating the therapeutic effect. Can not.
  • the present invention has been made in view of the above problems, and an cancer immunotherapy evaluation method, a cancer immunotherapy evaluation apparatus, which can accurately evaluate the therapeutic effect of cancer immunotherapy using the blood amino acid concentration,
  • An object is to provide a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a cancer immunotherapy evaluation system, and an information communication terminal device.
  • the present inventors have accurately determined the therapeutic effect of cancer immunotherapy using amino acids in blood (specifically, the anti-tumor immune effect in cancer immunotherapy). The present inventors have found that this can be done and have completed the present invention.
  • the method for evaluating cancer immunotherapy includes blood collected before the treatment is started from an evaluation subject receiving treatment by cancer immunotherapy.
  • An acquisition step of acquiring amino acid concentration data before starting treatment regarding the concentration value of amino acids in the blood, and amino acid concentration data after starting treatment regarding the concentration value of amino acids in blood collected after the treatment is started from the evaluation target;
  • the post-treatment amino acid concentration data may be data (after treatment amino acid concentration data) corresponding to “after treatment” amino acid concentration data described later.
  • cancer immunotherapy includes immune cell therapy, peptide / vaccine therapy, BMR (Biological Response Modifiers) therapy, cytokine therapy, antibody therapy, and induction of antitumor effect by release of immunosuppressive mechanism, Etc. are included.
  • BMR Bio Response Modifiers
  • before treatment is started may be referred to as “before treatment” or “before treatment start”
  • after treatment is started may be referred to as “after treatment start”.
  • before treatment start includes, for example, before the first narrow treatment in a broad sense over a certain period of time.
  • after the start of treatment includes, for example, after the first narrow treatment in the broad sense treatment for a certain period of time and before the final narrow treatment (for example, After being treated in a broad sense over a certain period of time (for example, after being treated generally), etc. included.
  • the concentration value reference evaluation step is included in the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment. Based on the concentration value of at least one of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro. It is characterized by evaluating the effect of the said treatment with respect to an evaluation object.
  • the cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step is based on the concentration value and the treatment is effective for the evaluation object. (E.g., further comprising a concentration value criterion determining step for determining whether or not the treatment is effective for the evaluation object based on the concentration value).
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step includes the pre-treatment amino acid concentration data, the post-treatment amino acid concentration data, and the amino acid Based on a preset multivariate discriminant that includes the concentration of as a variable, a discriminant value calculation that calculates a discriminant value that is a value of the multivariate discriminant and that corresponds to an evaluation result regarding the effect of the treatment on the evaluation target
  • the method further includes a step.
  • the discriminant value calculating step is based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, and the amino acid concentration value before starting treatment and the amino acid after starting treatment.
  • the discriminant value may be calculated by calculating the ratio or difference with the concentration value and substituting the calculated concentration value ratio or difference of each amino acid into each variable included in the multivariate discriminant.
  • the amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the multivariate discriminant is a logistic regression equation, a fractional equation, a linear discriminant, a multiple regression equation, or a support vector machine. It is any one of a created formula, a formula created by Mahalanobis distance method, a formula created by canonical discriminant analysis, and a formula created by a decision tree.
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the discriminant value calculating step includes Val included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. , Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Based on the multivariate discriminant including at least one of Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro as the variables. The discriminant value is calculated.
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the multivariate discriminant is Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His.
  • the fractional expression including at least one of the variables as the variable, or including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, Leu as the variable
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step is based on the discriminant value calculated in the discriminant value calculation step.
  • the method further comprises a discriminant value criterion evaluation step for evaluating the effect of the treatment on.
  • the cancer immunotherapy evaluation method is the above-described cancer immunotherapy evaluation method, wherein the discriminant value criterion evaluation step is based on the discriminant value and the treatment is effective for the evaluation object. (E.g., further including a discrimination value criterion discrimination step for discriminating whether or not the treatment is effective for the evaluation object based on the discrimination value).
  • the cancer immunotherapy evaluation apparatus is a cancer immunotherapy evaluation apparatus including a control unit and a storage unit, and the control unit is configured to perform the treatment before treatment by cancer immunotherapy is started.
  • the control unit is configured to perform the treatment before treatment by cancer immunotherapy is started.
  • a discriminant value calculating means for calculating a discriminant value that is a value of the multivariate discriminant and corresponds to an evaluation result relating to the effect of the treatment on the evaluation target. It is characterized by this.
  • the discriminant value calculating means is based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, and the amino acid concentration value before starting the treatment and the amino acid after starting the treatment.
  • the discriminant value may be calculated by calculating the ratio or difference with the concentration value and substituting the calculated concentration value ratio or difference of each amino acid into each variable included in the multivariate discriminant.
  • the amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
  • the cancer immunotherapy evaluation apparatus is the cancer immunotherapy evaluation apparatus according to the present invention, wherein the control unit has an effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation means. And a discriminant value criterion evaluating means for evaluating the above.
  • the cancer immunotherapy evaluation apparatus is the above-described cancer immunotherapy evaluation apparatus, wherein (i) the control unit includes amino acid concentration data and cancer state index data relating to an index representing a cancer state.
  • a multivariate discriminant creating unit that creates the multivariate discriminant stored in the storage unit based on the cancer state information stored in the unit; and (ii) the multivariate discriminant creating unit includes the cancer Candidate multivariate discriminant creating means for creating a candidate multivariate discriminant that is a candidate for the multivariate discriminant based on a predetermined formula creating method from state information, and the candidate multivariate discriminant creating means created by the candidate multivariate discriminant creating means
  • the candidate multivariate discriminant verification means for verifying the candidate multivariate discriminant based on a predetermined verification method, and selecting the candidate multivariate discriminant variable based on the predetermined variable selection method,
  • Variable size Variable selection means for selecting a combination of the amino acid concentration data included in the cancer state information used when creating an expression, the candidate multivariate
  • the cancer immunotherapy evaluation method is a cancer immunotherapy evaluation method executed in an information processing apparatus including a control unit and a storage unit, and is based on cancer immunotherapy executed in the control unit.
  • Amino acid concentration data before the start of treatment regarding the concentration value of the evaluation target amino acid before the start of treatment, and an amino acid concentration data after the start of treatment regarding the concentration value of the amino acid of the evaluation target after the start of the treatment Based on the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation target is a value of the multivariate discriminant And a discriminant value calculating step for calculating.
  • the cancer immunotherapy evaluation method according to the present invention is the cancer immunotherapy evaluation method according to the present invention, based on the discriminant value calculated in the discriminant value calculation step executed in the control unit, with respect to the evaluation object.
  • the method further includes a discriminant value criterion evaluation step for evaluating the effect of treatment.
  • a cancer immunotherapy evaluation program is a cancer immunotherapy evaluation program to be executed in an information processing apparatus including a control unit and a storage unit, and the cancer immunity evaluation program to be executed in the control unit.
  • Amino acid concentration data before the start of treatment regarding the concentration value of the evaluation target amino acid before the treatment by the therapy, and the amino acid after the start of treatment regarding the concentration value of the evaluation target amino acid after the start of the treatment Based on the concentration data and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the value of the multivariate discriminant corresponds to the evaluation result regarding the effect of the treatment on the evaluation target.
  • a discriminant value calculating step for calculating a discriminant value.
  • the cancer immunotherapy evaluation program according to the present invention is based on the discriminant value calculated in the discriminant value calculation step for the control unit to execute in the cancer immunotherapy evaluation program.
  • the method further comprises a discriminant value criterion evaluation step for evaluating the effect of the treatment.
  • a recording medium according to the present invention is a non-transitory computer-readable recording medium, and includes a programmed instruction for causing an information processing apparatus to execute the cancer immunotherapy evaluation method. To do.
  • the cancer immunotherapy evaluation system includes a cancer immunotherapy evaluation apparatus including a control unit and a storage unit, and an amino acid related to a concentration value of an amino acid to be evaluated that includes a control unit and receives treatment by cancer immunotherapy.
  • a cancer immunotherapy evaluation system configured to connect an information communication terminal device that provides concentration data to be communicable via a network, wherein the control unit of the information communication terminal device starts the treatment Pre-treatment amino acid concentration data relating to the concentration value of the evaluation target amino acid before treatment, and post-treatment amino acid concentration data relating to the concentration value of the evaluation target amino acid after initiation of the treatment, the cancer immunotherapy evaluation
  • a result receiving means for receiving a discriminant value corresponding to an evaluation result relating to the effect of the treatment on the evaluation target, wherein the control unit of the cancer immunotherapy evaluation device receives the information from the information communication terminal device.
  • Amino acid concentration data receiving means for receiving the transmitted pre-treatment amino acid concentration data and the post-treatment amino acid concentration data, and the pre-treatment amino acid concentration data and the post-treatment amino acid received by the amino acid concentration data receiving means Based on the density data and the multivariate discriminant stored in the storage unit, the discriminant value calculating means for calculating the discriminant value, and the discriminant value calculated by the discriminant value calculating means to the information communication terminal device And a result transmitting means for transmitting.
  • the cancer immunotherapy evaluation system is the cancer immunotherapy evaluation system, wherein the control unit of the cancer immunotherapy evaluation device is based on the discriminant value calculated by the discriminant value calculation means.
  • the apparatus further comprises discriminant value reference evaluation means for evaluating the effect of the treatment on the evaluation object, and the result transmitting means transmits the evaluation result of the evaluation object obtained by the discriminant value reference evaluation means to the information communication terminal device.
  • the result receiving means receives the evaluation result of the evaluation target transmitted from the cancer immunotherapy evaluation apparatus.
  • the information communication terminal device is an information communication terminal device that includes a control unit, and provides amino acid concentration data related to the concentration value of the amino acid to be evaluated that receives treatment by cancer immunotherapy, wherein the control unit includes: , Comprising a result acquisition means for acquiring a discriminant value corresponding to an evaluation result relating to the effect of the treatment on the evaluation target, which is a value of a multivariate discriminant including an amino acid concentration as a variable.
  • Amino acid concentration data before starting treatment regarding the concentration value of the amino acid to be evaluated before starting amino acid concentration data after starting treatment regarding the concentration value of the amino acid to be evaluated after starting the treatment, and multivariate discrimination It is calculated based on a formula.
  • the result acquisition unit acquires the evaluation result regarding the effect of the treatment on the evaluation target, and the evaluation result is the discriminant value. This is a result obtained by evaluating the effect of the treatment on the evaluation object.
  • the information communication terminal device is connected to the cancer immunotherapy evaluation device that stores the multivariate discriminant and calculates the discriminant value via the network in the information communication terminal device.
  • the control unit further comprises amino acid concentration data transmitting means for transmitting the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data to the cancer immunotherapy evaluation device, and the result acquisition means includes the The discriminant value transmitted from the cancer immunotherapy evaluation device is received.
  • the information communication terminal device is the information communication terminal device, wherein the cancer immunotherapy evaluation device further evaluates the effect of the treatment, and the result acquisition means includes the cancer immunotherapy evaluation device.
  • the transmitted evaluation result relating to the effect of the treatment on the evaluation target is transmitted.
  • the cancer immunotherapy evaluation apparatus is communicably connected via an information communication terminal device that provides amino acid concentration data related to the concentration value of an amino acid to be evaluated that receives treatment by cancer immunotherapy,
  • a cancer immunotherapy evaluation device including a control unit and a storage unit, wherein the control unit relates to a concentration value of the evaluation target amino acid transmitted from the information communication terminal device before the treatment is started.
  • Amino acid concentration data receiving means for receiving amino acid concentration data before starting treatment and amino acid concentration data after starting treatment related to the concentration value of the amino acid to be evaluated after the treatment is started, and received by the amino acid concentration data receiving means
  • the discriminant value calculating means calculates the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation object, which is the value of the multivariate discriminant.
  • a result transmitting means for transmitting the discriminant value calculated by the discriminant value calculating means to the information communication terminal device.
  • the cancer immunotherapy evaluation apparatus is the cancer immunotherapy evaluation apparatus according to the present invention, wherein the control unit has an effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation means. Further comprising: a discriminant value criterion-evaluating unit, wherein the result transmitting unit transmits the evaluation result of the evaluation target obtained by the discriminant value criterion-evaluating unit to the information communication terminal device. .
  • amino acid concentration data before starting treatment related to the concentration value of amino acids in blood collected before starting treatment from an evaluation subject receiving treatment by cancer immunotherapy, and treatment was started from the evaluation subject.
  • the amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
  • Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr which are included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data.
  • the effect of treatment on the evaluation target is evaluated. This produces an effect that the therapeutic effect can be accurately evaluated using the amino acid concentration useful for evaluating the therapeutic effect of cancer immunotherapy.
  • the treatment is evaluated whether the treatment is effective for the evaluation object based on the concentration value (for example, whether the treatment is effective for the evaluation object based on the concentration value). To determine).
  • the value of the multivariate discriminant is calculated. For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated.
  • the discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant.
  • the multivariate discriminant including the amino acid concentration as a variable, it is possible to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target.
  • the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment).
  • the multivariate discriminant is a logistic regression equation, a fractional equation, a linear discriminant equation, a multiple regression equation, an equation created by a support vector machine, an equation created by the Mahalanobis distance method, a canonical discriminant.
  • Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr which are included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data.
  • a discriminant value is calculated based on a multivariate discriminant including at least one of Arg, Gly, Cys2, and Pro as a variable.
  • a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. There is an effect that can be.
  • the multivariate discriminant is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable, or Trp, Thr,
  • This is a logistic regression equation including at least one of His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable.
  • This makes it possible to use a multivariate discriminant that is particularly useful for the evaluation including the concentration of amino acids that are particularly useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. There is an effect that can be obtained.
  • the effect of treatment on the evaluation target is evaluated based on the discriminant value. Accordingly, the therapeutic effect can be accurately evaluated by using the discriminant value obtained by the multivariate discriminant including the amino acid concentration as a variable.
  • the treatment is evaluated whether the treatment is effective for the evaluation target based on the discriminant value (for example, whether the treatment is effective for the evaluation target based on the discriminant value). To determine).
  • the discriminant value obtained by the multivariate discriminant useful for the evaluation for example, discrimination
  • the concentration of amino acid useful for evaluation for example, discrimination
  • the effect can be performed with high accuracy.
  • the multivariate discriminant stored in the storage unit is created based on the cancer state information stored in the storage unit including the amino acid concentration data and the cancer state index data relating to the index representing the cancer state. May be. Specifically, (i) creating a candidate multivariate discriminant based on a predetermined formula creation method from cancer state information, (ii) verifying the created candidate multivariate discriminant based on a predetermined verification method, (Iii) by selecting a variable of the candidate multivariate discriminant based on a predetermined variable selection method, selecting a combination of amino acid concentration data included in the cancer state information used when creating the candidate multivariate discriminant, (Iv) A candidate multivariate discriminant to be adopted as a multivariate discriminant from among a plurality of candidate multivariate discriminants based on the verification results accumulated by repeatedly executing (i), (ii) and (iii) A multivariate discriminant may be created by selection. Thereby, there exists an effect that the multivariate discriminant most suitable for evaluation
  • FIG. 1 is a principle configuration diagram showing the basic principle of the first embodiment.
  • FIG. 2 is a flowchart showing an example of the cancer immunotherapy evaluation method according to the first embodiment.
  • FIG. 3 is a principle configuration diagram showing the basic principle of the second embodiment.
  • FIG. 4 is a diagram illustrating an example of the overall configuration of the present system.
  • FIG. 5 is a diagram showing another example of the overall configuration of the present system.
  • FIG. 6 is a block diagram showing an example of the configuration of the cancer immunotherapy evaluation apparatus 100 of the present system.
  • FIG. 7 is a diagram illustrating an example of information stored in the user information file 106a.
  • FIG. 8 is a diagram showing an example of information stored in the amino acid concentration data file 106b.
  • FIG. 9 is a diagram illustrating an example of information stored in the cancer state information file 106c.
  • FIG. 10 is a diagram illustrating an example of information stored in the designated cancer state information file 106d.
  • FIG. 11 is a diagram illustrating an example of information stored in the candidate multivariate discriminant file 106e1.
  • FIG. 12 is a diagram illustrating an example of information stored in the verification result file 106e2.
  • FIG. 13 is a diagram illustrating an example of information stored in the selected cancer state information file 106e3.
  • FIG. 14 is a diagram illustrating an example of information stored in the multivariate discriminant file 106e4.
  • FIG. 15 is a diagram illustrating an example of information stored in the discrimination value file 106f.
  • FIG. 16 is a diagram illustrating an example of information stored in the evaluation result file 106g.
  • FIG. 17 is a block diagram showing a configuration of the multivariate discriminant-preparing part 102h.
  • FIG. 18 is a block diagram illustrating a configuration of the discriminant value criterion-evaluating unit 102j.
  • FIG. 19 is a block diagram illustrating an example of the configuration of the client apparatus 200 of the present system.
  • FIG. 20 is a block diagram showing an example of the configuration of the database apparatus 400 of this system.
  • FIG. 21 is a flowchart showing an example of a cancer immunotherapy evaluation service process performed in the present system.
  • FIG. 22 is a flowchart showing an example of multivariate discriminant creation processing performed by the cancer immunotherapy evaluation apparatus 100 of the present system.
  • FIG. 23 is a diagram showing an experimental protocol of Example 1.
  • FIG. 24 shows changes in responder tumor growth relative to immunotherapy.
  • FIG. 25 shows changes in tumor growth of non-responders with respect to immunotherapy.
  • FIG. 26 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in the responder.
  • FIG. 27 is a diagram showing the distribution of amino acid variables between the two groups before and after the start of treatment in the responder.
  • FIG. 28 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder.
  • FIG. 29 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder.
  • FIG. 24 shows changes in responder tumor growth relative to immunotherapy.
  • FIG. 25 shows changes in tumor growth of non-responders with respect to immunotherapy.
  • FIG. 26 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in the responder.
  • FIG. 30 is a radar chart showing the distribution of amino acid variables after the start of treatment in the responder.
  • FIG. 31 is a radar chart showing the distribution of amino acid variables after the start of treatment in a non-responder.
  • FIG. 32 is a diagram showing the AUC of the ROC curve of each amino acid variable.
  • FIG. 33 is a diagram showing the AUC of the ROC curve of each amino acid variable.
  • FIG. 34 is a diagram showing a list of fractional expressions.
  • FIG. 35 is a diagram showing the appearance frequency of amino acid variables included in the fractional expression given in FIG.
  • FIG. 36 is a diagram showing a list of logistic regression equations.
  • FIG. 37 is a diagram showing a list of logistic regression equations.
  • FIG. 38 is a diagram showing a list of logistic regression equations.
  • FIG. 39 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations listed in FIGS. 36, 37 and 38.
  • FIG. 40 is a diagram showing a list of logistic regression equations.
  • FIG. 41 is a diagram showing a list of logistic regression equations.
  • FIG. 42 is a diagram showing a list of logistic regression equations.
  • FIG. 43 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS. 40, 41, and 42.
  • FIG. 44 is a diagram showing a list of logistic regression equations.
  • FIG. 45 is a diagram showing a list of logistic regression equations.
  • FIG. 46 is a diagram showing a list of logistic regression equations.
  • FIG. 47 is a diagram showing a list of logistic regression equations.
  • FIG. 48 is a diagram showing a list of logistic regression equations.
  • FIG. 49 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS. 44-48.
  • FIG. 50 is a diagram showing a list of logistic regression equations.
  • FIG. 51 is a diagram showing a list of logistic regression equations.
  • FIG. 52 is a diagram showing a list of logistic regression equations.
  • FIG. 53 is a diagram showing a list of logistic regression equations.
  • FIG. 54 is a diagram showing a list of logistic regression equations.
  • FIG. 55 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS.
  • FIG. 56 is a diagram showing the experimental protocol of Example 6.
  • FIG. 57 is a diagram showing a section image of a tumor tissue.
  • FIG. 58 is a diagram showing the distribution of amino acid variables between the two groups before and after the start of treatment in the responder.
  • FIG. 59 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a responder.
  • FIG. 60 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder.
  • FIG. 61 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder.
  • FIG. 62 is a radar chart showing the distribution of amino acid variables after the start of treatment in the responder.
  • FIG. 63 is a radar chart showing the distribution of amino acid variables after the start of treatment in a non-responder.
  • FIG. 64 is a diagram showing the AUC of the ROC curve of each amino acid variable.
  • FIG. 65 is a diagram showing the AUC of the ROC curve of each amino acid variable.
  • FIG. 66 is a diagram showing a list of fractional expressions.
  • FIG. 67 is a diagram showing the appearance frequency of amino acid variables included in the fractional expression given in FIG.
  • FIG. 68 is a diagram showing a list of logistic regression equations.
  • FIG. 69 is a diagram showing a list of logistic regression equations.
  • FIG. 70 is a diagram showing a list of logistic regression equations.
  • 71 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations listed in FIGS. 68, 69, and 70.
  • FIG. 72 is a diagram showing a list of logistic regression equations.
  • FIG. 73 is a diagram showing a list of logistic regression equations.
  • FIG. 74 is a diagram showing a list of logistic regression equations.
  • FIG. 75 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations given in FIGS. 72, 73 and 74.
  • FIG. 76 is a diagram showing a list of logistic regression equations.
  • FIG. 77 is a diagram showing a list of logistic regression equations.
  • FIG. 78 is a diagram showing a list of logistic regression equations.
  • FIG. 79 is a diagram showing a list of logistic regression equations.
  • FIG. 80 is a diagram showing a list of logistic regression equations.
  • FIG. 80 is a diagram showing a list of logistic regression equations.
  • FIG. 81 is a diagram showing a list of logistic regression equations.
  • FIG. 82 is a diagram showing a list of logistic regression equations.
  • FIG. 83 is a diagram showing a list of logistic regression equations.
  • FIG. 84 is a diagram showing a list of logistic regression equations.
  • FIG. 85 is a diagram showing a list of logistic regression equations.
  • Embodiments of cancer immunotherapy evaluation method (first embodiment), cancer immunotherapy evaluation apparatus, cancer immunotherapy evaluation method, cancer immunotherapy evaluation program, recording medium, cancer immunotherapy evaluation system, and information communication terminal
  • An apparatus embodiment (second embodiment) will be described in detail with reference to the drawings. Note that the present invention is not limited to these embodiments.
  • FIG. 1 is a principle configuration diagram showing the basic principle of the first embodiment.
  • the amino acid concentration data after the start of treatment regarding the concentration value of the amino acid in the blood collected after the start of the treatment from the evaluation target is acquired (step S11).
  • amino acid concentration data measured by a company or the like that performs amino acid concentration measurement may be acquired.
  • the following (A) or (B) may be obtained from blood collected from an evaluation target.
  • Amino acid concentration data may be obtained by measuring amino acid concentration data by a measurement method.
  • the unit of amino acid concentration may be obtained by, for example, molar concentration, weight concentration, or by adding / subtracting / subtracting an arbitrary constant to / from these concentrations.
  • Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at ⁇ 80 ° C. until the measurement of amino acid concentration.
  • acetonitrile was added to remove protein, followed by precolumn derivatization using a labeling reagent (3-aminopyridyl-N-hydroxysuccinimidyl carbamate), and liquid chromatography mass spectrometry The amino acid concentration was analyzed by a total (LC / MS) (see International Publication No.
  • step S12 based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data acquired in step S11, the effect of treatment by cancer immunotherapy on the evaluation target is evaluated (step S12).
  • the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data to be evaluated are acquired, and evaluation is performed based on the acquired pre-treatment amino acid concentration data and post-treatment amino acid concentration data.
  • Evaluate the effect of treatment on the subject in short, provide information to assess the effect of treatment on the subject to be evaluated).
  • the amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
  • step S12 data such as missing values and outliers may be removed from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data acquired in step S11. Thereby, the therapeutic effect of cancer immunotherapy can be accurately evaluated.
  • the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, and Pro
  • the effect of treatment on the evaluation target may be evaluated based on at least one concentration value.
  • the therapeutic effect can be accurately evaluated using the amino acid concentration useful for evaluating the therapeutic effect of cancer immunotherapy.
  • the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, and Pro, whether or not the treatment is effective may be evaluated based on the concentration value. For example, it is determined whether or not the treatment is effective, and the evaluation target is assigned to any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It may be classified. Thereby, the said evaluation (for example, discrimination, classification, etc.) can be accurately performed using the amino acid concentration useful for evaluation (for example, discrimination, classification, etc.) regarding the effectiveness of the therapeutic effect of cancer immunotherapy.
  • the range that the density value can take is a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • a predetermined range for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • an arbitrary value is added / subtracted / multiplied / divided with respect to the density value so as to fall within a range from 0 to 10.0, or the density value is converted into a predetermined conversion method (for example, exponential conversion, logarithmic conversion,
  • the density value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, reciprocal conversion, or the like, or by combining these calculations with respect to the density value.
  • the value of the exponential function with the concentration value as the index and the Napier number as the base (specifically, the natural logarithm ln (p / (1-p)) when defining the probability p that the treatment is effective is the concentration value.
  • the value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value (specifically, The value of the probability p) may be further calculated.
  • the density value may be converted so that the value after conversion under a specific condition becomes a specific value. For example, the density value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
  • step S12 based on the pre-treatment amino acid concentration data and post-treatment amino acid concentration data obtained in step S11, and a preset multivariate discriminant including the amino acid concentration as a variable, the multivariate discriminant And the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation target may be calculated, and the effect of the treatment on the evaluation target may be evaluated based on the calculated discriminant value. For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated.
  • the discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant.
  • a multivariate discriminant including the amino acid concentration as a variable can be used to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target, or the obtained discriminant value can be used to obtain cancer. It is possible to accurately evaluate the therapeutic effect of immunotherapy.
  • the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment). Thereby, the said therapeutic effect can be continuously monitored even after completion
  • Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used. This makes it possible to obtain a discriminant value corresponding to the evaluation result related to the therapeutic effect on the evaluation target using the multivariate discriminant useful for evaluating the therapeutic effect of cancer immunotherapy, or to use the obtained discriminant value. Thus, the therapeutic effect can be accurately evaluated.
  • the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met , Lys, Arg, Gly, Cys2, and Pro, the discriminant value may be calculated based on a multivariate discriminant including at least one as a variable, and the treatment for the evaluation target is further performed based on the calculated discriminant value. You may evaluate the effect.
  • a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. Or the obtained discrimination value can be used to accurately evaluate the therapeutic effect.
  • the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met , Lys, Arg, Gly, Cys2, and Pro, the discriminant value may be calculated based on a multivariate discriminant including at least one as a variable, and further, based on the calculated discriminant value, To assess whether the treatment is effective.
  • the treatment is effective for the evaluation target, and any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It is also possible to classify the evaluation target.
  • This makes it possible to use a multivariate discriminant useful for the evaluation (for example, discrimination, classification, etc.) including the concentration of amino acids useful for evaluation (for example, discrimination, classification, etc.) as a variable regarding the effectiveness of the therapeutic effect of cancer immunotherapy.
  • a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target, or to accurately perform the evaluation (for example, discrimination, classification, etc.) using the obtained discriminant value.
  • the multivariate discriminant used in the evaluation is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable. Or a logistic regression equation including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable.
  • This makes it possible to use a multivariate discriminant that is particularly useful for evaluating the effectiveness of the therapeutic effect of cancer immunotherapy (for example, discrimination, classification, etc.), and to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target.
  • the evaluation (for example, discrimination, classification, etc.) can be performed with higher accuracy by using the obtained discrimination value.
  • the discriminant value can have a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • a predetermined range for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • an arbitrary value is added / subtracted / multiplied / divided with respect to the discriminant value so that the discriminant value falls within a range from 0 to 10.0, etc.
  • the discriminant value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, or reciprocal conversion, or by combining these calculations with respect to the discriminant value.
  • the value of the exponential function with the discriminant value as the exponent and the Napier number as the base is the discriminant value.
  • the value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value may be further calculated.
  • the discriminant value may be converted so that the value after conversion under a specific condition becomes a specific value.
  • the discriminant value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
  • the discriminant value in this specification may be the value of the multivariate discriminant itself, or may be a value after converting the value of the multivariate discriminant.
  • the treatment method to be evaluated can be selected before treatment.
  • a treatment method to be received by the evaluation object may be predicted before treatment.
  • each multivariate discriminant described above is described in, for example, the method described in International Publication No. 2004/052191 which is an international application by the present applicant or International Publication No. 2006/098192 which is an international application by the present applicant. You may create by the method of description. If the multivariate discriminant obtained by these methods is used, the multivariate discriminant is suitable for evaluating the therapeutic effect of cancer immunotherapy regardless of the unit of amino acid concentration in the amino acid concentration data as input data. Can be used.
  • the multivariate discriminant means a formula format generally used in multivariate analysis. For example, a fractional equation, multiple regression equation, multiple logistic regression equation, linear discriminant, Mahalanobis distance, canonical discriminant function, support vector Includes machines, decision trees, etc. Also included are expressions as indicated by the sum of different forms of multivariate discriminants. In addition, in multiple regression equations, multiple logistic regression equations, canonical discriminant functions, etc., coefficients and constant terms are added to each variable.
  • the coefficients and constant terms are preferably real numbers, more preferably Values belonging to the 99% confidence interval range of the coefficient and constant term obtained for discrimination from the data, more preferably within the 95% confidence interval range of the coefficient and constant term obtained from the data It doesn't matter if it belongs. Further, the value of each coefficient and its confidence interval may be obtained by multiplying it by a real number, and the value of the constant term and its confidence interval may be obtained by adding / subtracting / multiplying / dividing an arbitrary real constant thereto.
  • linear transformation addition of constants, constant multiplication
  • monotonic increase (decrease) conversion eg logit transformation
  • the fractional expression means that the numerator of the fractional expression is represented by the sum of amino acids A, B, C,... And / or the denominator of the fractional expression is the sum of amino acids a, b, c,. It is represented by
  • the fractional expression includes a sum of fractional expressions ⁇ , ⁇ , ⁇ ,.
  • the fractional expression also includes a divided fractional expression.
  • An appropriate coefficient may be added to each amino acid used in the numerator and denominator.
  • amino acids used in the numerator and denominator may overlap.
  • an appropriate coefficient may be attached to each fractional expression.
  • the value of the coefficient of each variable and the value of the constant term may be real numbers.
  • the combination of the numerator variable and the denominator variable is generally reversed in the sign of the correlation with the target variable, but since the correlation is maintained, it can be considered equivalent in discriminability. Combinations of swapping numerator and denominator variables are also included.
  • the first embodiment when evaluating the therapeutic effect of cancer immunotherapy, in addition to the concentration of amino acids, other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, immunocompetent intracellular cytokine, A delayed excessive reaction (DTH) or the like may be further used.
  • other biological information eg, tumor marker, blood cytokine, immunocompetent cell
  • Number immunocompetent intracellular cytokines, delayed hyperfractionation (DTH), etc.
  • DTH delayed hyperfractionation
  • FIG. 2 is a flowchart showing an example of the cancer immunotherapy evaluation method according to the first embodiment.
  • amino acid concentration data before starting treatment and amino acid concentration data after starting treatment of an individual (for example, an animal or a human) who are treated by cancer immunotherapy are acquired (step SA11).
  • amino acid concentration data measured by a company or the like that measures amino acid concentration may be acquired, and a measuring method such as (A) or (B) described above from blood collected from an individual.
  • the amino acid concentration data may be obtained by measuring the amino acid concentration data.
  • step SA12 data such as missing values and outliers are removed from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data obtained in step SA11 (step SA12).
  • Val, Ile, Leu, His, Phe, Trp, Gln Val, Ile, Leu, His, Phe, Trp, Gln included in the pre-treatment amino acid concentration data and post-treatment amino acid concentration data of the individual from which data such as missing values and outliers have been removed in step SA12.
  • treatment by cancer immunotherapy is effective for an individual based on the concentration value of at least one of Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro Is determined (step SA13).
  • FIG. 3 is a principle configuration diagram showing the basic principle of the second embodiment.
  • control unit determines the amino acid concentration data before the start of treatment of the evaluation target (for example, an individual such as an animal or a human) to be treated by cancer immunotherapy, the amino acid concentration data before the start of treatment, and the amino acid after the start of the treatment of the evaluation target. Based on the amino acid concentration data after the start of treatment related to the concentration value of the substance and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the value of the multivariate discriminant and the effect of the treatment on the evaluation target A discriminant value corresponding to the evaluation result is calculated (step S21).
  • the evaluation target for example, an individual such as an animal or a human
  • control unit evaluates the effect of treatment by cancer immunotherapy on the evaluation target based on the discriminant value calculated in step S21 (step S22).
  • the discriminant value corresponding to the evaluation result regarding the effect of the treatment on the evaluation target. And the effect of the treatment on the evaluation object is evaluated based on the calculated discriminant value (in short, information for evaluating the effect of the treatment on the evaluation object is provided). For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated.
  • the discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant.
  • a multivariate discriminant including the amino acid concentration as a variable can be used to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target, or the obtained discriminant value can be used to obtain cancer. It is possible to accurately evaluate the therapeutic effect of immunotherapy. Further, the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment). Thereby, the said therapeutic effect can be continuously monitored even after completion
  • Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used. This makes it possible to obtain a discriminant value corresponding to the evaluation result related to the therapeutic effect on the evaluation target using the multivariate discriminant useful for evaluating the therapeutic effect of cancer immunotherapy, or to use the obtained discriminant value. Thus, the therapeutic effect can be accurately evaluated.
  • step S21 Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, and the amino acid concentration data before and after the treatment start are included. At least one concentration value of Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, A discriminant value is calculated based on a multivariate discriminant including at least one of Gly, Cys2, and Pro as a variable.
  • step S22 the effect of treatment on the evaluation target is calculated based on the discriminant value calculated in step S21. You may evaluate.
  • a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. Or the obtained discrimination value can be used to accurately evaluate the therapeutic effect.
  • step S21 Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, and the amino acid concentration data before and after the treatment start are included. At least one concentration value of Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, A discriminant value is calculated based on a multivariate discriminant including at least one of Gly, Cys2, and Pro as a variable.
  • step S22 treatment is performed on the evaluation target based on the discriminant value calculated in step S21. You may evaluate whether it is effective.
  • the treatment is effective for the evaluation target, and any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It is also possible to classify the evaluation target.
  • This makes it possible to use a multivariate discriminant useful for the evaluation (for example, discrimination, classification, etc.) including the concentration of amino acids useful for evaluation (for example, discrimination, classification, etc.) as a variable regarding the effectiveness of the therapeutic effect of cancer immunotherapy.
  • a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target, or to accurately perform the evaluation (for example, discrimination, classification, etc.) using the obtained discriminant value.
  • the multivariate discriminant used in the evaluation is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable. Or a logistic regression equation including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable.
  • This makes it possible to use a multivariate discriminant that is particularly useful for evaluating the effectiveness of the therapeutic effect of cancer immunotherapy (for example, discrimination, classification, etc.), and to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target.
  • the evaluation (for example, discrimination, classification, etc.) can be performed with higher accuracy by using the obtained discrimination value.
  • the discriminant value can have a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • a predetermined range for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or ⁇ 10.
  • an arbitrary value is added / subtracted / multiplied / divided with respect to the discriminant value so that the discriminant value falls within a range from 0 to 10.0, etc.
  • the discriminant value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, or reciprocal conversion, or by combining these calculations with respect to the discriminant value.
  • the value of the exponential function with the discriminant value as the exponent and the Napier number as the base is the discriminant value.
  • the value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value may be further calculated.
  • the discriminant value may be converted so that the value after conversion under a specific condition becomes a specific value.
  • the discriminant value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
  • the discriminant value in this specification may be the value of the multivariate discriminant itself, or may be a value after converting the value of the multivariate discriminant.
  • the treatment method to be evaluated can be selected before treatment.
  • a treatment method to be received by the evaluation object may be predicted before treatment.
  • each multivariate discriminant described above is described in, for example, the method described in International Publication No. 2004/052191 which is an international application by the present applicant or International Publication No. 2006/098192 which is an international application by the present applicant. You may create by the method of description. If the multivariate discriminant obtained by these methods is used, the multivariate discriminant is suitable for evaluating the therapeutic effect of cancer immunotherapy regardless of the unit of amino acid concentration in the amino acid concentration data as input data. Can be used.
  • the multivariate discriminant means a formula format generally used in multivariate analysis. For example, a fractional equation, multiple regression equation, multiple logistic regression equation, linear discriminant, Mahalanobis distance, canonical discriminant function, support vector Includes machines, decision trees, etc. Also included are expressions as indicated by the sum of different forms of multivariate discriminants. In addition, in multiple regression equations, multiple logistic regression equations, canonical discriminant functions, etc., coefficients and constant terms are added to each variable.
  • the coefficients and constant terms are preferably real numbers, more preferably Values belonging to the 99% confidence interval range of the coefficient and constant term obtained for discrimination from the data, more preferably within the 95% confidence interval range of the coefficient and constant term obtained from the data It doesn't matter if it belongs. Further, the value of each coefficient and its confidence interval may be obtained by multiplying it by a real number, and the value of the constant term and its confidence interval may be obtained by adding / subtracting / multiplying / dividing an arbitrary real constant thereto.
  • linear transformation addition of constants, constant multiplication
  • monotonic increase (decrease) conversion eg logit transformation
  • the fractional expression means that the numerator of the fractional expression is represented by the sum of amino acids A, B, C,... And / or the denominator of the fractional expression is the sum of amino acids a, b, c,. It is represented by
  • the fractional expression includes a sum of fractional expressions ⁇ , ⁇ , ⁇ ,.
  • the fractional expression also includes a divided fractional expression.
  • An appropriate coefficient may be added to each amino acid used in the numerator and denominator.
  • amino acids used in the numerator and denominator may overlap.
  • an appropriate coefficient may be attached to each fractional expression.
  • the value of the coefficient of each variable and the value of the constant term may be real numbers.
  • the combination of the numerator variable and the denominator variable is generally reversed in the sign of the correlation with the target variable, but since the correlation is maintained, it can be considered equivalent in discriminability. Combinations of swapping numerator and denominator variables are also included.
  • the second embodiment when evaluating the therapeutic effect of cancer immunotherapy, in addition to the concentration of amino acids, other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, immunocompetent intracellular cytokine, A delayed excessive reaction (DTH) or the like may be further used.
  • other biological information eg, tumor marker, blood cytokine, immunocompetent cell
  • Number immunocompetent intracellular cytokines, delayed hyperfractionation (DTH), etc.
  • DTH delayed hyperfractionation
  • step 1 to step 4 the outline of the multivariate discriminant creation process (step 1 to step 4) will be described in detail. Note that the processing described here is merely an example, and the method of creating the multivariate discriminant is not limited to this.
  • the control unit determines amino acid concentration data (for example, data relating to amino acid concentration or data relating to the amount of change in amino acid concentration) and cancer state index data relating to an index (for example, tumor size) indicating a cancer state.
  • amino acid concentration data for example, data relating to amino acid concentration or data relating to the amount of change in amino acid concentration
  • cancer state index data relating to an index (for example, tumor size) indicating a cancer state.
  • an index for example, tumor size
  • y cancer state index data
  • x i amino acid concentration data
  • Step 1 a plurality of different formula creation methods (principal component analysis, discriminant analysis, support vector machine, multiple regression analysis, logistic regression analysis, k-means method, cluster analysis, decision tree, etc.) are obtained from cancer status information.
  • a plurality of candidate multivariate discriminants may be created by using the above in combination. Specifically, for cancer status information that is multivariate data composed of amino acid concentration data and cancer status index data obtained by analyzing blood obtained from a large number of pre-treatment groups and a large number of post-treatment groups.
  • a plurality of groups of candidate multivariate discriminants may be created concurrently using a plurality of different algorithms. For example, two different candidate multivariate discriminants may be created by performing discriminant analysis and logistic regression analysis simultaneously using different algorithms.
  • the candidate multivariate discriminant is created by converting the cancer state information using the candidate multivariate discriminant created by performing the principal component analysis and performing the discriminant analysis on the converted cancer state information Good. Thereby, finally, an appropriate multivariate discriminant suitable for the evaluation condition can be created.
  • the candidate multivariate discriminant prepared using principal component analysis is a linear expression including each amino acid variable that maximizes the variance of all amino acid concentration data.
  • the candidate multivariate discriminant created using discriminant analysis is a high-order formula (exponential or exponential) including each amino acid variable that minimizes the ratio of the sum of variances within each group to the variance of all amino acid concentration data. Including logarithm).
  • the candidate multivariate discriminant created using the support vector machine is a higher-order formula (including a kernel function) including each amino acid variable that maximizes the boundary between groups.
  • the candidate multivariate discriminant created using multiple regression analysis is a high-order expression including each amino acid variable that minimizes the sum of distances from all amino acid concentration data.
  • a candidate multivariate discriminant created using logistic regression analysis is a linear model representing the log odds of probability, and is a linear expression including each amino acid variable that maximizes the likelihood of the probability.
  • the k-means method searches k neighborhoods of each amino acid concentration data, defines the largest group among the groups to which the neighboring points belong as the group to which the data belongs, This is a method of selecting an amino acid variable that best matches the group to which the group belongs.
  • Cluster analysis is a method of clustering (grouping) points that are closest to each other in all amino acid concentration data.
  • the decision tree is a technique for predicting a group of amino acid concentration data based on patterns that can be taken by amino acid variables having higher ranks by adding ranks to amino acid variables.
  • control unit verifies (mutually verifies) the candidate multivariate discriminant created in step 1 based on a predetermined verification method (step 2).
  • the candidate multivariate discriminant is verified for each candidate multivariate discriminant created in step 1.
  • step 2 the discrimination rate, sensitivity, specificity, etc. of the candidate multivariate discriminant based on at least one of random sampling method, bootstrap method, holdout method, N-fold method, leave one out method, etc.
  • the verification may be performed with respect to at least one of the information criterion, ROC_AUC (area under the curve of the receiver characteristic curve), and the like.
  • the discrimination rate is a ratio in which the result of the therapeutic effect evaluated in the present embodiment is negative as a true state and is evaluated as negative correctly, and a positive result as a true state is correctly evaluated as positive. is there.
  • Sensitivity is the ratio at which positive results are positively evaluated as positive as the result of the therapeutic effect evaluated in this embodiment.
  • specificity is a ratio at which negative results are correctly evaluated as negative as the true result of the therapeutic effect evaluated in the present embodiment.
  • the information criterion is the difference between the number of amino acid variables of the candidate multivariate discriminant created in step 1, the result of the treatment effect evaluated in this embodiment and the result of the treatment effect described in the input data, Are added together.
  • ROC_AUC area under the curve of the receiver characteristic curve
  • ROC receiver characteristic curve
  • the value of ROC_AUC is 1 in complete discrimination, and the closer this value is to 1, the higher the discriminability.
  • the predictability is an average of the discrimination rate, sensitivity, and specificity obtained by repeating the verification of the candidate multivariate discriminant.
  • Robustness is the variance of discrimination rate, sensitivity, and specificity obtained by repeating verification of candidate multivariate discriminants.
  • the control unit selects cancer candidate variable variables based on a predetermined variable selection method, and thus cancer state information used when creating a candidate multivariate discriminant A combination of amino acid concentration data contained in is selected (step 3).
  • the selection of amino acid variables may be performed for each candidate multivariate discriminant created in step 1. Thereby, the amino acid variable of a candidate multivariate discriminant can be selected appropriately.
  • Step 1 is executed again using the cancer state information including the amino acid concentration data selected in Step 3.
  • step 3 the amino acid variable of the candidate multivariate discriminant may be selected from the verification result in step 2 based on at least one of stepwise method, best path method, neighborhood search method, and genetic algorithm. .
  • the best path method is a method of selecting amino acid variables by sequentially reducing amino acid variables included in the candidate multivariate discriminant one by one and optimizing the evaluation index given by the candidate multivariate discriminant. is there.
  • the control unit repeatedly executes the above-described step 1, step 2 and step 3, and based on the verification results accumulated thereby, the control unit can select from a plurality of candidate multivariate discriminants.
  • a multivariate discriminant is created by selecting candidate multivariate discriminants to be adopted as the multivariate discriminant (step 4).
  • candidate multivariate discriminants for example, selecting the optimal one from among candidate multivariate discriminants created by the same formula creation method, and selecting the optimum from all candidate multivariate discriminants Sometimes there is a choice.
  • the multivariate discriminant creation process processing related to the creation of a candidate multivariate discriminant, verification of the candidate multivariate discriminant, and selection of a variable of the candidate multivariate discriminant based on the cancer state information.
  • systematization systematization
  • the amino acid concentration is used for multivariate statistical analysis, and the variable selection method and cross-validation are combined in order to select the optimal and robust variable set. Extract the variable discriminant.
  • logistic regression, linear discrimination, support vector machine, Mahalanobis distance method, multiple regression analysis, cluster analysis, decision tree, and the like can be used.
  • FIG. 4 is a diagram showing an example of the overall configuration of the present system.
  • FIG. 5 is a diagram showing another example of the overall configuration of the present system.
  • the present system includes a cancer immunotherapy evaluation apparatus 100 that evaluates the effect of treatment on an evaluation target that receives treatment by cancer immunotherapy, and a client that provides amino acid concentration data relating to the concentration value of the amino acid to be evaluated.
  • the apparatus 200 (corresponding to the information communication terminal apparatus of the present invention) is configured to be communicably connected via the network 300.
  • this system uses cancer status information used when creating a multivariate discriminant in the cancer immunotherapy evaluation apparatus 100
  • a database apparatus 400 storing a multivariate discriminant used for evaluating the therapeutic effect of immunotherapy may be configured to be communicably connected via the network 300. Accordingly, cancer in the treatment by cancer immunotherapy from the cancer immunotherapy evaluation apparatus 100 to the client apparatus 200 or the database apparatus 400, or from the client apparatus 200 or database apparatus 400 to the cancer immunotherapy evaluation apparatus 100 via the network 300. Information about the state of the is provided.
  • the information on the cancer state in the treatment with cancer immunotherapy is information on the value measured for a specific item related to the cancer state of an organism including humans in the treatment with cancer immunotherapy (for example, presence or absence of therapeutic effect) Or a difference value of tumor size).
  • information related to cancer status in cancer immunotherapy treatment is generated by the cancer immunotherapy evaluation apparatus 100, the client apparatus 200, and other apparatuses (for example, various measurement apparatuses), and is mainly stored in the database apparatus 400.
  • FIG. 6 is a block diagram showing an example of the configuration of the cancer immunotherapy evaluation apparatus 100 of the present system, and conceptually shows only the portion related to the present invention in the configuration.
  • the cancer immunotherapy evaluation apparatus 100 includes a control unit 102 such as a CPU that comprehensively controls the cancer immunotherapy evaluation apparatus, a communication device such as a router, and a wired or wireless communication line such as a dedicated line.
  • a communication interface unit 104 that connects the therapy evaluation device to the network 300 so as to be communicable, a storage unit 106 that stores various databases, tables, files, and the like, and an input / output interface unit 108 that connects to the input device 112 and the output device 114 These parts are connected to be communicable via an arbitrary communication path.
  • the cancer immunotherapy evaluation apparatus 100 may be configured in the same housing as various analysis apparatuses (for example, an amino acid analyzer or the like).
  • the storage unit 106 is a storage means, and for example, a memory device such as a RAM / ROM, a fixed disk device such as a hard disk, a flexible disk, an optical disk, or the like can be used.
  • the storage unit 106 stores a computer program for giving instructions to the CPU and performing various processes in cooperation with an OS (Operating System).
  • the storage unit 106 includes a user information file 106a, an amino acid concentration data file 106b, a cancer state information file 106c, a designated cancer state information file 106d, a multivariate discriminant-related information database 106e, and a discriminant value.
  • a file 106f and an evaluation result file 106g are stored.
  • the user information file 106a stores user information related to users.
  • FIG. 7 is a diagram illustrating an example of information stored in the user information file 106a.
  • the information stored in the user information file 106a includes a user ID for uniquely identifying a user and authentication for whether or not the user is a valid person.
  • the amino acid concentration data file 106b stores amino acid concentration data before starting treatment and amino acid concentration data after starting treatment.
  • FIG. 8 is a diagram showing an example of information stored in the amino acid concentration data file 106b.
  • the information stored in the amino acid concentration data file 106b includes an individual number for uniquely identifying an individual (sample) to be evaluated, amino acid concentration data before starting treatment, and amino acid concentration after starting treatment. It is configured to correlate with data.
  • the amino acid concentration data is treated as a numerical value, that is, a continuous scale, but the amino acid concentration data may be a nominal scale or an order scale. In the case of a nominal scale or an order scale, analysis may be performed by giving an arbitrary numerical value to each state.
  • amino acid concentration data may be combined with other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, cytokine in immunocompetent cell, delayed excessive reaction (DTH), etc.).
  • other biological information eg, tumor marker, blood cytokine, number of immuno
  • the cancer state information file 106 c stores cancer state information used when creating a multivariate discriminant.
  • FIG. 9 is a diagram illustrating an example of information stored in the cancer state information file 106c.
  • the information stored in the cancer state information file 106c includes cancer state index data relating to individual numbers and indices (index T 1 , index T 2 , index T 3 ...) Representing the cancer state. (T) and amino acid concentration data are associated with each other.
  • the cancer state index data and the amino acid concentration data are treated as numerical values (that is, a continuous scale), but the cancer state index data and the amino acid concentration data may be a nominal scale or an order scale. In the case of a nominal scale or an order scale, analysis may be performed by giving an arbitrary numerical value to each state.
  • the cancer state index data may be a known single state index serving as a marker of cancer state, or numerical data may be used.
  • the designated cancer state information file 106d stores the cancer state information designated by the cancer state information designation unit 102g described later.
  • FIG. 10 is a diagram illustrating an example of information stored in the designated cancer state information file 106d. As shown in FIG. 10, the information stored in the designated cancer state information file 106d is configured by associating individual numbers, designated cancer state index data, and designated amino acid concentration data with each other.
  • the multivariate discriminant-related information database 106e includes a candidate multivariate discriminant file 106e1 for storing the candidate multivariate discriminant created by the candidate multivariate discriminant-preparing part 102h1, which will be described later, and a candidate multivariate discriminant described later.
  • a verification result file 106e2 for storing a verification result in the discriminant verification unit 102h2
  • a selected cancer state information file 106e3 for storing cancer state information including a combination of amino acid concentration data selected by a variable selection unit 102h3 described later, and a later-described
  • a multivariate discriminant file 106e4 that stores the multivariate discriminant created by the multivariate discriminant creation unit 102h.
  • the candidate multivariate discriminant file 106e1 stores the candidate multivariate discriminant created by the candidate multivariate discriminant creation unit 102h1 described later.
  • FIG. 11 is a diagram illustrating an example of information stored in the candidate multivariate discriminant file 106e1.
  • information stored in the candidate multivariate discriminant file 106e1 includes a rank, a candidate multivariate discriminant (in FIG. 11, F 1 (Gly, Leu, Phe,%)) And F 2. (Gly, Leu, Phe,%), F 3 (Gly, Leu, Phe,...)) Are associated with each other.
  • FIG. 12 is a diagram illustrating an example of information stored in the verification result file 106e2.
  • the information stored in the verification result file 106e2 includes rank, candidate multivariate discriminant (in FIG. 12, F k (Gly, Leu, Phe,%) And F m (Gly, Le, Phe,%), Fl (Gly, Leu, Phe, etc) And the verification results of each candidate multivariate discriminant (for example, the evaluation value of each candidate multivariate discriminant). They are related to each other.
  • the selected cancer state information file 106e3 stores cancer state information including a combination of amino acid concentration data corresponding to variables selected by the variable selection unit 102h3 described later.
  • FIG. 13 is a diagram illustrating an example of information stored in the selected cancer state information file 106e3. As shown in FIG. 13, the information stored in the selected cancer state information file 106e3 is selected by an individual number, cancer state index data designated by a cancer state information designation unit 102g described later, and a variable selection unit 102h3 described later. The amino acid concentration data is associated with each other.
  • the multivariate discriminant file 106e4 stores the multivariate discriminant created by the multivariate discriminant-preparing part 102h described later.
  • FIG. 14 is a diagram illustrating an example of information stored in the multivariate discriminant file 106e4.
  • the information stored in the multivariate discriminant file 106e4 includes the rank, the multivariate discriminant (in FIG. 14, F p (Phe,%) And F p (Gly, Leu, Phe). ), F k (Gly, Leu, Phe,...)), A threshold corresponding to each formula creation method, a verification result of each multivariate discriminant (for example, an evaluation value of each multivariate discriminant), Are related to each other.
  • the discriminant value file 106f stores the discriminant value calculated by the discriminant value calculator 102i described later.
  • FIG. 15 is a diagram illustrating an example of information stored in the discrimination value file 106f. As shown in FIG. 15, information stored in the discriminant value file 106f includes an individual number for uniquely identifying an individual (sample) to be evaluated and a rank (for uniquely identifying a multivariate discriminant). Number) and the discriminant value are associated with each other.
  • the evaluation result file 106g stores the evaluation result in the discriminant value criterion-evaluating unit 102j described later (specifically, the discrimination result / classification result in the discriminant value criterion-discriminating unit 102j1 described later).
  • FIG. 16 is a diagram illustrating an example of information stored in the evaluation result file 106g.
  • Information stored in the evaluation result file 106g includes an individual number for uniquely identifying an individual (sample) to be evaluated, and a plurality of evaluation target amino acid concentration data (amino acid concentration data before treatment start and amino acid after treatment start).
  • Concentration data one or more discriminant values calculated by a multivariate discriminant (for example, a discriminant value before treatment start, a discriminant value after treatment start or a discriminant value before and after treatment start), and the therapeutic effect of cancer immunotherapy
  • a multivariate discriminant for example, a discriminant value before treatment start, a discriminant value after treatment start or a discriminant value before and after treatment start
  • the evaluation results related to the evaluation are associated with each other.
  • the storage unit 106 stores various types of Web data for providing the Web site to the client device 200, CGI programs, and the like as other information in addition to the information described above.
  • the Web data includes data for displaying various Web pages to be described later, and these data are formed as text files described in HTML or XML, for example.
  • a part file, a work file, and other temporary files for creating Web data are also stored in the storage unit 106.
  • the storage unit 106 stores audio for transmission to the client device 200 as an audio file such as WAVE format or AIFF format, and stores still images or moving images as image files such as JPEG format or MPEG2 format as necessary. Can be stored.
  • the communication interface unit 104 mediates communication between the cancer immunotherapy evaluation device 100 and the network 300 (or a communication device such as a router). That is, the communication interface unit 104 has a function of communicating data with other terminals via a communication line.
  • the input / output interface unit 108 is connected to the input device 112 and the output device 114.
  • a monitor including a home television
  • a speaker or a printer can be used as the output device 114 (hereinafter, the output device 114 may be described as the monitor 114).
  • the input device 112 a monitor that realizes a pointing device function in cooperation with a mouse can be used in addition to a keyboard, a mouse, and a microphone.
  • the control unit 102 has an internal memory for storing a control program such as an OS (Operating System), a program defining various processing procedures, and necessary data, and performs various information processing based on these programs. Execute. As shown in the figure, the control unit 102 is roughly divided into a request interpretation unit 102a, a browsing processing unit 102b, an authentication processing unit 102c, an email generation unit 102d, a Web page generation unit 102e, a reception unit 102f, and a cancer state information designation unit 102g.
  • a multivariate discriminant creation unit 102h, a discriminant value calculation unit 102i, a discriminant value criterion evaluation unit 102j, a result output unit 102k, and a transmission unit 102m are provided.
  • the control unit 102 removes data with missing values, removes data with many outliers, and has missing values with respect to the cancer state information transmitted from the database device 400 and the amino acid concentration data transmitted from the client device 200. Data processing such as removal of variables with a lot of data is also performed.
  • the request interpretation unit 102a interprets the request content from the client device 200 or the database device 400, and passes the processing to each unit of the control unit 102 according to the interpretation result.
  • the browsing processing unit 102b Upon receiving browsing requests for various screens from the client device 200, the browsing processing unit 102b generates and transmits Web data for these screens.
  • the authentication processing unit 102c makes an authentication determination.
  • the e-mail generation unit 102d generates an e-mail including various types of information.
  • the web page generation unit 102e generates a web page that the user browses on the client device 200.
  • the receiving unit 102 f receives information (specifically, amino acid concentration data, cancer state information, multivariate discriminant, etc.) transmitted from the client device 200 or the database device 400 via the network 300.
  • information specifically, amino acid concentration data, cancer state information, multivariate discriminant, etc.
  • the cancer state information specifying unit 102g specifies target cancer state index data and amino acid concentration data.
  • the multivariate discriminant creating unit 102h creates a multivariate discriminant based on the cancer state information received by the receiving unit 102f and the cancer state information designated by the cancer state information designating unit 102g. Specifically, the multivariate discriminant-preparing part 102h is accumulated by repeatedly executing the candidate multivariate discriminant-preparing part 102h1, the candidate multivariate discriminant-verifying part 102h2, and the variable selecting part 102h3 from the cancer state information. A multivariate discriminant is created by selecting a candidate multivariate discriminant to be adopted as a multivariate discriminant from a plurality of candidate multivariate discriminants based on the verification result.
  • the multivariate discriminant-preparing unit 102h selects a desired multivariate discriminant from the storage unit 106, A multivariate discriminant may be created.
  • the multivariate discriminant creation unit 102h creates a multivariate discriminant by selecting and downloading a desired multivariate discriminant from another computer device (for example, the database device 400) that stores the multivariate discriminant in advance. May be.
  • FIG. 17 is a block diagram showing the configuration of the multivariate discriminant-preparing part 102h, and conceptually shows only the part related to the present invention.
  • the multivariate discriminant creation unit 102h further includes a candidate multivariate discriminant creation unit 102h1, a candidate multivariate discriminant verification unit 102h2, and a variable selection unit 102h3.
  • the candidate multivariate discriminant creation unit 102h1 creates a candidate multivariate discriminant that is a candidate for the multivariate discriminant based on a predetermined formula creation method from the cancer state information.
  • the candidate multivariate discriminant-preparing part 102h1 may create a plurality of candidate multivariate discriminants from the cancer state information by using a plurality of different formula creation methods.
  • the candidate multivariate discriminant verification unit 102h2 verifies the candidate multivariate discriminant created by the candidate multivariate discriminant creation unit 102h1 based on a predetermined verification method.
  • the candidate multivariate discriminant verification unit 102h2 determines the discrimination rate of the candidate multivariate discriminant based on at least one of a random sampling method, a bootstrap method, a holdout method, an N-fold method, or a leave one out method.
  • the variable selection unit 102h3 selects a variable of the candidate multivariate discriminant based on a predetermined variable selection method, so that a combination of amino acid concentration data included in the cancer state information used when creating the candidate multivariate discriminant is selected. select. Note that the variable selection unit 102h3 may select a variable of the candidate multivariate discriminant from the verification result based on at least one of the stepwise method, the best path method, the neighborhood search method, and the genetic algorithm.
  • the discriminant value calculation unit 102 i includes the multivariate discriminant created by the multivariate discriminant creation unit 102 h and the evaluation target amino acid concentration data received by the receiving unit 102 f (specifically, the amino acid concentration before starting treatment). Based on the concentration data, amino acid concentration data after treatment start, etc., one or more discriminant values that are the values of the multivariate discriminant (specifically, discriminant value before treatment start, discriminant value after treatment start, treatment Calculate the discriminant value before and after the start.
  • Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used.
  • the discriminant value calculation unit 102i is configured to include the Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, and the like included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. At least one concentration value among Thr, Met, Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, One or more discriminant values may be calculated based on a multivariate discriminant including at least one of Lys, Arg, Gly, Cys2, and Pro as a variable.
  • the discriminant value criterion discriminating unit 102j1 to be described later discriminates whether or not the cancer immunotherapy treatment is effective for the evaluation target, and considers the degree of possibility that the treatment is effective.
  • the discriminant value calculation unit 102i uses the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment.
  • the multivariate discriminant is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable, or Trp, Thr, His, Arg, Ile,
  • a logistic regression equation including at least one of Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable may be used.
  • the discriminant value criterion-evaluating unit 102j evaluates the effect of treatment by cancer immunotherapy on the evaluation target based on one or more discriminant values calculated by the discriminant value calculating unit 102i.
  • the discriminant value criterion-evaluating unit 102j evaluates, for example, whether the treatment is effective for the evaluation target.
  • the discrimination value criterion evaluation unit 102j further includes a discrimination value criterion discrimination unit 102j1.
  • FIG. 18 is a block diagram showing the configuration of the discriminant value criterion-evaluating unit 102j, and conceptually shows only the portion related to the present invention.
  • the discriminant value criterion discriminating unit 102j1 discriminates whether or not the cancer immunotherapy treatment is effective for the evaluation target based on the one or more discriminant values calculated by the discriminant value calculating unit 102i.
  • the evaluation target is classified into any one of a plurality of categories (ranks) defined in consideration of the possibility of being effective.
  • the discriminant value criterion discriminating unit 102j1 compares the one or more discriminant values calculated by the discriminant value calculating unit 102i with one or more threshold values (cut-off values) to be evaluated. In contrast, whether cancer immunotherapy treatment is effective or not, and evaluate to any one of multiple categories (ranks) defined taking into consideration the degree of possibility that the treatment is effective Or classify objects.
  • the result output unit 102k outputs the processing results in each processing unit of the control unit 102 (evaluation results in the discrimination value criterion evaluation unit 102j (specifically, discrimination results or classification in the discrimination value criterion discrimination unit 102j1). Including the result) is output to the output device 114.
  • the transmission unit 102m transmits, for example, a discrimination value, an evaluation result (eg, a discrimination result, a classification result, etc.) to the client device 200 that is a transmission source of the amino acid concentration data to be evaluated, or a database device 400.
  • a multivariate discriminant created by the cancer immunotherapy evaluation apparatus 100, an evaluation result (for example, a discrimination result, a classification result, etc.), etc. are transmitted.
  • FIG. 19 is a block diagram showing an example of the configuration of the client apparatus 200 of the present system, and conceptually shows only the portion related to the present invention in the configuration.
  • the client device 200 includes a control unit 210, a ROM 220, an HD 230, a RAM 240, an input device 250, an output device 260, an input / output IF 270, and a communication IF 280. These units are communicably connected via an arbitrary communication path. Has been.
  • the control unit 210 includes a web browser 211, an electronic mailer 212, a reception unit 213, and a transmission unit 214.
  • the web browser 211 performs browse processing for interpreting the web data and displaying the interpreted web data on a monitor 261 described later.
  • the Web browser 211 may be plugged in with various software such as a stream player having a function of receiving, displaying, and feeding back a stream video.
  • the electronic mailer 212 transmits and receives electronic mail according to a predetermined communication protocol (for example, SMTP (Simple Mail Transfer Protocol), POP3 (Post Office Protocol version 3), etc.).
  • SMTP Simple Mail Transfer Protocol
  • POP3 Post Office Protocol version 3
  • the receiving unit 213 (corresponding to an example of the result acquisition unit of the present invention), such as a discrimination value and an evaluation result (for example, a discrimination result, a classification result, etc.) transmitted from the cancer immunotherapy evaluation apparatus 100 via the communication IF 280 Receive various information.
  • the client device has a function of acquiring various information such as a discrimination value and an evaluation result.
  • the transmission unit 214 sends various information such as amino acid concentration data to be evaluated (specifically, pre-treatment amino acid concentration data, post-treatment amino acid concentration data, etc.) to the cancer immunotherapy evaluation apparatus 100 via the communication IF 280. Send.
  • the input device 250 is a keyboard, a mouse, a microphone, or the like.
  • a monitor 261 which will be described later, also realizes a pointing device function in cooperation with the mouse.
  • the output device 260 is an output unit that outputs information received via the communication IF 280, and includes a monitor (including a home television) 261 and a printer 262. In addition, the output device 260 may be provided with a speaker or the like.
  • the input / output IF 270 is connected to the input device 250 and the output device 260.
  • the communication IF 280 connects the client device 200 and the network 300 (or a communication device such as a router) so that they can communicate with each other.
  • the client device 200 is connected to the network 300 via a communication device such as a modem, TA, or router and a telephone line, or via a dedicated line.
  • the client apparatus 200 can access the cancer immunotherapy evaluation apparatus 100 according to a predetermined communication protocol.
  • an information processing device for example, a known personal computer, workstation, home game device, Internet TV, PHS terminal, portable terminal, mobile object
  • peripheral devices such as a printer, a monitor, and an image scanner as necessary.
  • the client device 200 may be realized by installing software (including programs, data, and the like) that realizes a Web data browsing function and an e-mail function in a communication terminal / information processing terminal such as a PDA).
  • control unit 210 of the client device 200 may be realized by a CPU and a program that is interpreted and executed by the CPU and all or any part of the processing performed by the control unit 210.
  • the ROM 220 or the HD 230 stores computer programs for giving instructions to the CPU and performing various processes in cooperation with an OS (Operating System).
  • the computer program is executed by being loaded into the RAM 240, and constitutes the control unit 210 in cooperation with the CPU.
  • the computer program may be recorded in an application program server connected to the client apparatus 200 via an arbitrary network, and the client apparatus 200 may download all or a part thereof as necessary. .
  • all or any part of the processing performed by the control unit 210 may be realized by hardware such as wired logic.
  • the network 300 has a function of connecting the cancer immunotherapy evaluation apparatus 100, the client apparatus 200, and the database apparatus 400 so that they can communicate with each other, such as the Internet, an intranet, or a LAN (including both wired and wireless).
  • the network 300 includes a VAN, a personal computer communication network, a public telephone network (including both analog / digital), a dedicated line network (including both analog / digital), a CATV network, and a mobile line switching network.
  • mobile packet switching network including IMT2000 system, GSM (registered trademark) system or PDC / PDC-P system
  • wireless paging network including local wireless network such as Bluetooth (registered trademark)
  • PHS network including CS, BS or ISDB
  • satellite A communication network including CS, BS or ISDB
  • FIG. 20 is a block diagram showing an example of the configuration of the database apparatus 400 of this system, and conceptually shows only the portion related to the present invention in the configuration.
  • the database device 400 is a cancer immunotherapy evaluation device 100 or cancer state information used when creating a multivariate discriminant in the database device, a multivariate discriminant created in the cancer immunotherapy evaluation device 100, and a cancer immunotherapy evaluation device.
  • 100 has a function of storing an evaluation result (specifically, a discrimination result) obtained in 100.
  • the database device 400 includes a control unit 402 such as a CPU that comprehensively controls the database device, a communication device such as a router, and a wired or wireless communication circuit such as a dedicated line.
  • a communication interface unit 404 that connects the apparatus to the network 300 to be communicable, a storage unit 406 that stores various databases, tables, and files (for example, files for Web pages), and an input unit that connects to the input unit 412 and the output unit 414. And an output interface unit 408. These units are communicably connected via an arbitrary communication path.
  • the storage unit 406 is a storage means, and for example, a memory device such as a RAM / ROM, a fixed disk device such as a hard disk, a flexible disk, an optical disk, or the like can be used.
  • the storage unit 406 stores various programs used for various processes.
  • the communication interface unit 404 mediates communication between the database device 400 and the network 300 (or a communication device such as a router). That is, the communication interface unit 404 has a function of communicating data with other terminals via a communication line.
  • the input / output interface unit 408 is connected to the input device 412 and the output device 414.
  • the output device 414 in addition to a monitor (including a home TV), a speaker or a printer can be used as the output device 414 (hereinafter, the output device 414 may be described as the monitor 414).
  • the input device 412 can be a monitor that realizes a pointing device function in cooperation with the mouse.
  • the control unit 402 has an internal memory for storing a control program such as an OS (Operating System), a program that defines various processing procedures, and necessary data, and performs various information processing based on these programs. Execute. As shown in the figure, the control unit 402 is roughly divided into a request interpreting unit 402a, a browsing processing unit 402b, an authentication processing unit 402c, an e-mail generating unit 402d, a Web page generating unit 402e, and a transmitting unit 402f.
  • a control program such as an OS (Operating System)
  • OS Operating System
  • the request interpretation unit 402a interprets the request content from the cancer immunotherapy evaluation apparatus 100, and passes the processing to each unit of the control unit 402 according to the interpretation result.
  • the browsing processing unit 402b Upon receiving browsing requests for various screens from the cancer immunotherapy evaluation apparatus 100, the browsing processing unit 402b generates and transmits Web data for these screens.
  • the authentication processing unit 402c receives an authentication request from the cancer immunotherapy evaluation device 100 and makes an authentication determination.
  • the e-mail generation unit 402d generates an e-mail including various types of information.
  • the web page generation unit 402e generates a web page that the user browses on the client device 200.
  • the transmission unit 402f transmits various types of information such as cancer state information and multivariate discriminants to the cancer immunotherapy evaluation apparatus 100.
  • FIG. 21 is a flowchart illustrating an example of a cancer immunotherapy evaluation service process according to the second embodiment.
  • the amino acid concentration data used in this processing is a specialist who uses blood (including plasma, serum, etc.) collected in advance from an individual such as an animal or human, for example, using a measurement method such as (A) or (B) below. Is related to the concentration value of amino acids obtained by analysis or independent analysis.
  • the unit of amino acid concentration may be obtained by, for example, molar concentration, weight concentration, or by adding / subtracting / subtracting an arbitrary constant to / from these concentrations.
  • Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at ⁇ 80 ° C. until the measurement of amino acid concentration.
  • amino acid concentration measurement For amino acid concentration measurement, acetonitrile was added to remove protein, followed by precolumn derivatization using a labeling reagent (3-aminopyridyl-N-hydroxysuccinimidyl carbamate), and liquid chromatography mass spectrometry The amino acid concentration was analyzed by a total (LC / MS) (see International Publication No. 2003/069328 and International Publication No. 2005/116629).
  • Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at ⁇ 80 ° C. until the measurement of amino acid concentration.
  • amino acid concentration When measuring the amino acid concentration, sulfosalicylic acid was added to remove the protein, and then the amino acid concentration was analyzed by an amino acid analyzer based on the post-column derivatization method using a ninhydrin reagent.
  • the client apparatus 200 causes the cancer immunotherapy evaluation apparatus to be displayed. 100 is accessed. Specifically, when the user instructs to update the screen of the Web browser 211 of the client device 200, the Web browser 211 uses the predetermined communication protocol to evaluate the cancer immunotherapy evaluation according to a predetermined communication protocol. By transmitting to the apparatus 100, a transmission request for a Web page corresponding to the amino acid concentration data transmission screen is made to the cancer immunotherapy evaluation apparatus 100 by routing based on the address.
  • an address such as URL
  • the cancer immunotherapy evaluation device 100 receives the transmission from the client device 200 at the request interpretation unit 102a, analyzes the content of the transmission, and moves the processing to each unit of the control unit 102 according to the analysis result.
  • the cancer immunotherapy evaluation apparatus 100 is a predetermined storage area of the storage unit 106 mainly in the browsing processing unit 102b. Web data for displaying the Web page stored in is acquired, and the acquired Web data is transmitted to the client device 200.
  • the cancer immunotherapy evaluation device 100 when there is a transmission request for a Web page corresponding to the amino acid concentration data transmission screen from the user, the cancer immunotherapy evaluation device 100 first uses the control unit 102 to check the user ID and the user password. Ask the user for input. Then, when the user ID and password are input, the cancer immunotherapy evaluation device 100 causes the authentication processing unit 102c to input the input user ID and password and the user ID stored in the user information file 106a. Make authentication with user password. The cancer immunotherapy evaluation apparatus 100 transmits Web data for displaying a Web page corresponding to the amino acid concentration data transmission screen to the client apparatus 200 by the browsing processing unit 102b only when authentication is possible. The client device 200 is identified by the IP address transmitted from the client device 200 together with the transmission request.
  • the client device 200 receives the Web data (for displaying a Web page corresponding to the amino acid concentration data transmission screen) transmitted from the cancer immunotherapy evaluation device 100 by the receiving unit 213, and receives the received Web data. Is interpreted by the Web browser 211, and the amino acid concentration data transmission screen is displayed on the monitor 261.
  • the client device 200 transmits input information and an identifier for specifying selection items to the cancer immunotherapy evaluation apparatus 100, so that the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment are evaluated for cancer immunotherapy. It transmits to the apparatus 100 (step SA21).
  • the transmission of amino acid concentration data in step SA21 may be realized by an existing file transfer technique such as FTP.
  • the cancer immunotherapy evaluation device 100 interprets the request contents of the client device 200 by interpreting the identifier transmitted from the client device 200 by the request interpretation unit 102a, and whether the cancer immunotherapy treatment is effective.
  • a request for transmission of a multivariate discriminant for determining whether or not is sent to the database apparatus 400.
  • the database apparatus 400 interprets the transmission request from the cancer immunotherapy evaluation apparatus 100 by the request interpretation unit 402a and stores the multivariate discriminant (for example, the updated latest data) stored in a predetermined storage area of the storage unit 406. Is transmitted to the cancer immunotherapy evaluation apparatus 100 (step SA22). Specifically, in step SA22, at least one of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro. A multivariate discriminant including one as a variable is transmitted to the cancer immunotherapy evaluation apparatus 100.
  • the cancer immunotherapy evaluation apparatus 100 uses the receiving unit 102f to determine the pre-treatment amino acid concentration data and post-treatment amino acid concentration data transmitted from the client device 200, and the multivariate discrimination transmitted from the database device 400.
  • the received pre-treatment amino acid concentration data and post-treatment amino acid concentration data are stored in a predetermined storage area of the amino acid concentration data file 106b, and the received multivariate discriminant is stored in the multivariate discriminant file 106e4.
  • the data is stored in a predetermined storage area (step SA23).
  • the controller 102 removes data such as missing values and outliers from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data of the individual received in step SA23 (step S23). SA24).
  • the cancer immunotherapy evaluation apparatus 100 includes the discriminant value calculation unit 102i in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data of the individual from which data such as a missing value and an outlier have been removed in step SA24.
  • At least one concentration value of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro and in step SA23 Multivariate including at least one of received Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro as a variable
  • one or more discriminant values eg, pre-treatment start Discriminant value obtained by substituting noic acid concentration data into a multivariate discriminant (discriminant value before treatment start), discriminant value obtained by substituting amino acid concentration data after treatment start
  • the cancer immunotherapy evaluation apparatus 100 compares the discriminant value calculated in step SA25 with a preset threshold value in the discriminant value criterion discriminating unit 102j1, so that treatment by cancer immunotherapy is effective for the individual. And the determination result is stored in a predetermined storage area of the evaluation result file 106g (step SA26).
  • the cancer immunotherapy evaluation apparatus 100 uses the transmission unit 102m to send the determination result obtained in step SA26 (which may include the determination value calculated in step SA25) to the client apparatus 200 that is the transmission source of amino acid concentration data.
  • the data is transmitted to the database device 400 (step SA27).
  • the cancer immunotherapy evaluation apparatus 100 creates a web page for displaying the discrimination result in the web page generation unit 102e, and stores Web data corresponding to the created web page in the storage unit 106. Stored in the storage area.
  • the client device 200 issues a request for browsing the Web page to the cancer immunotherapy evaluation device 100. Send to.
  • the browsing processing unit 102b interprets the browsing request transmitted from the client device 200, and stores Web data corresponding to the Web page for displaying the determination result in the storage unit 106. Read from storage area. Then, the cancer immunotherapy evaluation device 100 transmits the read Web data to the client device 200 and transmits the Web data or the determination result to the database device 400 by the transmission unit 102m.
  • the cancer immunotherapy evaluation apparatus 100 may notify the user client apparatus 200 of the determination result by e-mail at the control unit 102. Specifically, the cancer immunotherapy evaluation apparatus 100 first refers to the user information stored in the user information file 106a based on the user ID or the like in the e-mail generation unit 102d according to the transmission timing, Get the user's email address. Next, the cancer immunotherapy evaluation apparatus 100 uses the e-mail generation unit 102d to generate data related to the e-mail including the name and discrimination result of the user with the acquired e-mail address as the destination. Next, the cancer immunotherapy evaluation apparatus 100 transmits the generated data to the user client apparatus 200 by the transmission unit 102m.
  • the cancer immunotherapy evaluation apparatus 100 may transmit the determination result to the user client apparatus 200 using an existing file transfer technique such as FTP.
  • control unit 402 receives the discrimination result or Web data transmitted from the cancer immunotherapy evaluation device 100, and stores the received discrimination result or Web data in a predetermined unit of the storage unit 406. Save (accumulate) in the storage area (step SA28).
  • the client device 200 receives the Web data transmitted from the cancer immunotherapy evaluation device 100 by the receiving unit 213, interprets the received Web data by the Web browser 211, and the Web page on which the individual determination result is written. Is displayed on the monitor 261 (step SA29).
  • the client apparatus 200 arbitrarily selects the e-mail transmitted from the cancer immunotherapy evaluation apparatus 100 by a known function of the e-mailer 212.
  • the received e-mail is displayed on the monitor 261.
  • the user can check the determination result by browsing the Web page displayed on the monitor 261.
  • the user may print the display content of the Web page displayed on the monitor 261 with the printer 262.
  • the user can check the discrimination result by browsing the e-mail displayed on the monitor 261.
  • the user may print the content of the e-mail displayed on the monitor 261 with the printer 262.
  • a cancer immunotherapy evaluation device a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a recording medium, a cancer immunotherapy evaluation system, and an information communication terminal device according to the present invention are claimed in addition to the second embodiment described above.
  • the present invention may be implemented in various different embodiments within the scope of the technical idea described in the above.
  • each illustrated component is functionally conceptual and does not necessarily need to be physically configured as illustrated.
  • the processing functions provided in the cancer immunotherapy evaluation apparatus 100 is interpreted and executed by a CPU (Central Processing Unit) and the CPU. It may be realized by a program or hardware based on wired logic.
  • the program is recorded on a non-transitory computer-readable recording medium including programmed instructions for causing the information processing apparatus to execute the cancer immunotherapy evaluation method according to the present invention. It is mechanically read by the immunotherapy evaluation apparatus 100. That is, in the storage unit 106 such as a ROM or an HDD, computer programs for performing various processes by giving instructions to the CPU in cooperation with an OS (Operating System) are recorded. This computer program is executed by being loaded into the RAM, and constitutes a control unit in cooperation with the CPU.
  • OS Operating System
  • the computer program may be stored in an application program server connected to the cancer immunotherapy evaluation apparatus 100 via an arbitrary network, and may be downloaded in whole or in part as necessary. Is possible.
  • the cancer immunotherapy evaluation program according to the present invention may be stored in a non-temporary computer-readable recording medium, or may be configured as a program product.
  • the “recording medium” means a memory card, USB memory, SD card, flexible disk, magneto-optical disk, ROM, EPROM, EEPROM (registered trademark), CD-ROM, MO, DVD, and Blu-ray. (Registered trademark) It shall include any “portable physical medium” such as Disc.
  • the “program” is a data processing method described in an arbitrary language or description method, and may be in the form of source code or binary code. Note that the “program” is not necessarily limited to a single configuration, but is distributed in the form of a plurality of modules and libraries, or in cooperation with a separate program typified by an OS (Operating System). Including those that achieve the function. In addition, a well-known structure and procedure can be used about the specific structure and reading procedure for reading a recording medium in each apparatus shown to embodiment, the installation procedure after reading, etc.
  • Various databases and the like stored in the storage unit 106 are storage devices such as a memory device such as a RAM and a ROM, a fixed disk device such as a hard disk, a flexible disk, and an optical disk. Programs, tables, databases, web page files, and the like.
  • the cancer immunotherapy evaluation apparatus 100 may be configured as an information processing apparatus such as a known personal computer or workstation, or may be configured as the information processing apparatus connected to an arbitrary peripheral device.
  • the cancer immunotherapy evaluation apparatus 100 may be realized by installing software (including a program or data) that realizes the cancer immunotherapy evaluation method of the present invention in the information processing apparatus.
  • the specific form of distribution / integration of the devices is not limited to that shown in the figure, and all or a part of them may be functionally or physically in arbitrary units according to various additions or according to functional loads. It can be configured to be distributed and integrated. That is, the above-described embodiments may be arbitrarily combined and may be selectively implemented.
  • FIG. 22 is a flowchart illustrating an example of multivariate discriminant creation processing.
  • the multivariate discriminant creation process may be performed by the database apparatus 400 that manages cancer state information.
  • the cancer immunotherapy evaluation device 100 stores cancer state information acquired in advance from the database device 400 in a predetermined storage area of the cancer state information file 106c.
  • the cancer immunotherapy evaluation apparatus 100 uses the cancer state index data specified in advance by the cancer state information specifying unit 102g (for example, data related to an index indicating a cancer state (for example, presence or absence of a therapeutic effect obtained based on the index). ), Or data relating to the amount of change in an indicator of cancer status (for example, a difference in tumor size) and amino acid concentration data (eg, data relating to amino acid concentration or data relating to the amount of change in amino acid concentration) , Etc.) is stored in a predetermined storage area of the designated cancer state information file 106d.
  • the cancer state index data specified in advance by the cancer state information specifying unit 102g for example, data related to an index indicating a cancer state (for example, presence or absence of a therapeutic effect obtained based on the index).
  • data relating to the amount of change in an indicator of cancer status for
  • the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1 based on a predetermined formula creation method from cancer state information stored in a predetermined storage area of the designated cancer state information file 106d. A multivariate discriminant is created, and the created candidate multivariate discriminant is stored in a predetermined storage area of the candidate multivariate discriminant file 106e1 (step SB21).
  • the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1, and a plurality of different formula creation methods (principal component analysis, discriminant analysis, support vector machine, multiple regression analysis, logistic regression) Analysis, k-means method, cluster analysis, decision tree, etc.
  • the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1, and executes various calculations (for example, average and variance) corresponding to the selected formula selection method based on the cancer state information. .
  • the multivariate discriminant-preparing part 102h determines the calculation result and parameters of the determined candidate multivariate discriminant-expression in the candidate multivariate discriminant-preparing part 102h1. Thereby, a candidate multivariate discriminant is created based on the selected formula creation method.
  • a candidate multivariate discriminant when created in parallel and in parallel by using a plurality of different formula creation methods, the above-described processing may be executed in parallel for each selected formula creation method.
  • the candidate multivariate discriminant when creating a candidate multivariate discriminant serially using a combination of multiple different formula creation methods, for example, transform cancer status information using a candidate multivariate discriminant created by performing principal component analysis. Then, the candidate multivariate discriminant may be created by performing discriminant analysis on the converted cancer state information.
  • the multivariate discriminant-preparing part 102h verifies (mutually verifies) the candidate multivariate discriminant created in step SB21 with the candidate multivariate discriminant-verifying part 102h2, and verifies the verification result.
  • the result is stored in a predetermined storage area of the verification result file 106e2 (step SB22).
  • the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-verifying part 102h2, based on the cancer state information stored in a predetermined storage area of the designated cancer state information file 106d.
  • the verification data used when verifying the formula is created, and the candidate multivariate discriminant is verified based on the created verification data.
  • the multivariate discriminant creation unit 102h creates each formula in the candidate multivariate discriminant verification unit 102h2.
  • Each candidate multivariate discriminant corresponding to the method is verified based on a predetermined verification method.
  • the discrimination rate, sensitivity, and specificity of the candidate multivariate discriminant based on at least one of the random sampling method, the bootstrap method, the holdout method, the N-fold method, the leave one-out method, etc. , Information criteria, ROC_AUC (area under the receiver characteristic curve), etc.
  • the multivariate discriminant-preparing part 102h creates a candidate multivariate discriminant by selecting a variable of the candidate multivariate discriminant based on a predetermined variable selection method in the variable selector 102h3.
  • a combination of amino acid concentration data included in the cancer state information to be used is selected, and cancer state information including the selected combination of amino acid concentration data is stored in a predetermined storage area of the selected cancer state information file 106e3 (step SB23).
  • step SB21 a plurality of candidate multivariate discriminants are created in combination with a plurality of different formula creation methods, and in step SB22, each candidate multivariate discriminant corresponding to each formula creation method is verified based on a predetermined verification method.
  • the multivariate discriminant-preparing part 102h selects a variable of the candidate multivariate discriminant based on a predetermined variable selection method for each candidate multivariate discriminant in the variable selector 102h3. Also good.
  • the variable of the candidate multivariate discriminant may be selected based on at least one of the stepwise method, the best path method, the neighborhood search method, and the genetic algorithm from the verification result.
  • the best path method is a method of selecting variables by sequentially reducing the variables included in the candidate multivariate discriminant one by one and optimizing the evaluation index given by the candidate multivariate discriminant.
  • the multivariate discriminant-preparing part 102h selects a combination of amino acid concentration data based on the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d by the variable selection part 102h3. May be.
  • the multivariate discriminant-preparing part 102h determines whether or not all combinations of amino acid concentration data included in the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d have been completed. If the determination result is “end” (step SB24: Yes), the process proceeds to the next step (step SB25). If the determination result is not “end” (step SB24: No), the process proceeds to step SB21. Return. The multivariate discriminant-preparing part 102h determines whether or not the preset number of times has ended, and if the determination result is “end” (step SB24: Yes), the next step (step SB25). If the determination result is not “end” (step SB24: No), the process may return to step SB21.
  • the multivariate discriminant-preparing part 102h has a combination of amino acid concentration data included in the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d as the combination of the amino acid concentration data selected in step SB23.
  • the multivariate discriminant creation unit 102h compares the evaluation value with a predetermined threshold corresponding to each formula creation method. Based on the result, it may be determined whether to proceed to step SB25 or to return to step SB21.
  • the multivariate discriminant-preparing part 102h selects a multivariate discriminant by selecting a candidate multivariate discriminant to be adopted as a multivariate discriminant from a plurality of candidate multivariate discriminants based on the verification result.
  • the determined multivariate discriminant (selected candidate multivariate discriminant) is stored in a predetermined storage area of the multivariate discriminant file 106e4 (step SB25).
  • step SB25 for example, when the optimum one is selected from candidate multivariate discriminants created by the same formula creation method, and when the optimum one is selected from all candidate multivariate discriminants There is.
  • FIGS. The horizontal axis of the graphs shown in FIG. 24 and FIG. 25 represents the number of days when the day of 5FU administration is day 0, and the vertical axis represents the average value of tumor size (mm 2 ).
  • FIG. 24 shows WT (responder) data and
  • FIG. 25 shows Nude (non-responder) data. 24 and 25 show a group with treatment and a group without treatment, respectively.
  • Measurement of amino acid concentration in plasma was performed by the measurement method (A) described in the above embodiment.
  • FIG. 26 and FIG. 27 show data on amino acid concentrations in plasma of WT mice. 26 and 27, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration ( ⁇ M).
  • Pre pre-treatment start
  • post post
  • ⁇ M average value of each amino acid concentration
  • FIG. 28 and FIG. 29 show data on amino acid concentrations in plasma of Nude mice. 28 and 29, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration ( ⁇ M).
  • pre pre-treatment start
  • post post
  • ⁇ M average value of each amino acid concentration
  • FIG. 30 shows a radar chart showing the distribution of each amino acid after the start of treatment, assuming that each amino acid before the start of treatment is 100% for WT (responder).
  • FIG. 31 shows a radar chart showing the distribution of each amino acid after the start of treatment when Nude (non-responder) is 100% of each amino acid before the start of treatment.
  • the amino acid profile change in WT and the amino acid profile change in Nude were different, and a characteristic amino acid profile change in plasma was clarified when an antitumor effect was obtained.
  • the amino acid variables Val, Leu, Ile, His, Phe, Trp, Gln, Asp, and Orn that were significantly different before and after the start of WT treatment were found to have the ability to discriminate the effects of cancer immunotherapy. It was also found that the amino acid variables Ala, Thr, Lys, and Pro that had a significant difference before and after the start of Nude treatment also had the ability to discriminate cancer immunotherapy effects.
  • the discrimination performance of 2-group discrimination before and after the start of treatment with each amino acid variable in WT mice was evaluated by the area under the ROC curve (ROC_AUC).
  • the AUC was greater than 0.7 for the amino acid variables Val, Leu, Ile, Lys, His, Phe, Trp, Gln, Asp, and Orn (FIGS. 32 and 33).
  • the amino acid variables Val, Leu, Ile, Lys, His, Phe, Trp, Gln, Asp, and Orn were found to have the ability to discriminate the effects of cancer immunotherapy.
  • a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained.
  • a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • a combination of variables (4 or less) to be included in a fractional expression as a multivariate discriminant is searched from the following 22 types of amino acids, and the bootstrap method is used as cross validation Was used to search for a fractional expression that maximizes the ability to discriminate the effects of cancer immunotherapy.
  • 22 kinds of amino acids are Ala, Arg, Asn, Asp, Cit, Cys2, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Orn, Phe, Pro, Ser, Thr, Trp, Tyr. , Val.
  • FIG. 34 shows a list of fractional expressions having a good discrimination ability that the value of ROC_AUC is 1.
  • FIG. 35 shows the frequency of appearance of variables in the formula included in FIG. The amino acid variables up to position 10 in the order of appearance frequency were Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, and His. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • FIG. 36, 37, and 38 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC.
  • FIG. 36, FIG. 37, and FIG. 38 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 39 shows the appearance frequency of variables in the expressions included in FIGS. 36, 37, and 38.
  • the amino acid variables up to position 10 in the order of appearance frequency were Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • a difference (post-pre) was calculated by subtracting each plasma amino acid concentration after the start of treatment from each plasma amino acid concentration before the start of the treatment, and a change related to the difference. Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • FIG. 40, FIG. 41 and FIG. 42 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC.
  • FIG. 40, FIG. 41, and FIG. 42 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 43 shows the appearance frequency of variables in the expressions included in FIGS. 40, 41, and 42.
  • the amino acid variables up to position 10 in the order of appearance frequency were Trp, Asp, Ile, Thr, Arg, Pro, Ser, Leu, Met, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained.
  • a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • FIGS. 44-48 shows a list of logistic regression equations with equally good discriminating ability evaluated by the value of ROC_AUC.
  • FIGS. 44 to 48 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 49 shows the frequency of occurrence of variables in the expressions included in FIGS.
  • the amino acid variables up to position 10 in the order of appearance frequency were His, Thr, Lys, Phe, Arg, Ile, Met, Ser, Val, Asn. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • a difference (post-pre) was calculated by subtracting each plasma amino acid concentration after the start of treatment from each plasma amino acid concentration before the start of the treatment, and a change related to the difference. Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • Figure 50-54 shows a list of logistic regression equations with equally good discriminating ability evaluated by the value of ROC_AUC.
  • 50-54 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 55 shows the appearance frequency of variables in the expressions included in FIGS.
  • the amino acid variables up to position 10 in the order of appearance frequency were Lys, Gln, Thr, Phe, Met, Pro, Ser, Ala, Asn, and Val. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • T cells were isolated from the spleen of 4T1 tumor-bearing mice, and anti-4T1 activated T cells were prepared by activating and proliferating in the presence of anti-CD3 antibody, anti-CD28 antibody and IL-2.
  • 4T1 cells or CT26 cells were subcutaneously transplanted (1 ⁇ 10 6 cells / 100 ⁇ l) into 10 Balb / c mice, respectively, and the above-mentioned anti-4T1 activated T cells were transplanted from the tail vein 10 days after the transplantation.
  • tumor tissues of 4T1 tumor-bearing mice and CT26 tumor-bearing mice were excised and frozen sections were prepared, and then H / E staining, anti-CD4 antibody, anti-CD8 antibody and anti-F4 / 80 antibody (macrophage marker) ) Fluorescence immunostaining was performed (FIG. 57).
  • Measurement of amino acid concentration in plasma was performed by the measurement method (A) described in the above embodiment.
  • FIG. 58 and FIG. 58 and 59 Data of amino acid concentrations in plasma of 4T1 tumor-bearing mice are shown in FIG. 58 and FIG. 58 and 59, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration ( ⁇ M).
  • pre pre-treatment start
  • post post
  • ⁇ M average value of each amino acid concentration
  • FIGS. radar charts showing the distribution of each amino acid after the start of treatment when each amino acid before the start of treatment is taken as 100% are shown in FIGS.
  • the pattern of amino acid profiles was different.
  • the pattern of amino acid profile change in plasma differs depending on the presence or absence of antitumor effects.
  • Arg, His, Met, and Cys2 have the ability to discriminate the effects of cancer immunotherapy.
  • Ala, Thr, Pro among the amino acid variables that had a significant difference before and after the start of treatment of CT26 tumor-bearing mice also had the ability to discriminate cancer immunotherapy effects.
  • FIG. 66 shows a list of fractional expressions having good discrimination ability that the value of ROC_AUC is 1.
  • FIG. 67 shows the frequency of occurrence of variables in the formula included in FIG.
  • the amino acid variables up to position 10 in the order of appearance frequency were Pro, Glu, Orn, Arg, Gly, Ile, Thr, Trp, Ser, Tyr. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • FIG. 68, FIG. 69, and FIG. 70 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC.
  • FIG. 68, FIG. 69, and FIG. 70 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 71 shows the frequency of occurrence of variables in the expressions included in FIGS.
  • the amino acid variables up to position 10 in the order of appearance frequency were Trp, His, Thr, Ile, Pro, Phe, Asn, Arg, Cys2, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • Example 6 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post-pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and a change related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • FIG. 72, 73, and 74 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC.
  • FIG. 72, FIG. 73, and FIG. 74 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • FIG. 75 shows the appearance frequency of variables in the expressions included in FIGS. 72, 73, and 74.
  • the amino acid variables up to position 10 in the order of appearance frequency were Trp, His, Thr, Ile, Pro, Phe, Asn, Arg, Cys2, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
  • FIG. 76-80 shows a logistic regression equation (including variable combinations, coefficients, and constants) and a value of ROC_AUC with cross validation.
  • Example 6 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post-pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and a change related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
  • FIGS. 81 to 85 show a list of logistic regression equations with discriminability evaluated by ROC_AUC value of 0.7 or more.
  • FIGS. 81 to 85 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
  • the cancer immunotherapy evaluation method and the like according to the present invention can be widely implemented in many industrial fields, particularly in the fields of pharmaceuticals, foods, and medicine, and in particular, the therapeutic effect of cancer immunotherapy. It is extremely useful in the field of bioinformatics for evaluating the above.

Abstract

The purpose of the present invention is to provide a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation device, a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a cancer immunotherapy evaluation system and an information communication terminal device, each of which can evaluate the therapeutic effect of a cancer immunotherapy with high accuracy utilizing a blood amino acid concentration. In the present embodiment, both pre-therapy amino acid concentration data, which relates to a value of the concentration of an amino acid in blood collected from a subject of interest who is intended to treated by a cancer immunotherapy prior to the start of the therapy, and post-therapy amino acid concentration data, which relates to a value of the concentration of the amino acid in blood collected from the subject of interest after the start of the therapy, are obtained, and the effect of the therapy on the subject is evaluated on the basis of the pre-therapy amino acid concentration data and the post-therapy amino acid concentration data obtained.

Description

[規則37.2に基づきISAが決定した発明の名称] 癌免疫療法の評価方法、癌免疫療法評価装置、癌免疫療法評価プログラム、癌免疫療法評価システムおよび情報通信端末装置[Name of invention determined by ISA based on Rule 37.2] Cancer immunotherapy evaluation method, cancer immunotherapy evaluation device, cancer immunotherapy evaluation program, cancer immunotherapy evaluation system, and information communication terminal device
 本発明は、癌免疫療法の評価方法、癌免疫療法評価装置、癌免疫療法評価方法、癌免疫療法評価プログラム、癌免疫療法評価システムおよび情報通信端末装置に関するものである。 The present invention relates to a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation device, a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a cancer immunotherapy evaluation system, and an information communication terminal device.
 免疫療法(免疫治療)とは、免疫担当細胞、サイトカインまたは抗体等を活性化する物質を用いて生体に備わっている免疫機能を目的の方向に導く治療法であり、種々の癌に対する免疫療法は公知である。癌免疫療法は一定の効果を発揮し得るが、治療に対するレスポンダーとノンレスポンダーが存在し、その効果には個人差が生じる。ここで、レスポンダーには、癌が根絶、縮小もしくは改善されている(混合型レスポンダーまたは部分的レスポンダー)個体、または癌が進行していない個体などが含まれる。また、癌が進行していない個体とは、治療を受けていない個体と比較して、癌の増大が認められない、QOL(Quality Of Life)が向上または維持されている、および/または、平均余命が増加している個体などを指す。 Immunotherapy (immunotherapy) is a treatment that uses the substance that activates immunocompetent cells, cytokines, antibodies, etc. to direct the immune function of the living body in the intended direction, and immunotherapy for various cancers It is known. Although cancer immunotherapy can exert certain effects, there are responders and non-responders to treatment, and the effects vary among individuals. Here, the responder includes an individual whose cancer has been eradicated, reduced or improved (mixed responder or partial responder), an individual whose cancer has not progressed, and the like. In addition, an individual in which cancer has not progressed is an increase in cancer, quality of life (QOL) is improved or maintained, and / or average compared to an individual who has not been treated. An individual whose life expectancy has increased.
 癌に対する治療方法の有効性を評価する手法については、画像により評価する手法が基本的に提唱されており、また、生存期間の延長により評価する手法が通常用いられている。しかし、癌に対する治療方法の有効性を評価するこれまでの手法の多くは、新薬の評価及び再評価に際して用いられているものである。そのため、当該これまでの手法を通常の癌治療の評価に適用するのは難しいのが現状である。 As a method of evaluating the effectiveness of a treatment method for cancer, a method of evaluating by image is basically proposed, and a method of evaluating by extending the survival period is usually used. However, many of the conventional methods for evaluating the effectiveness of therapeutic methods for cancer are used in the evaluation and re-evaluation of new drugs. For this reason, it is difficult to apply the conventional method to the evaluation of normal cancer treatment.
 癌免疫療法による治療が開始されたのち早期にその治療効果を判定することができることは、癌患者のQOL低下を防ぐことや、治療法変更への判断材料を提供することに繋がる。よって、少なくとも治療途中で効果の有無が判り且つ腫瘍組織での癌免疫応答を直接反映しているバイオマーカーが望まれる。 The ability to determine the therapeutic effect at an early stage after the start of cancer immunotherapy treatment leads to prevention of QOL reduction in cancer patients and provision of judgment materials for treatment change. Therefore, a biomarker that can be determined whether or not it is effective at least during treatment and directly reflects the cancer immune response in the tumor tissue is desired.
 現在、評価またはモニタリングのために、免疫細胞治療を受けた患者における免疫応答や癌に対する生体の免疫応答の詳細な解析が行われている。例えば、末梢血中の抗腫瘍T細胞の測定が広く実施されており、測定には、フローサイトメーターによる細胞内サイトカイン測定や、ELISPOT法、ELISA法、テトラマー法などが用いられている。しかしながら、末梢血中の抗腫瘍T細胞の評価だけでは、抗腫瘍効果の予測は困難である(非特許文献1、2)。また、腫瘍組織や所属リンパ節での免疫応答を直接測定する試みもある。例えば、癌局所の抗原特異活性化T細胞の検出は臨床治療成績(clinical outcome)への関与が高いことが報告される(非特許文献3)。しかしながら、患者側への負担も大きく、手間も要し、臨床検体が得られない場合も多い。 Currently, for the purpose of evaluation or monitoring, detailed analysis of the immune response in patients receiving immune cell therapy and the immune response of the living body against cancer is being performed. For example, measurement of anti-tumor T cells in peripheral blood is widely carried out, and intracellular cytokine measurement using a flow cytometer, ELISPOT method, ELISA method, tetramer method, etc. are used for the measurement. However, it is difficult to predict an antitumor effect only by evaluating antitumor T cells in peripheral blood (Non-Patent Documents 1 and 2). There are also attempts to directly measure immune responses in tumor tissues and regional lymph nodes. For example, it has been reported that detection of antigen-specific activated T cells in a local cancer region is highly involved in clinical outcome (Non-patent Document 3). However, the burden on the patient side is large, labor is required, and clinical specimens are often not obtained.
 このように、癌免疫療法の抗腫瘍効果を判別する良い方法はなく、そのため、精度の良い、血液などの簡便なバイオマーカーが望まれている。 Thus, there is no good method for discriminating the antitumor effect of cancer immunotherapy, and therefore, a simple biomarker such as blood with high accuracy is desired.
 ここで、血液中アミノ酸の濃度が、癌に対する治療により変化することが知られている。例えば、乳癌患者では外科的手術により血中セリンとグルタミン酸濃度が正常化することが報告されている(非特許文献4)。また、癌患者の外科的手術直後における血中非必須アミノ酸の低下が報告される(非特許文献5,6)。また、乳癌患者のウクライン治療により血中プロリン、タウリン、グルタミン酸濃度が増加し、アラニン濃度は減少することが報告されている(非特許文献7)。更に、シスプラチンを含む化学療法で効果のある患者では効果のない患者に比べて1サイクル目で17種類の血中アミノ酸濃度が増加していたという報告がある(非特許文献8)。 Here, it is known that the concentration of amino acids in blood changes due to treatment for cancer. For example, it has been reported that blood serum serine and glutamic acid concentrations are normalized by surgery in breast cancer patients (Non-patent Document 4). In addition, a decrease in non-essential amino acids in blood immediately after surgery for cancer patients is reported (Non-Patent Documents 5 and 6). In addition, it has been reported that blood proline, taurine, and glutamic acid concentrations increase and alanine concentration decreases by Ukrain treatment of breast cancer patients (Non-patent Document 7). Furthermore, there is a report that 17 types of blood amino acid concentrations were increased in the first cycle in patients who were effective with chemotherapy including cisplatin compared to patients who were not effective (Non-patent Document 8).
 また、先行特許として、アミノ酸濃度と生体状態とを関連付ける方法に関する特許文献1、特許文献2、および特許文献3が公開されている。また、アミノ酸濃度を用いて肺癌の状態を評価する方法に関する特許文献4、アミノ酸濃度を用いて乳癌の状態を評価する方法に関する特許文献5、アミノ酸濃度を用いて大腸癌の状態を評価する方法に関する特許文献6、アミノ酸濃度を用いて癌の状態を評価する方法に関する特許文献7、アミノ酸濃度を用いて胃癌の状態を評価する方法に関する特許文献8、アミノ酸濃度を用いて癌種を評価する方法に関する特許文献9、アミノ酸濃度を用いて女性生殖器癌の状態を評価する特許文献10、およびアミノ酸濃度を用いて前立腺癌を含む前立腺疾患の状態を評価する特許文献11が公開されている。 Also, Patent Literature 1, Patent Literature 2, and Patent Literature 3 relating to a method for associating an amino acid concentration with a biological state are disclosed as prior patents. In addition, Patent Document 4 relating to a method for evaluating lung cancer status using amino acid concentration, Patent Literature 5 relating to a method for evaluating breast cancer status using amino acid concentration, and a method for evaluating colon cancer status using amino acid concentration Patent Document 6, Patent Document 7 related to a method for evaluating cancer status using amino acid concentration, Patent Document 8 related to a method for evaluating gastric cancer status using amino acid concentration, and a method for evaluating a cancer type using amino acid concentration Patent Document 9, Patent Document 10 that evaluates the state of female genital cancer using amino acid concentration, and Patent Document 11 that evaluates the state of prostate disease including prostate cancer using amino acid concentration are disclosed.
国際公開第2004/052191号International Publication No. 2004/052191 国際公開第2006/098192号International Publication No. 2006/098192 国際公開第2009/054351号International Publication No. 2009/054351 国際公開第2008/016111号International Publication No. 2008/016111 国際公開第2008/075662号International Publication No. 2008/077562 国際公開第2008/075663号International Publication No. 2008/077563 国際公開第2008/075664号International Publication No. 2008/077564 国際公開第2009/099005号International Publication No. 2009/099005 国際公開第2009/110517号International Publication No. 2009/110517 国際公開第2009/154296号International Publication No. 2009/154296 国際公開第2009/154297号International Publication No. 2009/154297
 しかしながら、非特許文献4から8はいずれも外科的手術や化学療法における報告であり、癌免疫療法においては血液中アミノ酸濃度変化に関連した報告はない。また、特許文献1-特許文献11に開示されている指標式群を癌免疫療法の治療効果の評価に用いても、評価の対象が異なるので、治療効果の評価について十分な精度を得ることはできない。 However, all of Non-Patent Documents 4 to 8 are reports on surgical operation and chemotherapy, and there are no reports related to changes in blood amino acid concentration in cancer immunotherapy. Further, even if the index formula groups disclosed in Patent Document 1 to Patent Document 11 are used for evaluating the therapeutic effect of cancer immunotherapy, the evaluation target is different, so that sufficient accuracy can be obtained for evaluating the therapeutic effect. Can not.
 つまり、血中アミノ酸濃度を利用して癌免疫療法の治療効果を評価する方法の開発は行われておらず、また実用化もされていないという問題点があった。 That is, there has been a problem that a method for evaluating the therapeutic effect of cancer immunotherapy using blood amino acid concentration has not been developed and has not been put into practical use.
 本発明は、上記問題点に鑑みてなされたもので、血中アミノ酸濃度を利用して癌免疫療法の治療効果を精度よく評価することができる癌免疫療法の評価方法、癌免疫療法評価装置、癌免疫療法評価方法、癌免疫療法評価プログラム、癌免疫療法評価システムおよび情報通信端末装置を提供することを目的とする。 The present invention has been made in view of the above problems, and an cancer immunotherapy evaluation method, a cancer immunotherapy evaluation apparatus, which can accurately evaluate the therapeutic effect of cancer immunotherapy using the blood amino acid concentration, An object is to provide a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a cancer immunotherapy evaluation system, and an information communication terminal device.
 本発明者らは、上述した課題を解決するために鋭意検討した結果、血液中のアミノ酸で癌免疫療法の治療効果(具体的には、癌免疫療法における抗腫瘍免疫効果)の判別を精度よくできることを見出し、本発明を完成するに至った。 As a result of intensive studies to solve the above-described problems, the present inventors have accurately determined the therapeutic effect of cancer immunotherapy using amino acids in blood (specifically, the anti-tumor immune effect in cancer immunotherapy). The present inventors have found that this can be done and have completed the present invention.
 すなわち、上述した課題を解決し、目的を達成するために、本発明にかかる癌免疫療法の評価方法は、癌免疫療法による治療を受ける評価対象から前記治療が開始される前に採取された血液中のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記評価対象から前記治療が開始された後に採取された血液中のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを取得する取得ステップと、前記取得ステップで取得した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに基づいて、前記評価対象に対する前記治療の効果を評価する濃度値基準評価ステップとを含むことを特徴とする。なお、前記治療開始後アミノ酸濃度データは、後述する「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。 That is, in order to solve the above-described problems and achieve the object, the method for evaluating cancer immunotherapy according to the present invention includes blood collected before the treatment is started from an evaluation subject receiving treatment by cancer immunotherapy. An acquisition step of acquiring amino acid concentration data before starting treatment regarding the concentration value of amino acids in the blood, and amino acid concentration data after starting treatment regarding the concentration value of amino acids in blood collected after the treatment is started from the evaluation target; And a concentration value reference evaluation step for evaluating the effect of the treatment on the evaluation object based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data acquired in the acquisition step. The post-treatment amino acid concentration data may be data (after treatment amino acid concentration data) corresponding to “after treatment” amino acid concentration data described later.
 ここで、本明細書において、癌免疫療法には、免疫細胞療法、ペプチド・ワクチン療法、BMR(Biological Response Modifiers)療法、サイトカイン療法、抗体療法、および、免疫抑制機構の解除による抗腫瘍効果誘導、などが含まれる。 Here, in the present specification, cancer immunotherapy includes immune cell therapy, peptide / vaccine therapy, BMR (Biological Response Modifiers) therapy, cytokine therapy, antibody therapy, and induction of antitumor effect by release of immunosuppressive mechanism, Etc. are included.
 また、本明細書では、「治療が開始される前」を「治療前」または「治療開始前」と記し、「治療が開始された後」を「治療開始後」と記す場合がある。また、本明細書において、「治療開始前」には、例えば、一定期間に亘る広義の治療における初回の狭義の治療が行われる前、などが含まれる。また、本明細書において、「治療開始後」には、例えば、一定期間に亘る広義の治療における初回の狭義の治療が行われた後で且つ最終回の狭義の治療が行われる前(例えば、一般的に言われる「治療中」など)、または、一定期間に亘る広義の治療における最終回の狭義の治療が行われた後(例えば、一般的に言われる「治療後」など)、などが含まれる。 In this specification, “before treatment is started” may be referred to as “before treatment” or “before treatment start”, and “after treatment is started” may be referred to as “after treatment start”. In the present specification, “before treatment start” includes, for example, before the first narrow treatment in a broad sense over a certain period of time. In the present specification, “after the start of treatment” includes, for example, after the first narrow treatment in the broad sense treatment for a certain period of time and before the final narrow treatment (for example, After being treated in a broad sense over a certain period of time (for example, after being treated generally), etc. included.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記濃度値基準評価ステップは、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの前記濃度値に基づいて、前記評価対象に対する前記治療の効果を評価すること、を特徴とする。 In the cancer immunotherapy evaluation method according to the present invention, the concentration value reference evaluation step is included in the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment. Based on the concentration value of at least one of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro. It is characterized by evaluating the effect of the said treatment with respect to an evaluation object.
 ここで、本明細書では各種アミノ酸を主に略称で表記するが、それらの正式名称は以下の通りである。
(略称)     (正式名称)
Ala      Alanine
Arg      Arginine
Asp      Aspartic acid
Cys2     Cystine
Gln      Glutamine
Glu      Glutamic acid
Gly      Glycine
His      Histidine
Ile      Isoleucine
Leu      Leucine
Lys      Lysine
Met      Methionine
Orn      Ornithine
Phe      Phenylalanine
Pro      Proline
Ser      Serine
Thr      Threonine
Trp      Tryptophan
Val      Valine
Here, although various amino acids are mainly represented by abbreviations in the present specification, their formal names are as follows.
(Abbreviation) (official name)
Ala Alanine
Arg Arginine
Asp Aspartic acid
Cys2 Cystein
Gln Glutamine
Glu Glutamic acid
Gly Glycine
His Histide
Ile Isolucine
Leu Leucine
Lys Lysine
Met Methionine
Orn Origine
Phe Phenylalanine
Pro Proline
Ser Serine
Thr Threoneine
Trp Tryptophan
Val Valine
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記濃度値基準評価ステップは、前記濃度値に基づいて、前記評価対象に対して前記治療が有効であるかを評価する(例えば、前記濃度値に基づいて、前記評価対象に対して前記治療が有効であるか否かを判別する濃度値基準判別ステップをさらに含む)こと、を特徴とする。 Further, the cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step is based on the concentration value and the treatment is effective for the evaluation object. (E.g., further comprising a concentration value criterion determining step for determining whether or not the treatment is effective for the evaluation object based on the concentration value).
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記濃度値基準評価ステップは、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む予め設定された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップをさらに含むこと、を特徴とする。なお、前記判別値算出ステップは、前記治療開始前アミノ酸濃度データと前記治療開始後アミノ酸濃度データに基づいて、前記治療が開始される前のアミノ酸の濃度値と前記治療が開始された後のアミノ酸の濃度値との比または差分を算出し、算出した各アミノ酸の濃度値の比または差分を多変量判別式に含まれる各変数に代入することで、判別値を算出してもよい。また、前記治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。 Further, the cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step includes the pre-treatment amino acid concentration data, the post-treatment amino acid concentration data, and the amino acid Based on a preset multivariate discriminant that includes the concentration of as a variable, a discriminant value calculation that calculates a discriminant value that is a value of the multivariate discriminant and that corresponds to an evaluation result regarding the effect of the treatment on the evaluation target The method further includes a step. The discriminant value calculating step is based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, and the amino acid concentration value before starting treatment and the amino acid after starting treatment. Alternatively, the discriminant value may be calculated by calculating the ratio or difference with the concentration value and substituting the calculated concentration value ratio or difference of each amino acid into each variable included in the multivariate discriminant. The amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つであること、を特徴とする。 The cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the multivariate discriminant is a logistic regression equation, a fractional equation, a linear discriminant, a multiple regression equation, or a support vector machine. It is any one of a created formula, a formula created by Mahalanobis distance method, a formula created by canonical discriminant analysis, and a formula created by a decision tree.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記判別値算出ステップは、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの前記濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを前記変数として含む前記多変量判別式に基づいて、前記判別値を算出すること、を特徴とする。 The cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the discriminant value calculating step includes Val included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. , Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Based on the multivariate discriminant including at least one of Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro as the variables. The discriminant value is calculated.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを前記変数として含む前記分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを前記変数として含む前記ロジスティック回帰式であること、を特徴とする。 The cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the multivariate discriminant is Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His. The fractional expression including at least one of the variables as the variable, or including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, Leu as the variable The logistic regression equation.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記濃度値基準評価ステップは、前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップをさらに含むこと、を特徴とする。 Further, the cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the concentration value reference evaluation step is based on the discriminant value calculated in the discriminant value calculation step. The method further comprises a discriminant value criterion evaluation step for evaluating the effect of the treatment on.
 また、本発明にかかる癌免疫療法の評価方法は、前記の癌免疫療法の評価方法において、前記判別値基準評価ステップは、前記判別値に基づいて、前記評価対象に対して前記治療が有効であるかを評価する(例えば、前記判別値に基づいて、前記評価対象に対して前記治療が有効であるか否かを判別する判別値基準判別ステップをさらに含む)こと、を特徴とする。 Further, the cancer immunotherapy evaluation method according to the present invention is the above-described cancer immunotherapy evaluation method, wherein the discriminant value criterion evaluation step is based on the discriminant value and the treatment is effective for the evaluation object. (E.g., further including a discrimination value criterion discrimination step for discriminating whether or not the treatment is effective for the evaluation object based on the discrimination value).
 また、本発明にかかる癌免疫療法評価装置は、制御部と記憶部とを備えた癌免疫療法評価装置であって、前記制御部は、癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出手段を備えたこと、を特徴とする。なお、前記判別値算出手段は、前記治療開始前アミノ酸濃度データと前記治療開始後アミノ酸濃度データに基づいて、前記治療が開始される前のアミノ酸の濃度値と前記治療が開始された後のアミノ酸の濃度値との比または差分を算出し、算出した各アミノ酸の濃度値の比または差分を多変量判別式に含まれる各変数に代入することで、判別値を算出してもよい。また、前記治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。 The cancer immunotherapy evaluation apparatus according to the present invention is a cancer immunotherapy evaluation apparatus including a control unit and a storage unit, and the control unit is configured to perform the treatment before treatment by cancer immunotherapy is started. Including the amino acid concentration data before the start of treatment regarding the concentration value of the amino acid to be evaluated, the amino acid concentration data after the start of treatment regarding the concentration value of the amino acid to be evaluated after the start of the treatment, and the amino acid concentration as variables Based on the multivariate discriminant stored in the storage unit, there is provided a discriminant value calculating means for calculating a discriminant value that is a value of the multivariate discriminant and corresponds to an evaluation result relating to the effect of the treatment on the evaluation target. It is characterized by this. In addition, the discriminant value calculating means is based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, and the amino acid concentration value before starting the treatment and the amino acid after starting the treatment. Alternatively, the discriminant value may be calculated by calculating the ratio or difference with the concentration value and substituting the calculated concentration value ratio or difference of each amino acid into each variable included in the multivariate discriminant. The amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data.
 また、本発明にかかる癌免疫療法評価装置は、前記の癌免疫療法評価装置において、前記制御部は、前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段をさらに備えたこと、を特徴とする。 The cancer immunotherapy evaluation apparatus according to the present invention is the cancer immunotherapy evaluation apparatus according to the present invention, wherein the control unit has an effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation means. And a discriminant value criterion evaluating means for evaluating the above.
 なお、本発明にかかる癌免疫療法評価装置は、前記の癌免疫療法評価装置において、(i)前記制御部は、アミノ酸濃度データと癌の状態を表す指標に関する癌状態指標データとを含む前記記憶部に記憶された癌状態情報に基づいて、前記記憶部で記憶する前記多変量判別式を作成する多変量判別式作成手段をさらに備え、(ii)前記多変量判別式作成手段は、前記癌状態情報から所定の式作成手法に基づいて、前記多変量判別式の候補である候補多変量判別式を作成する候補多変量判別式作成手段と、前記候補多変量判別式作成手段で作成した前記候補多変量判別式を、所定の検証手法に基づいて検証する候補多変量判別式検証手段と、所定の変数選択手法に基づいて前記候補多変量判別式の変数を選択することで、前記候補多変量判別式を作成する際に用いる前記癌状態情報に含まれる前記アミノ酸濃度データの組み合わせを選択する変数選択手段と、をさらに備え、前記候補多変量判別式作成手段、前記候補多変量判別式検証手段および前記変数選択手段を繰り返し実行して蓄積した前記検証結果に基づいて、複数の前記候補多変量判別式の中から前記多変量判別式として採用する前記候補多変量判別式を選出することで、前記多変量判別式を作成すること、を特徴としてもよい。 The cancer immunotherapy evaluation apparatus according to the present invention is the above-described cancer immunotherapy evaluation apparatus, wherein (i) the control unit includes amino acid concentration data and cancer state index data relating to an index representing a cancer state. A multivariate discriminant creating unit that creates the multivariate discriminant stored in the storage unit based on the cancer state information stored in the unit; and (ii) the multivariate discriminant creating unit includes the cancer Candidate multivariate discriminant creating means for creating a candidate multivariate discriminant that is a candidate for the multivariate discriminant based on a predetermined formula creating method from state information, and the candidate multivariate discriminant creating means created by the candidate multivariate discriminant creating means The candidate multivariate discriminant verification means for verifying the candidate multivariate discriminant based on a predetermined verification method, and selecting the candidate multivariate discriminant variable based on the predetermined variable selection method, Variable size Variable selection means for selecting a combination of the amino acid concentration data included in the cancer state information used when creating an expression, the candidate multivariate discriminant creation means, the candidate multivariate discriminant verification means, and By selecting the candidate multivariate discriminant to be adopted as the multivariate discriminant from a plurality of the candidate multivariate discriminants based on the verification results accumulated by repeatedly executing the variable selection unit, It may be characterized by creating a multivariate discriminant.
 また、本発明にかかる癌免疫療法評価方法は、制御部と記憶部とを備えた情報処理装置において実行される癌免疫療法評価方法であって、前記制御部において実行される、癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップを含むこと、を特徴とする。 The cancer immunotherapy evaluation method according to the present invention is a cancer immunotherapy evaluation method executed in an information processing apparatus including a control unit and a storage unit, and is based on cancer immunotherapy executed in the control unit. Amino acid concentration data before the start of treatment regarding the concentration value of the evaluation target amino acid before the start of treatment, and an amino acid concentration data after the start of treatment regarding the concentration value of the amino acid of the evaluation target after the start of the treatment Based on the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation target is a value of the multivariate discriminant And a discriminant value calculating step for calculating.
 また、本発明にかかる癌免疫療法評価方法は、前記の癌免疫療法評価方法において、前記制御部において実行される、前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップをさらに含むこと、を特徴とする。 Further, the cancer immunotherapy evaluation method according to the present invention is the cancer immunotherapy evaluation method according to the present invention, based on the discriminant value calculated in the discriminant value calculation step executed in the control unit, with respect to the evaluation object. The method further includes a discriminant value criterion evaluation step for evaluating the effect of treatment.
 また、本発明にかかる癌免疫療法評価プログラムは、制御部と記憶部とを備えた情報処理装置において実行させるための癌免疫療法評価プログラムであって、前記制御部において実行させるための、癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップを含むこと、を特徴とする。 A cancer immunotherapy evaluation program according to the present invention is a cancer immunotherapy evaluation program to be executed in an information processing apparatus including a control unit and a storage unit, and the cancer immunity evaluation program to be executed in the control unit. Amino acid concentration data before the start of treatment regarding the concentration value of the evaluation target amino acid before the treatment by the therapy, and the amino acid after the start of treatment regarding the concentration value of the evaluation target amino acid after the start of the treatment Based on the concentration data and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the value of the multivariate discriminant corresponds to the evaluation result regarding the effect of the treatment on the evaluation target. And a discriminant value calculating step for calculating a discriminant value.
 また、本発明にかかる癌免疫療法評価プログラムは、前記の癌免疫療法評価プログラムにおいて、前記制御部において実行させるための、前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップをさらに含むこと、を特徴とする。 The cancer immunotherapy evaluation program according to the present invention is based on the discriminant value calculated in the discriminant value calculation step for the control unit to execute in the cancer immunotherapy evaluation program. The method further comprises a discriminant value criterion evaluation step for evaluating the effect of the treatment.
 また、本発明にかかる記録媒体は、一時的でないコンピュータ読み取り可能な記録媒体であって、情報処理装置に前記癌免疫療法評価方法を実行させるためのプログラム化された命令を含むこと、を特徴とする。 A recording medium according to the present invention is a non-transitory computer-readable recording medium, and includes a programmed instruction for causing an information processing apparatus to execute the cancer immunotherapy evaluation method. To do.
 また、本発明にかかる癌免疫療法評価システムは、制御部と記憶部とを備えた癌免疫療法評価装置と、制御部を備え、癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置とを、ネットワークを介して通信可能に接続して構成された癌免疫療法評価システムであって、前記情報通信端末装置の前記制御部は、前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを、前記癌免疫療法評価装置へ送信するアミノ酸濃度データ送信手段と、前記癌免疫療法評価装置から送信された、アミノ酸の濃度を変数として含む多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を受信する結果受信手段とを備え、前記癌免疫療法評価装置の前記制御部は、前記情報通信端末装置から送信された前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データを受信するアミノ酸濃度データ受信手段と、前記アミノ酸濃度データ受信手段で受信した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびに前記記憶部に記憶された前記多変量判別式に基づいて、前記判別値を算出する判別値算出手段と、前記判別値算出手段で算出した前記判別値を前記情報通信端末装置へ送信する結果送信手段と、を備えたこと、を特徴とする。 The cancer immunotherapy evaluation system according to the present invention includes a cancer immunotherapy evaluation apparatus including a control unit and a storage unit, and an amino acid related to a concentration value of an amino acid to be evaluated that includes a control unit and receives treatment by cancer immunotherapy. A cancer immunotherapy evaluation system configured to connect an information communication terminal device that provides concentration data to be communicable via a network, wherein the control unit of the information communication terminal device starts the treatment Pre-treatment amino acid concentration data relating to the concentration value of the evaluation target amino acid before treatment, and post-treatment amino acid concentration data relating to the concentration value of the evaluation target amino acid after initiation of the treatment, the cancer immunotherapy evaluation An amino acid concentration data transmitting means for transmitting to the apparatus; and a multivariate format including the amino acid concentration transmitted from the cancer immunotherapy evaluation apparatus as a variable. And a result receiving means for receiving a discriminant value corresponding to an evaluation result relating to the effect of the treatment on the evaluation target, wherein the control unit of the cancer immunotherapy evaluation device receives the information from the information communication terminal device. Amino acid concentration data receiving means for receiving the transmitted pre-treatment amino acid concentration data and the post-treatment amino acid concentration data, and the pre-treatment amino acid concentration data and the post-treatment amino acid received by the amino acid concentration data receiving means Based on the density data and the multivariate discriminant stored in the storage unit, the discriminant value calculating means for calculating the discriminant value, and the discriminant value calculated by the discriminant value calculating means to the information communication terminal device And a result transmitting means for transmitting.
 また、本発明にかかる癌免疫療法評価システムは、前記の癌免疫療法評価システムにおいて、前記癌免疫療法評価装置の前記制御部は、前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段をさらに備え、前記結果送信手段は、前記判別値基準評価手段で得られた前記評価対象の前記評価結果を前記情報通信端末装置へ送信し、前記結果受信手段は、前記癌免疫療法評価装置から送信された前記評価対象の前記評価結果を受信すること、を特徴とする。 Further, the cancer immunotherapy evaluation system according to the present invention is the cancer immunotherapy evaluation system, wherein the control unit of the cancer immunotherapy evaluation device is based on the discriminant value calculated by the discriminant value calculation means. The apparatus further comprises discriminant value reference evaluation means for evaluating the effect of the treatment on the evaluation object, and the result transmitting means transmits the evaluation result of the evaluation object obtained by the discriminant value reference evaluation means to the information communication terminal device. The result receiving means receives the evaluation result of the evaluation target transmitted from the cancer immunotherapy evaluation apparatus.
 また、本発明にかかる情報通信端末装置は、制御部を備え、癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置であって、前記制御部は、アミノ酸の濃度を変数として含む多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を取得する結果取得手段を備え、前記判別値は、前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、および前記多変量判別式に基づいて算出したものであること、を特徴とする。 The information communication terminal device according to the present invention is an information communication terminal device that includes a control unit, and provides amino acid concentration data related to the concentration value of the amino acid to be evaluated that receives treatment by cancer immunotherapy, wherein the control unit includes: , Comprising a result acquisition means for acquiring a discriminant value corresponding to an evaluation result relating to the effect of the treatment on the evaluation target, which is a value of a multivariate discriminant including an amino acid concentration as a variable. Amino acid concentration data before starting treatment regarding the concentration value of the amino acid to be evaluated before starting, amino acid concentration data after starting treatment regarding the concentration value of the amino acid to be evaluated after starting the treatment, and multivariate discrimination It is calculated based on a formula.
 また、本発明にかかる情報通信端末装置は、前記の情報通信端末装置において、前記結果取得手段は、前記評価対象に対する前記治療の効果に関する前記評価結果を取得し、前記評価結果は、前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価して得られた結果であること、を特徴とする。 Moreover, in the information communication terminal device according to the present invention, in the information communication terminal device, the result acquisition unit acquires the evaluation result regarding the effect of the treatment on the evaluation target, and the evaluation result is the discriminant value. This is a result obtained by evaluating the effect of the treatment on the evaluation object.
 また、本発明にかかる情報通信端末装置は、前記の情報通信端末装置において、前記多変量判別式を記憶し、前記判別値を算出する癌免疫療法評価装置とネットワークを介して通信可能に接続されており、前記制御部は、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データを、前記癌免疫療法評価装置へ送信するアミノ酸濃度データ送信手段をさらに備え、前記結果取得手段は、前記癌免疫療法評価装置から送信された前記判別値を受信すること、を特徴とする。 The information communication terminal device according to the present invention is connected to the cancer immunotherapy evaluation device that stores the multivariate discriminant and calculates the discriminant value via the network in the information communication terminal device. The control unit further comprises amino acid concentration data transmitting means for transmitting the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data to the cancer immunotherapy evaluation device, and the result acquisition means includes the The discriminant value transmitted from the cancer immunotherapy evaluation device is received.
 また、本発明にかかる情報通信端末装置は、前記の情報通信端末装置において、前記癌免疫療法評価装置は、前記治療の効果をさらに評価し、前記結果取得手段は、前記癌免疫療法評価装置から送信された、前記評価対象に対する前記治療の効果に関する前記評価結果を受信すること、を特徴とする。 Moreover, the information communication terminal device according to the present invention is the information communication terminal device, wherein the cancer immunotherapy evaluation device further evaluates the effect of the treatment, and the result acquisition means includes the cancer immunotherapy evaluation device. The transmitted evaluation result relating to the effect of the treatment on the evaluation target is transmitted.
 また、本発明にかかる癌免疫療法評価装置は、癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置とネットワークを介して通信可能に接続された、制御部と記憶部とを備えた癌免疫療法評価装置であって、前記制御部は、前記情報通信端末装置から送信された、前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを受信するアミノ酸濃度データ受信手段と、前記アミノ酸濃度データ受信手段で受信した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出手段と、前記判別値算出手段で算出した前記判別値を前記情報通信端末装置へ送信する結果送信手段と、を備えたこと、を特徴とする。 In addition, the cancer immunotherapy evaluation apparatus according to the present invention is communicably connected via an information communication terminal device that provides amino acid concentration data related to the concentration value of an amino acid to be evaluated that receives treatment by cancer immunotherapy, A cancer immunotherapy evaluation device including a control unit and a storage unit, wherein the control unit relates to a concentration value of the evaluation target amino acid transmitted from the information communication terminal device before the treatment is started. Amino acid concentration data receiving means for receiving amino acid concentration data before starting treatment and amino acid concentration data after starting treatment related to the concentration value of the amino acid to be evaluated after the treatment is started, and received by the amino acid concentration data receiving means The amino acid concentration data before the start of treatment and the amino acid concentration data after the start of treatment, and the concentration of amino acids as variables Based on the multivariate discriminant stored in the storage unit, the discriminant value calculating means calculates the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation object, which is the value of the multivariate discriminant. And a result transmitting means for transmitting the discriminant value calculated by the discriminant value calculating means to the information communication terminal device.
 また、本発明にかかる癌免疫療法評価装置は、前記の癌免疫療法評価装置において、前記制御部は、前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段をさらに備え、前記結果送信手段は、前記判別値基準評価手段で得られた前記評価対象の前記評価結果を前記情報通信端末装置へ送信すること、を特徴とする。 The cancer immunotherapy evaluation apparatus according to the present invention is the cancer immunotherapy evaluation apparatus according to the present invention, wherein the control unit has an effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation means. Further comprising: a discriminant value criterion-evaluating unit, wherein the result transmitting unit transmits the evaluation result of the evaluation target obtained by the discriminant value criterion-evaluating unit to the information communication terminal device. .
 本発明によれば、癌免疫療法による治療を受ける評価対象から治療が開始される前に採取された血液中のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および評価対象から治療が開始された後に採取された血液中のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを取得し、取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに基づいて、評価対象に対する治療の効果を評価する。これにより、血液中のアミノ酸の濃度を利用して癌免疫療法の治療効果を精度よく評価することができるという効果を奏する。なお、治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。これにより、当該治療効果を、治療終了後も継続的にモニタリングすることができるという効果を奏する。 According to the present invention, amino acid concentration data before starting treatment related to the concentration value of amino acids in blood collected before starting treatment from an evaluation subject receiving treatment by cancer immunotherapy, and treatment was started from the evaluation subject. Acquire amino acid concentration data after the start of treatment regarding the concentration value of amino acids in blood collected later, and evaluate the effect of treatment on the evaluation target based on the acquired amino acid concentration data before starting treatment and amino acid concentration data after starting treatment. . Thereby, there exists an effect that the therapeutic effect of cancer immunotherapy can be accurately evaluated using the concentration of amino acids in blood. The amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data. Thereby, there exists an effect that the said therapeutic effect can be continuously monitored even after completion | finish of a treatment.
 また、本発明によれば、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値に基づいて、評価対象に対する治療の効果を評価する。これにより、癌免疫療法の治療効果の評価に有用なアミノ酸の濃度を利用して、当該治療効果を精度よく評価することができるという効果を奏する。 Further, according to the present invention, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, which are included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. Based on the concentration value of at least one of Met, Lys, Arg, Gly, Cys2, and Pro, the effect of treatment on the evaluation target is evaluated. This produces an effect that the therapeutic effect can be accurately evaluated using the amino acid concentration useful for evaluating the therapeutic effect of cancer immunotherapy.
 また、本発明によれば、濃度値に基づいて、評価対象に対して治療が有効であるかを評価する(例えば、濃度値に基づいて、評価対象に対して治療が有効であるか否かを判別する)。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別)に有用なアミノ酸の濃度を利用して、当該評価(例えば判別)を精度よく行うことができるという効果を奏する。 Further, according to the present invention, it is evaluated whether the treatment is effective for the evaluation object based on the concentration value (for example, whether the treatment is effective for the evaluation object based on the concentration value). To determine). This produces an effect that the evaluation (for example, discrimination) can be accurately performed by using the amino acid concentration useful for the evaluation (for example, discrimination) regarding the effectiveness of the therapeutic effect of cancer immunotherapy.
 また、本発明によれば、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む予め設定された多変量判別式に基づいて、当該多変量判別式の値であって評価対象に対する治療の効果に関する評価結果に相当する判別値を算出する。例えば、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データに基づいて、治療が開始される前のアミノ酸の濃度値と治療が開始された後のアミノ酸の濃度値との比または差分を算出し、算出した各アミノ酸の濃度値の比または差分を多変量判別式に含まれる各変数に代入することで、判別値を算出してもよい。これにより、アミノ酸の濃度を変数として含む多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができるという効果を奏する。また、治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。これにより、当該治療効果を、治療終了後も継続的にモニタリングすることができるという効果を奏する。 Further, according to the present invention, based on the pre-treatment amino acid concentration data and post-treatment amino acid concentration data, and a preset multivariate discriminant including the amino acid concentration as a variable, the value of the multivariate discriminant Then, a discriminant value corresponding to the evaluation result regarding the effect of the treatment on the evaluation target is calculated. For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated. The discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant. Thereby, using the multivariate discriminant including the amino acid concentration as a variable, it is possible to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target. Further, the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment). Thereby, there exists an effect that the said therapeutic effect can be continuously monitored even after completion | finish of a treatment.
 また、本発明によれば、多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つである。これにより、癌免疫療法の治療効果の評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができるという効果を奏する。 Further, according to the present invention, the multivariate discriminant is a logistic regression equation, a fractional equation, a linear discriminant equation, a multiple regression equation, an equation created by a support vector machine, an equation created by the Mahalanobis distance method, a canonical discriminant. One of an expression created by analysis and an expression created by a decision tree. Thereby, using the multivariate discriminant useful for evaluating the therapeutic effect of cancer immunotherapy, it is possible to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target.
 また、本発明によれば、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、判別値を算出する。これにより、癌免疫療法の治療効果の評価に有用なアミノ酸の濃度を変数として含む当該評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができるという効果を奏する。 Further, according to the present invention, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, which are included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. At least one concentration value of Met, Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, A discriminant value is calculated based on a multivariate discriminant including at least one of Arg, Gly, Cys2, and Pro as a variable. As a result, a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. There is an effect that can be.
 また、本発明によれば、多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを変数として含む分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを変数として含むロジスティック回帰式である。これにより、癌免疫療法の治療効果の評価に特に有用なアミノ酸の濃度を変数として含む当該評価に特に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができるという効果を奏する。 According to the present invention, the multivariate discriminant is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable, or Trp, Thr, This is a logistic regression equation including at least one of His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable. This makes it possible to use a multivariate discriminant that is particularly useful for the evaluation including the concentration of amino acids that are particularly useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. There is an effect that can be obtained.
 また、本発明によれば、判別値に基づいて、評価対象に対する治療の効果を評価する。これにより、アミノ酸の濃度を変数として含む多変量判別式で得られる判別値を利用して、当該治療効果を精度よく評価することができるという効果を奏する。 Further, according to the present invention, the effect of treatment on the evaluation target is evaluated based on the discriminant value. Accordingly, the therapeutic effect can be accurately evaluated by using the discriminant value obtained by the multivariate discriminant including the amino acid concentration as a variable.
 また、本発明によれば、判別値に基づいて、評価対象に対して治療が有効であるかを評価する(例えば、判別値に基づいて、評価対象に対して治療が有効であるか否かを判別する)。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別)に有用なアミノ酸の濃度を変数として含む当該評価(例えば判別)に有用な多変量判別式で得られる判別値を利用して、当該評価(例えば判別)を精度よく行うことができるという効果を奏する。 Further, according to the present invention, it is evaluated whether the treatment is effective for the evaluation target based on the discriminant value (for example, whether the treatment is effective for the evaluation target based on the discriminant value). To determine). By using the discriminant value obtained by the multivariate discriminant useful for the evaluation (for example, discrimination) including the concentration of amino acid useful for evaluation (for example, discrimination) regarding the effectiveness of the therapeutic effect of cancer immunotherapy as a variable. The effect (for example, discrimination) can be performed with high accuracy.
 なお、本発明によれば、アミノ酸濃度データと癌の状態を表す指標に関する癌状態指標データとを含む記憶手段に記憶された癌状態情報に基づいて、記憶手段で記憶する多変量判別式を作成してもよい。具体的には、(i)癌状態情報から所定の式作成手法に基づいて候補多変量判別式を作成し、(ii)作成した候補多変量判別式を所定の検証手法に基づいて検証し、(iii)所定の変数選択手法に基づいて候補多変量判別式の変数を選択することで、候補多変量判別式を作成する際に用いる癌状態情報に含まれるアミノ酸濃度データの組み合わせを選択し、(iv)(i)、(ii)および(iii)を繰り返し実行して蓄積した検証結果に基づいて、複数の候補多変量判別式の中から多変量判別式として採用する候補多変量判別式を選出することで、多変量判別式を作成してもよい。これにより、癌免疫療法の治療効果の評価に最適な多変量判別式を作成することができるという効果を奏する。 According to the present invention, the multivariate discriminant stored in the storage unit is created based on the cancer state information stored in the storage unit including the amino acid concentration data and the cancer state index data relating to the index representing the cancer state. May be. Specifically, (i) creating a candidate multivariate discriminant based on a predetermined formula creation method from cancer state information, (ii) verifying the created candidate multivariate discriminant based on a predetermined verification method, (Iii) by selecting a variable of the candidate multivariate discriminant based on a predetermined variable selection method, selecting a combination of amino acid concentration data included in the cancer state information used when creating the candidate multivariate discriminant, (Iv) A candidate multivariate discriminant to be adopted as a multivariate discriminant from among a plurality of candidate multivariate discriminants based on the verification results accumulated by repeatedly executing (i), (ii) and (iii) A multivariate discriminant may be created by selection. Thereby, there exists an effect that the multivariate discriminant most suitable for evaluation of the therapeutic effect of cancer immunotherapy can be created.
 なお、本発明では、癌免疫療法の治療効果を評価する際、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。また、本発明では、癌免疫療法の治療効果を評価する際、多変量判別式における変数として、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。 In the present invention, when evaluating the therapeutic effect of cancer immunotherapy, in addition to the amino acid concentration, other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, immunocompetent intracellular cytokine, delayed type) An excessive reaction (DTH, etc.) may be further used. Further, in the present invention, when evaluating the therapeutic effect of cancer immunotherapy, as a variable in the multivariate discriminant, in addition to the amino acid concentration, other biological information (for example, tumor marker, blood cytokine, number of immunocompetent cells, Intracellular cytokines such as immunocompetent cells and delayed hyperfractionation (DTH) may be further used.
図1は、第1実施形態の基本原理を示す原理構成図である。FIG. 1 is a principle configuration diagram showing the basic principle of the first embodiment. 図2は、第1実施形態にかかる癌免疫療法の評価方法の一例を示すフローチャートである。FIG. 2 is a flowchart showing an example of the cancer immunotherapy evaluation method according to the first embodiment. 図3は、第2実施形態の基本原理を示す原理構成図である。FIG. 3 is a principle configuration diagram showing the basic principle of the second embodiment. 図4は、本システムの全体構成の一例を示す図である。FIG. 4 is a diagram illustrating an example of the overall configuration of the present system. 図5は、本システムの全体構成の他の一例を示す図である。FIG. 5 is a diagram showing another example of the overall configuration of the present system. 図6は、本システムの癌免疫療法評価装置100の構成の一例を示すブロック図である。FIG. 6 is a block diagram showing an example of the configuration of the cancer immunotherapy evaluation apparatus 100 of the present system. 図7は、利用者情報ファイル106aに格納される情報の一例を示す図である。FIG. 7 is a diagram illustrating an example of information stored in the user information file 106a. 図8は、アミノ酸濃度データファイル106bに格納される情報の一例を示す図である。FIG. 8 is a diagram showing an example of information stored in the amino acid concentration data file 106b. 図9は、癌状態情報ファイル106cに格納される情報の一例を示す図である。FIG. 9 is a diagram illustrating an example of information stored in the cancer state information file 106c. 図10は、指定癌状態情報ファイル106dに格納される情報の一例を示す図である。FIG. 10 is a diagram illustrating an example of information stored in the designated cancer state information file 106d. 図11は、候補多変量判別式ファイル106e1に格納される情報の一例を示す図である。FIG. 11 is a diagram illustrating an example of information stored in the candidate multivariate discriminant file 106e1. 図12は、検証結果ファイル106e2に格納される情報の一例を示す図である。FIG. 12 is a diagram illustrating an example of information stored in the verification result file 106e2. 図13は、選択癌状態情報ファイル106e3に格納される情報の一例を示す図である。FIG. 13 is a diagram illustrating an example of information stored in the selected cancer state information file 106e3. 図14は、多変量判別式ファイル106e4に格納される情報の一例を示す図である。FIG. 14 is a diagram illustrating an example of information stored in the multivariate discriminant file 106e4. 図15は、判別値ファイル106fに格納される情報の一例を示す図である。FIG. 15 is a diagram illustrating an example of information stored in the discrimination value file 106f. 図16は、評価結果ファイル106gに格納される情報の一例を示す図である。FIG. 16 is a diagram illustrating an example of information stored in the evaluation result file 106g. 図17は、多変量判別式作成部102hの構成を示すブロック図である。FIG. 17 is a block diagram showing a configuration of the multivariate discriminant-preparing part 102h. 図18は、判別値基準評価部102jの構成を示すブロック図である。FIG. 18 is a block diagram illustrating a configuration of the discriminant value criterion-evaluating unit 102j. 図19は、本システムのクライアント装置200の構成の一例を示すブロック図である。FIG. 19 is a block diagram illustrating an example of the configuration of the client apparatus 200 of the present system. 図20は、本システムのデータベース装置400の構成の一例を示すブロック図である。FIG. 20 is a block diagram showing an example of the configuration of the database apparatus 400 of this system. 図21は、本システムで行う癌免疫療法評価サービス処理の一例を示すフローチャートである。FIG. 21 is a flowchart showing an example of a cancer immunotherapy evaluation service process performed in the present system. 図22は、本システムの癌免疫療法評価装置100で行う多変量判別式作成処理の一例を示すフローチャートである。FIG. 22 is a flowchart showing an example of multivariate discriminant creation processing performed by the cancer immunotherapy evaluation apparatus 100 of the present system. 図23は、実施例1の実験プロトコルを示す図である。FIG. 23 is a diagram showing an experimental protocol of Example 1. 図24は、免疫治療に対するレスポンダーの腫瘍増殖の変化を示す図である。FIG. 24 shows changes in responder tumor growth relative to immunotherapy. 図25は、免疫治療に対するノンレスポンダーの腫瘍増殖の変化を示す図である。FIG. 25 shows changes in tumor growth of non-responders with respect to immunotherapy. 図26は、レスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 26 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in the responder. 図27は、レスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 27 is a diagram showing the distribution of amino acid variables between the two groups before and after the start of treatment in the responder. 図28は、ノンレスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 28 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder. 図29は、ノンレスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 29 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder. 図30は、レスポンダーにおける治療開始後のアミノ酸変数の分布を示すレーダーチャートである。FIG. 30 is a radar chart showing the distribution of amino acid variables after the start of treatment in the responder. 図31は、ノンレスポンダーにおける治療開始後のアミノ酸変数の分布を示すレーダーチャートである。FIG. 31 is a radar chart showing the distribution of amino acid variables after the start of treatment in a non-responder. 図32は、各アミノ酸変数のROC曲線のAUCを示す図である。FIG. 32 is a diagram showing the AUC of the ROC curve of each amino acid variable. 図33は、各アミノ酸変数のROC曲線のAUCを示す図である。FIG. 33 is a diagram showing the AUC of the ROC curve of each amino acid variable. 図34は、分数式の一覧を示す図である。FIG. 34 is a diagram showing a list of fractional expressions. 図35は、図34に挙げられた分数式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 35 is a diagram showing the appearance frequency of amino acid variables included in the fractional expression given in FIG. 図36は、ロジスティック回帰式の一覧を示す図である。FIG. 36 is a diagram showing a list of logistic regression equations. 図37は、ロジスティック回帰式の一覧を示す図である。FIG. 37 is a diagram showing a list of logistic regression equations. 図38は、ロジスティック回帰式の一覧を示す図である。FIG. 38 is a diagram showing a list of logistic regression equations. 図39は、図36、図37および図38に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 39 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations listed in FIGS. 36, 37 and 38. 図40は、ロジスティック回帰式の一覧を示す図である。FIG. 40 is a diagram showing a list of logistic regression equations. 図41は、ロジスティック回帰式の一覧を示す図である。FIG. 41 is a diagram showing a list of logistic regression equations. 図42は、ロジスティック回帰式の一覧を示す図である。FIG. 42 is a diagram showing a list of logistic regression equations. 図43は、図40、図41および図42に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 43 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS. 40, 41, and 42. 図44は、ロジスティック回帰式の一覧を示す図である。FIG. 44 is a diagram showing a list of logistic regression equations. 図45は、ロジスティック回帰式の一覧を示す図である。FIG. 45 is a diagram showing a list of logistic regression equations. 図46は、ロジスティック回帰式の一覧を示す図である。FIG. 46 is a diagram showing a list of logistic regression equations. 図47は、ロジスティック回帰式の一覧を示す図である。FIG. 47 is a diagram showing a list of logistic regression equations. 図48は、ロジスティック回帰式の一覧を示す図である。FIG. 48 is a diagram showing a list of logistic regression equations. 図49は、図44-48に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 49 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS. 44-48. 図50は、ロジスティック回帰式の一覧を示す図である。FIG. 50 is a diagram showing a list of logistic regression equations. 図51は、ロジスティック回帰式の一覧を示す図である。FIG. 51 is a diagram showing a list of logistic regression equations. 図52は、ロジスティック回帰式の一覧を示す図である。FIG. 52 is a diagram showing a list of logistic regression equations. 図53は、ロジスティック回帰式の一覧を示す図である。FIG. 53 is a diagram showing a list of logistic regression equations. 図54は、ロジスティック回帰式の一覧を示す図である。FIG. 54 is a diagram showing a list of logistic regression equations. 図55は、図50-54に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 55 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations shown in FIGS. 図56は、実施例6の実験プロトコルを示す図である。FIG. 56 is a diagram showing the experimental protocol of Example 6. 図57は、腫瘍組織の切片画像を示す図である。FIG. 57 is a diagram showing a section image of a tumor tissue. 図58は、レスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 58 is a diagram showing the distribution of amino acid variables between the two groups before and after the start of treatment in the responder. 図59は、レスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 59 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a responder. 図60は、ノンレスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 60 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder. 図61は、ノンレスポンダーにおける治療開始前と治療開始後の2群間のアミノ酸変数の分布を示す図である。FIG. 61 is a diagram showing the distribution of amino acid variables between two groups before and after the start of treatment in a non-responder. 図62は、レスポンダーにおける治療開始後のアミノ酸変数の分布を示すレーダーチャートである。FIG. 62 is a radar chart showing the distribution of amino acid variables after the start of treatment in the responder. 図63は、ノンレスポンダーにおける治療開始後のアミノ酸変数の分布を示すレーダーチャートである。FIG. 63 is a radar chart showing the distribution of amino acid variables after the start of treatment in a non-responder. 図64は、各アミノ酸変数のROC曲線のAUCを示す図である。FIG. 64 is a diagram showing the AUC of the ROC curve of each amino acid variable. 図65は、各アミノ酸変数のROC曲線のAUCを示す図である。FIG. 65 is a diagram showing the AUC of the ROC curve of each amino acid variable. 図66は、分数式の一覧を示す図である。FIG. 66 is a diagram showing a list of fractional expressions. 図67は、図66に挙げられた分数式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 67 is a diagram showing the appearance frequency of amino acid variables included in the fractional expression given in FIG. 図68は、ロジスティック回帰式の一覧を示す図である。FIG. 68 is a diagram showing a list of logistic regression equations. 図69は、ロジスティック回帰式の一覧を示す図である。FIG. 69 is a diagram showing a list of logistic regression equations. 図70は、ロジスティック回帰式の一覧を示す図である。FIG. 70 is a diagram showing a list of logistic regression equations. 図71は、図68、図69および図70に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。71 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations listed in FIGS. 68, 69, and 70. FIG. 図72は、ロジスティック回帰式の一覧を示す図である。FIG. 72 is a diagram showing a list of logistic regression equations. 図73は、ロジスティック回帰式の一覧を示す図である。FIG. 73 is a diagram showing a list of logistic regression equations. 図74は、ロジスティック回帰式の一覧を示す図である。FIG. 74 is a diagram showing a list of logistic regression equations. 図75は、図72、図73および図74に挙げられたロジスティック回帰式に含まれるアミノ酸変数の出現頻度を示す図である。FIG. 75 is a diagram showing the appearance frequency of amino acid variables included in the logistic regression equations given in FIGS. 72, 73 and 74. 図76は、ロジスティック回帰式の一覧を示す図である。FIG. 76 is a diagram showing a list of logistic regression equations. 図77は、ロジスティック回帰式の一覧を示す図である。FIG. 77 is a diagram showing a list of logistic regression equations. 図78は、ロジスティック回帰式の一覧を示す図である。FIG. 78 is a diagram showing a list of logistic regression equations. 図79は、ロジスティック回帰式の一覧を示す図である。FIG. 79 is a diagram showing a list of logistic regression equations. 図80は、ロジスティック回帰式の一覧を示す図である。FIG. 80 is a diagram showing a list of logistic regression equations. 図81は、ロジスティック回帰式の一覧を示す図である。FIG. 81 is a diagram showing a list of logistic regression equations. 図82は、ロジスティック回帰式の一覧を示す図である。FIG. 82 is a diagram showing a list of logistic regression equations. 図83は、ロジスティック回帰式の一覧を示す図である。FIG. 83 is a diagram showing a list of logistic regression equations. 図84は、ロジスティック回帰式の一覧を示す図である。FIG. 84 is a diagram showing a list of logistic regression equations. 図85は、ロジスティック回帰式の一覧を示す図である。FIG. 85 is a diagram showing a list of logistic regression equations.
 以下に、癌免疫療法の評価方法の実施形態(第1実施形態)、ならびに癌免疫療法評価装置、癌免疫療法評価方法、癌免疫療法評価プログラム、記録媒体、癌免疫療法評価システムおよび情報通信端末装置の実施形態(第2実施形態)を、図面に基づいて詳細に説明する。なお、本発明はこれらの実施形態により限定されるものではない。 Embodiments of cancer immunotherapy evaluation method (first embodiment), cancer immunotherapy evaluation apparatus, cancer immunotherapy evaluation method, cancer immunotherapy evaluation program, recording medium, cancer immunotherapy evaluation system, and information communication terminal An apparatus embodiment (second embodiment) will be described in detail with reference to the drawings. Note that the present invention is not limited to these embodiments.
[第1実施形態]
[1-1.第1実施形態の概要]
 ここでは、第1実施形態の概要について図1を参照して説明する。図1は第1実施形態の基本原理を示す原理構成図である。
[First Embodiment]
[1-1. Overview of First Embodiment]
Here, an overview of the first embodiment will be described with reference to FIG. FIG. 1 is a principle configuration diagram showing the basic principle of the first embodiment.
 まず、癌免疫療法による治療を受ける評価対象(例えば動物やヒトなどの個体)から治療開始前に採取した血液(例えば血漿、血清などを含む)中のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および当該評価対象から治療開始後に採取した血液中のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを取得する(ステップS11)。なお、ステップS11では、例えば、アミノ酸濃度測定を行う企業等が測定したアミノ酸濃度データを取得してもよく、また、評価対象から採取した血液から、例えば以下の(A)または(B)などの測定方法でアミノ酸濃度データを測定することでアミノ酸濃度データを取得してもよい。ここで、アミノ酸濃度の単位は、例えばモル濃度や重量濃度、これらの濃度に任意の定数を加減乗除することで得られるものでもよい。
(A)採取した血液サンプルを遠心することにより血液から血漿を分離した。全ての血漿サンプルは、アミノ酸濃度の測定時まで-80℃で凍結保存した。アミノ酸濃度測定時には、アセトニトリルを添加し除蛋白処理を行った後、標識試薬(3-アミノピリジル-N-ヒドロキシスクシンイミジルカルバメート)を用いてプレカラム誘導体化を行い、そして、液体クロマトグラフ質量分析計(LC/MS)によりアミノ酸濃度を分析した(国際公開第2003/069328号、国際公開第2005/116629号を参照)。
(B)採取した血液サンプルを遠心することにより血液から血漿を分離した。全ての血漿サンプルは、アミノ酸濃度の測定時まで-80℃で凍結保存した。アミノ酸濃度測定時には、スルホサリチル酸を添加し除蛋白処理を行った後、ニンヒドリン試薬を用いたポストカラム誘導体化法を原理としたアミノ酸分析計によりアミノ酸濃度を分析した。
First, the amino acid concentration data before the start of treatment relating to the concentration value of amino acids in blood (including plasma, serum, etc.) collected before the start of treatment from an evaluation subject (for example, an individual such as an animal or a human) who receives treatment by cancer immunotherapy And the amino acid concentration data after the start of treatment regarding the concentration value of the amino acid in the blood collected after the start of the treatment from the evaluation target is acquired (step S11). In step S11, for example, amino acid concentration data measured by a company or the like that performs amino acid concentration measurement may be acquired. Further, for example, the following (A) or (B) may be obtained from blood collected from an evaluation target. Amino acid concentration data may be obtained by measuring amino acid concentration data by a measurement method. Here, the unit of amino acid concentration may be obtained by, for example, molar concentration, weight concentration, or by adding / subtracting / subtracting an arbitrary constant to / from these concentrations.
(A) Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at −80 ° C. until the measurement of amino acid concentration. For amino acid concentration measurement, acetonitrile was added to remove protein, followed by precolumn derivatization using a labeling reagent (3-aminopyridyl-N-hydroxysuccinimidyl carbamate), and liquid chromatography mass spectrometry The amino acid concentration was analyzed by a total (LC / MS) (see International Publication No. 2003/069328 and International Publication No. 2005/116629).
(B) Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at −80 ° C. until the measurement of amino acid concentration. When measuring the amino acid concentration, sulfosalicylic acid was added to remove the protein, and then the amino acid concentration was analyzed by an amino acid analyzer based on the post-column derivatization method using a ninhydrin reagent.
 つぎに、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに基づいて、評価対象に対する癌免疫療法による治療の効果を評価する(ステップS12)。 Next, based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data acquired in step S11, the effect of treatment by cancer immunotherapy on the evaluation target is evaluated (step S12).
 以上、第1実施形態によれば、評価対象の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データを取得し、取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに基づいて、評価対象に対する治療の効果を評価する(要するに、評価対象に対する治療の効果を評価するための情報を提供する)。これにより、血液中のアミノ酸の濃度を利用して癌免疫療法の治療効果を精度よく評価することができる(要するに、治療の効果を評価するための精度のよい情報を提供することができる)。なお、治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。これにより、当該治療効果を、治療終了後も継続的にモニタリングすることができる。 As described above, according to the first embodiment, the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data to be evaluated are acquired, and evaluation is performed based on the acquired pre-treatment amino acid concentration data and post-treatment amino acid concentration data. Evaluate the effect of treatment on the subject (in short, provide information to assess the effect of treatment on the subject to be evaluated). Thereby, it is possible to accurately evaluate the therapeutic effect of cancer immunotherapy using the concentration of amino acid in blood (in short, accurate information for evaluating the therapeutic effect can be provided). The amino acid concentration data after the start of treatment may be data (after treatment amino acid concentration data) corresponding to the above-mentioned “after treatment” amino acid concentration data. Thereby, the said therapeutic effect can be continuously monitored even after completion | finish of a treatment.
 ここで、ステップS12を実行する前に、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データから欠損値や外れ値などのデータを除去してもよい。これにより、癌免疫療法の治療効果を精度よく評価することができる。 Here, before executing step S12, data such as missing values and outliers may be removed from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data acquired in step S11. Thereby, the therapeutic effect of cancer immunotherapy can be accurately evaluated.
 また、ステップS12では、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値に基づいて、評価対象に対する治療の効果を評価してもよい。これにより、癌免疫療法の治療効果の評価に有用なアミノ酸の濃度を利用して、当該治療効果を精度よく評価することができる。 In step S12, the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, and Pro, the effect of treatment on the evaluation target may be evaluated based on at least one concentration value. Thereby, the therapeutic effect can be accurately evaluated using the amino acid concentration useful for evaluating the therapeutic effect of cancer immunotherapy.
 また、ステップS12では、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値に基づいて、評価対象に対して治療が有効であるかを評価してもよい。例えば、治療が有効であるか否かを判別したり、また、治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりしてもよい。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別、分類など)に有用なアミノ酸の濃度を利用して、当該評価(例えば判別、分類など)を精度よく行うことができる。 In step S12, the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, and Pro, whether or not the treatment is effective may be evaluated based on the concentration value. For example, it is determined whether or not the treatment is effective, and the evaluation target is assigned to any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It may be classified. Thereby, the said evaluation (for example, discrimination, classification, etc.) can be accurately performed using the amino acid concentration useful for evaluation (for example, discrimination, classification, etc.) regarding the effectiveness of the therapeutic effect of cancer immunotherapy.
 また、濃度値の取り得る範囲が所定範囲(例えば0.0から1.0までの範囲、0.0から10.0までの範囲、0.0から100.0までの範囲、又は-10.0から10.0までの範囲、など)に収まるように、例えば、濃度値に対して任意の値を加減乗除したり、また、濃度値を所定の変換手法(例えば、指数変換、対数変換、角変換、平方根変換、プロビット変換、又は逆数変換など)で変換したり、また、濃度値に対してこれらの計算を組み合わせて行ったりすることで、濃度値を変換してもよい。例えば、濃度値を指数としネイピア数を底とする指数関数の値(具体的には、治療が有効である確率pを定義したときの自然対数ln(p/(1-p))が濃度値と等しいとした場合におけるp/(1-p)の値)をさらに算出してもよく、また、算出した指数関数の値を1と当該値との和で割った値(具体的には、確率pの値)をさらに算出してもよい。
 また、特定の条件のときの変換後の値が特定の値となるように、濃度値を変換してもよい。例えば、感度が80%のときの変換後の値が5.0となり且つ感度が95%のときの変換後の値が8.0となるように濃度値を変換してもよい。
Further, the range that the density value can take is a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or −10. For example, an arbitrary value is added / subtracted / multiplied / divided with respect to the density value so as to fall within a range from 0 to 10.0, or the density value is converted into a predetermined conversion method (for example, exponential conversion, logarithmic conversion, The density value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, reciprocal conversion, or the like, or by combining these calculations with respect to the density value. For example, the value of the exponential function with the concentration value as the index and the Napier number as the base (specifically, the natural logarithm ln (p / (1-p)) when defining the probability p that the treatment is effective is the concentration value. (The value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value (specifically, The value of the probability p) may be further calculated.
Further, the density value may be converted so that the value after conversion under a specific condition becomes a specific value. For example, the density value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
 また、ステップS12では、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む予め設定された多変量判別式に基づいて、当該多変量判別式の値であって評価対象に対する治療の効果に関する評価結果に相当する判別値を算出してもよく、さらに、算出した判別値に基づいて、評価対象に対する治療の効果を評価してもよい。例えば、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データに基づいて、治療が開始される前のアミノ酸の濃度値と治療が開始された後のアミノ酸の濃度値との比または差分を算出し、算出した各アミノ酸の濃度値の比または差分を多変量判別式に含まれる各変数に代入することで、判別値を算出してもよい。これにより、アミノ酸の濃度を変数として含む多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、癌免疫療法の治療効果を精度よく評価することができたりする。また、治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。これにより、当該治療効果を、治療終了後も継続的にモニタリングすることができる。 In step S12, based on the pre-treatment amino acid concentration data and post-treatment amino acid concentration data obtained in step S11, and a preset multivariate discriminant including the amino acid concentration as a variable, the multivariate discriminant And the discriminant value corresponding to the evaluation result relating to the effect of the treatment on the evaluation target may be calculated, and the effect of the treatment on the evaluation target may be evaluated based on the calculated discriminant value. For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated. The discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant. As a result, a multivariate discriminant including the amino acid concentration as a variable can be used to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target, or the obtained discriminant value can be used to obtain cancer. It is possible to accurately evaluate the therapeutic effect of immunotherapy. Further, the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment). Thereby, the said therapeutic effect can be continuously monitored even after completion | finish of a treatment.
 なお、多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つでもよい。これにより、癌免疫療法の治療効果の評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該治療効果を精度よく評価することができたりする。 Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used. This makes it possible to obtain a discriminant value corresponding to the evaluation result related to the therapeutic effect on the evaluation target using the multivariate discriminant useful for evaluating the therapeutic effect of cancer immunotherapy, or to use the obtained discriminant value. Thus, the therapeutic effect can be accurately evaluated.
 また、ステップS12では、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、判別値を算出してもよく、さらに、算出した判別値に基づいて、評価対象に対する治療の効果を評価してもよい。これにより、癌免疫療法の治療効果の評価に有用なアミノ酸の濃度を変数として含む当該評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該治療効果を精度よく評価することができたりする。 In step S12, the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met , Lys, Arg, Gly, Cys2, and Pro, the discriminant value may be calculated based on a multivariate discriminant including at least one as a variable, and the treatment for the evaluation target is further performed based on the calculated discriminant value. You may evaluate the effect. As a result, a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. Or the obtained discrimination value can be used to accurately evaluate the therapeutic effect.
 また、ステップS12では、ステップS11で取得した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、判別値を算出してもよく、さらに、算出した判別値に基づいて、評価対象に対して治療が有効であるかを評価してもよい。例えば、評価対象に対して治療が有効であるか否かを判別したり、また、治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりしてもよい。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別、分類など)に有用なアミノ酸の濃度を変数として含む当該評価(例えば判別、分類など)に有用な多変量判別式を利用して、評価対象に対する治療の有効性に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該評価(例えば判別、分類など)を精度よく行うことができたりする。なお、当該評価(例えば判別、分類など)で用いられる多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを変数として含む分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを変数として含むロジスティック回帰式でもよい。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別、分類など)に特に有用な多変量判別式を利用して、評価対象に対する治療の有効性に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該評価(例えば判別、分類など)をさらに精度よく行うことができたりする。 In step S12, the pre-treatment amino acid concentration data acquired in step S11 and the post-treatment amino acid concentration data include Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser. , Thr, Met, Lys, Arg, Gly, Cys2, Pro, as well as Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met , Lys, Arg, Gly, Cys2, and Pro, the discriminant value may be calculated based on a multivariate discriminant including at least one as a variable, and further, based on the calculated discriminant value, To assess whether the treatment is effective. For example, it is determined whether or not the treatment is effective for the evaluation target, and any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It is also possible to classify the evaluation target. This makes it possible to use a multivariate discriminant useful for the evaluation (for example, discrimination, classification, etc.) including the concentration of amino acids useful for evaluation (for example, discrimination, classification, etc.) as a variable regarding the effectiveness of the therapeutic effect of cancer immunotherapy. Thus, it is possible to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target, or to accurately perform the evaluation (for example, discrimination, classification, etc.) using the obtained discriminant value. Or The multivariate discriminant used in the evaluation (for example, discrimination, classification, etc.) is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable. Or a logistic regression equation including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable. This makes it possible to use a multivariate discriminant that is particularly useful for evaluating the effectiveness of the therapeutic effect of cancer immunotherapy (for example, discrimination, classification, etc.), and to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target. The evaluation (for example, discrimination, classification, etc.) can be performed with higher accuracy by using the obtained discrimination value.
 また、判別値の取り得る範囲が所定範囲(例えば0.0から1.0までの範囲、0.0から10.0までの範囲、0.0から100.0までの範囲、又は-10.0から10.0までの範囲、など)に収まるように、例えば、判別値に対して任意の値を加減乗除したり、また、判別値を所定の変換手法(例えば、指数変換、対数変換、角変換、平方根変換、プロビット変換、又は逆数変換など)で変換したり、また、判別値に対してこれらの計算を組み合わせて行ったりすることで、判別値を変換してもよい。例えば、判別値を指数としネイピア数を底とする指数関数の値(具体的には、治療が有効である確率pを定義したときの自然対数ln(p/(1-p))が判別値と等しいとした場合におけるp/(1-p)の値)をさらに算出してもよく、また、算出した指数関数の値を1と当該値との和で割った値(具体的には、確率pの値)をさらに算出してもよい。
 また、特定の条件のときの変換後の値が特定の値となるように、判別値を変換してもよい。例えば、感度が80%のときの変換後の値が5.0となり且つ感度が95%のときの変換後の値が8.0となるように判別値を変換してもよい。
 なお、本明細書における判別値は、多変量判別式の値そのものであってもよく、多変量判別式の値を変換した後の値であってもよい。
In addition, the discriminant value can have a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or −10. For example, an arbitrary value is added / subtracted / multiplied / divided with respect to the discriminant value so that the discriminant value falls within a range from 0 to 10.0, etc. The discriminant value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, or reciprocal conversion, or by combining these calculations with respect to the discriminant value. For example, the value of the exponential function with the discriminant value as the exponent and the Napier number as the base (specifically, the natural logarithm ln (p / (1-p)) when defining the probability p that the treatment is effective is the discriminant value. (The value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value (specifically, The value of the probability p) may be further calculated.
Also, the discriminant value may be converted so that the value after conversion under a specific condition becomes a specific value. For example, the discriminant value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
Note that the discriminant value in this specification may be the value of the multivariate discriminant itself, or may be a value after converting the value of the multivariate discriminant.
 また、アミノ酸濃度データに基づいて、評価対象が受ける治療法の選択を治療前に行ってもよい。換言すると、アミノ酸濃度データに基づいて、評価対象が受ける治療法を治療前に予測してもよい。 Also, based on the amino acid concentration data, the treatment method to be evaluated can be selected before treatment. In other words, based on the amino acid concentration data, a treatment method to be received by the evaluation object may be predicted before treatment.
 ここで、上記した各多変量判別式は、例えば、本出願人による国際出願である国際公開第2004/052191号に記載の方法または本出願人による国際出願である国際公開第2006/098192号に記載の方法で作成してもよい。なお、これら方法で得られた多変量判別式であれば、入力データとしてのアミノ酸濃度データにおけるアミノ酸濃度の単位に因らず、当該多変量判別式を癌免疫療法の治療効果の評価に好適に用いることができる。 Here, each multivariate discriminant described above is described in, for example, the method described in International Publication No. 2004/052191 which is an international application by the present applicant or International Publication No. 2006/098192 which is an international application by the present applicant. You may create by the method of description. If the multivariate discriminant obtained by these methods is used, the multivariate discriminant is suitable for evaluating the therapeutic effect of cancer immunotherapy regardless of the unit of amino acid concentration in the amino acid concentration data as input data. Can be used.
 また、多変量判別式とは、一般に多変量解析で用いられる式の形式を意味し、例えば分数式、重回帰式、多重ロジスティック回帰式、線形判別式、マハラノビス距離、正準判別関数、サポートベクターマシン、決定木などを包含する。また、異なる形式の多変量判別式の和で示されるような式も含まれる。また、重回帰式、多重ロジスティック回帰式、正準判別関数などにおいては各変数に係数および定数項が付加されるが、この場合の係数および定数項は、好ましくは実数であること、より好ましくはデータから判別を行うために得られた係数および定数項の99%信頼区間の範囲に属する値、さらに好ましくはデータから判別を行うために得られた係数および定数項の95%信頼区間の範囲に属する値であればかまわない。また、各係数の値、及びその信頼区間は、それを実数倍したものでもよく、定数項の値、及びその信頼区間は、それに任意の実定数を加減乗除したものでもよい。ロジスティック回帰式、線形判別式、重回帰式などを多変量判別式として用いる場合、線形変換(定数の加算、定数倍)や単調増加(減少)の変換(例えばlogit変換など)は評価性能を変えるものではなく変換前と同等であるので、これらの変換が行われた後のものを用いてもよい。 The multivariate discriminant means a formula format generally used in multivariate analysis. For example, a fractional equation, multiple regression equation, multiple logistic regression equation, linear discriminant, Mahalanobis distance, canonical discriminant function, support vector Includes machines, decision trees, etc. Also included are expressions as indicated by the sum of different forms of multivariate discriminants. In addition, in multiple regression equations, multiple logistic regression equations, canonical discriminant functions, etc., coefficients and constant terms are added to each variable. In this case, the coefficients and constant terms are preferably real numbers, more preferably Values belonging to the 99% confidence interval range of the coefficient and constant term obtained for discrimination from the data, more preferably within the 95% confidence interval range of the coefficient and constant term obtained from the data It doesn't matter if it belongs. Further, the value of each coefficient and its confidence interval may be obtained by multiplying it by a real number, and the value of the constant term and its confidence interval may be obtained by adding / subtracting / multiplying / dividing an arbitrary real constant thereto. When logistic regression, linear discriminant, multiple regression, etc. are used as multivariate discriminants, linear transformation (addition of constants, constant multiplication) or monotonic increase (decrease) conversion (eg logit transformation) changes the evaluation performance. Since these are equivalent to those before conversion, those after these conversions may be used.
 なお、分数式とは、当該分数式の分子がアミノ酸A,B,C,・・・の和で表わされ及び/又は当該分数式の分母がアミノ酸a,b,c,・・・の和で表わされるものである。また、分数式には、このような構成の分数式α,β,γ,・・・の和(例えばα+βのようなもの)も含まれる。また、分数式には、分割された分数式も含まれる。なお、分子や分母に用いられるアミノ酸にはそれぞれ適当な係数がついてもかまわない。また、分子や分母に用いられるアミノ酸は重複してもかまわない。また、各分数式に適当な係数がついてもかまわない。また、各変数の係数の値や定数項の値は、実数であればかまわない。分数式で、分子の変数と分母の変数を入れ替えた組み合わせは、目的変数との相関の正負の符号は概して逆転するが、それらの相関性は保たれるので、判別性では同等と見なせるので、分子の変数と分母の変数を入れ替えた組み合わせも、包含するものである。 The fractional expression means that the numerator of the fractional expression is represented by the sum of amino acids A, B, C,... And / or the denominator of the fractional expression is the sum of amino acids a, b, c,. It is represented by In addition, the fractional expression includes a sum of fractional expressions α, β, γ,. The fractional expression also includes a divided fractional expression. An appropriate coefficient may be added to each amino acid used in the numerator and denominator. In addition, amino acids used in the numerator and denominator may overlap. Moreover, an appropriate coefficient may be attached to each fractional expression. The value of the coefficient of each variable and the value of the constant term may be real numbers. In the fractional expression, the combination of the numerator variable and the denominator variable is generally reversed in the sign of the correlation with the target variable, but since the correlation is maintained, it can be considered equivalent in discriminability. Combinations of swapping numerator and denominator variables are also included.
 そして、第1実施形態では、癌免疫療法の治療効果を評価する際、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。また、第1実施形態では、癌免疫療法の治療効果を評価する際、多変量判別式における変数として、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。 In the first embodiment, when evaluating the therapeutic effect of cancer immunotherapy, in addition to the concentration of amino acids, other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, immunocompetent intracellular cytokine, A delayed excessive reaction (DTH) or the like may be further used. In the first embodiment, when evaluating the therapeutic effect of cancer immunotherapy, as a variable in the multivariate discriminant, in addition to the amino acid concentration, other biological information (eg, tumor marker, blood cytokine, immunocompetent cell) Number, immunocompetent intracellular cytokines, delayed hyperfractionation (DTH), etc.) may be further used.
[1-2.第1実施形態の具体例]
 ここでは、第1実施形態の具体例について図2を参照して説明する。図2は、第1実施形態にかかる癌免疫療法の評価方法の一例を示すフローチャートである。
[1-2. Specific Example of First Embodiment]
Here, a specific example of the first embodiment will be described with reference to FIG. FIG. 2 is a flowchart showing an example of the cancer immunotherapy evaluation method according to the first embodiment.
 まず、癌免疫療法による治療を受ける個体(例えば動物やヒトなど)の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データを取得する(ステップSA11)。なお、ステップSA11では、例えば、アミノ酸濃度測定を行う企業等が測定したアミノ酸濃度データを取得してもよく、また、個体から採取した血液から例えば上述した(A)または(B)などの測定方法でアミノ酸濃度データを測定することでアミノ酸濃度データを取得してもよい。 First, amino acid concentration data before starting treatment and amino acid concentration data after starting treatment of an individual (for example, an animal or a human) who are treated by cancer immunotherapy are acquired (step SA11). In step SA11, for example, amino acid concentration data measured by a company or the like that measures amino acid concentration may be acquired, and a measuring method such as (A) or (B) described above from blood collected from an individual. The amino acid concentration data may be obtained by measuring the amino acid concentration data.
 つぎに、ステップSA11で取得した個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データから、欠損値や外れ値などのデータを除去する(ステップSA12)。 Next, data such as missing values and outliers are removed from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data obtained in step SA11 (step SA12).
 つぎに、ステップSA12で欠損値や外れ値などのデータが除去された個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値に基づいて、個体に対して癌免疫療法による治療が有効であるか否かを判別する(ステップSA13)。 Next, Val, Ile, Leu, His, Phe, Trp, Gln, Val, Ile, Leu, His, Phe, Trp, Gln included in the pre-treatment amino acid concentration data and post-treatment amino acid concentration data of the individual from which data such as missing values and outliers have been removed in step SA12. Whether treatment by cancer immunotherapy is effective for an individual based on the concentration value of at least one of Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro Is determined (step SA13).
 具体的には、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値と予め設定された閾値(カットオフ値)とを比較することで、個体に対して治療が有効であるか否かを判別する。 Specifically, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys included in the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment. , Arg, Gly, Cys2, and Pro, and a threshold value (cutoff value) set in advance is compared to determine whether or not the treatment is effective for the individual.
 また、具体的には、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、1つまたは複数の判別値(例えば、治療開始前アミノ酸濃度データを多変量判別式に代入して得られる判別値(治療開始前判別値)、治療開始後アミノ酸濃度データを多変量判別式に代入して得られる判別値(治療開始後判別値)、または、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データとの差分または比などに関する変化量データを多変量判別式に代入して得られる判別値(治療開始前後判別値)、など)を算出し、算出した判別値と予め設定された閾値とを比較することで、個体に対して治療が有効であるか否かを判別する。 Specifically, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. , Lys, Arg, Gly, Cys2, Pro, and at least one concentration value, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg , Gly, Cys2, Pro, based on a multivariate discriminant including at least one as a variable, obtained by substituting one or more discriminant values (for example, amino acid concentration data before starting treatment into the multivariate discriminant) Substituting the discriminant value (discriminant value before starting treatment) and amino acid concentration data after starting treatment into the multivariate discriminant Discriminant value (discriminant value after the start of treatment) or a discriminant value obtained by substituting change data relating to the difference or ratio between the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment into the multivariate discriminant ( And the like, and a comparison is made between the calculated discriminant value and a preset threshold value to discriminate whether or not the treatment is effective for the individual.
[第2実施形態]
[2-1.第2実施形態の概要]
 ここでは、第2実施形態の概要について図3を参照して説明する。図3は第2実施形態の基本原理を示す原理構成図である。
[Second Embodiment]
[2-1. Outline of Second Embodiment]
Here, an outline of the second embodiment will be described with reference to FIG. FIG. 3 is a principle configuration diagram showing the basic principle of the second embodiment.
 まず、制御部は、癌免疫療法による治療を受ける評価対象(例えば動物やヒトなどの個体)の治療開始前のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、当該評価対象の治療開始後のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって評価対象に対する治療の効果に関する評価結果に相当する判別値を算出する(ステップS21)。 First, the control unit determines the amino acid concentration data before the start of treatment of the evaluation target (for example, an individual such as an animal or a human) to be treated by cancer immunotherapy, the amino acid concentration data before the start of treatment, and the amino acid after the start of the treatment of the evaluation target. Based on the amino acid concentration data after the start of treatment related to the concentration value of the substance and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the value of the multivariate discriminant and the effect of the treatment on the evaluation target A discriminant value corresponding to the evaluation result is calculated (step S21).
 つぎに、制御部は、ステップS21で算出した判別値に基づいて、評価対象に対する癌免疫療法による治療の効果を評価する(ステップS22)。 Next, the control unit evaluates the effect of treatment by cancer immunotherapy on the evaluation target based on the discriminant value calculated in step S21 (step S22).
 以上、第2実施形態によれば、評価対象の治療開始前アミノ酸濃度データ、治療開始後アミノ酸濃度データ、および多変量判別式に基づいて、評価対象に対する治療の効果に関する評価結果に相当する判別値を算出し、算出した判別値に基づいて、評価対象に対する治療の効果を評価する(要するに、評価対象に対する治療の効果を評価するための情報を提供する)。例えば、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データに基づいて、治療が開始される前のアミノ酸の濃度値と治療が開始された後のアミノ酸の濃度値との比または差分を算出し、算出した各アミノ酸の濃度値の比または差分を多変量判別式に含まれる各変数に代入することで、判別値を算出してもよい。これにより、アミノ酸の濃度を変数として含む多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、癌免疫療法の治療効果を精度よく評価することができたりする。また、治療開始後アミノ酸濃度データは、上述した「治療後」のアミノ酸濃度データに対応するもの(治療後アミノ酸濃度データ)であってもよい。これにより、当該治療効果を、治療終了後も継続的にモニタリングすることができる。 As described above, according to the second embodiment, based on the pre-treatment amino acid concentration data, the post-treatment amino acid concentration data of the evaluation target, and the multivariate discriminant, the discriminant value corresponding to the evaluation result regarding the effect of the treatment on the evaluation target. And the effect of the treatment on the evaluation object is evaluated based on the calculated discriminant value (in short, information for evaluating the effect of the treatment on the evaluation object is provided). For example, based on the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment, the ratio or difference between the concentration value of the amino acid before starting treatment and the concentration value of the amino acid after starting treatment is calculated. The discriminant value may be calculated by substituting the ratio or difference of the calculated concentration values of each amino acid into each variable included in the multivariate discriminant. As a result, a multivariate discriminant including the amino acid concentration as a variable can be used to obtain a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target, or the obtained discriminant value can be used to obtain cancer. It is possible to accurately evaluate the therapeutic effect of immunotherapy. Further, the amino acid concentration data after the start of treatment may correspond to the above-mentioned “after treatment” amino acid concentration data (amino acid concentration data after treatment). Thereby, the said therapeutic effect can be continuously monitored even after completion | finish of a treatment.
 なお、多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つでもよい。これにより、癌免疫療法の治療効果の評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該治療効果を精度よく評価することができたりする。 Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used. This makes it possible to obtain a discriminant value corresponding to the evaluation result related to the therapeutic effect on the evaluation target using the multivariate discriminant useful for evaluating the therapeutic effect of cancer immunotherapy, or to use the obtained discriminant value. Thus, the therapeutic effect can be accurately evaluated.
 また、ステップS21では、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、判別値を算出し、ステップS22では、ステップS21で算出した判別値に基づいて、評価対象に対する治療の効果を評価してもよい。これにより、癌免疫療法の治療効果の評価に有用なアミノ酸の濃度を変数として含む当該評価に有用な多変量判別式を利用して、評価対象に対する治療効果に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該治療効果を精度よく評価することができたりする。 In step S21, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, and the amino acid concentration data before and after the treatment start are included. At least one concentration value of Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, A discriminant value is calculated based on a multivariate discriminant including at least one of Gly, Cys2, and Pro as a variable. In step S22, the effect of treatment on the evaluation target is calculated based on the discriminant value calculated in step S21. You may evaluate. As a result, a discriminant value corresponding to the evaluation result regarding the therapeutic effect on the evaluation target is obtained using a multivariate discriminant useful for the evaluation including the concentration of amino acid useful for evaluating the therapeutic effect of cancer immunotherapy as a variable. Or the obtained discrimination value can be used to accurately evaluate the therapeutic effect.
 また、ステップS21では、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、判別値を算出し、ステップS22では、ステップS21で算出した判別値に基づいて、評価対象に対して治療が有効であるかを評価してもよい。例えば、評価対象に対して治療が有効であるか否かを判別したり、また、治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりしてもよい。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別、分類など)に有用なアミノ酸の濃度を変数として含む当該評価(例えば判別、分類など)に有用な多変量判別式を利用して、評価対象に対する治療の有効性に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該評価(例えば判別、分類など)を精度よく行うことができたりする。なお、当該評価(例えば判別、分類など)で用いられる多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを変数として含む分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを変数として含むロジスティック回帰式でもよい。これにより、癌免疫療法の治療効果の有効性に関する評価(例えば判別、分類など)に特に有用な多変量判別式を利用して、評価対象に対する治療の有効性に関する評価結果に相当する判別値を得ることができたり、得られた判別値を利用して、当該評価(例えば判別、分類など)をさらに精度よく行うことができたりする。 In step S21, Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, and the amino acid concentration data before and after the treatment start are included. At least one concentration value of Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, A discriminant value is calculated based on a multivariate discriminant including at least one of Gly, Cys2, and Pro as a variable. In step S22, treatment is performed on the evaluation target based on the discriminant value calculated in step S21. You may evaluate whether it is effective. For example, it is determined whether or not the treatment is effective for the evaluation target, and any one of a plurality of categories (ranks) defined in consideration of the possibility of the treatment being effective. It is also possible to classify the evaluation target. This makes it possible to use a multivariate discriminant useful for the evaluation (for example, discrimination, classification, etc.) including the concentration of amino acids useful for evaluation (for example, discrimination, classification, etc.) as a variable regarding the effectiveness of the therapeutic effect of cancer immunotherapy. Thus, it is possible to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target, or to accurately perform the evaluation (for example, discrimination, classification, etc.) using the obtained discriminant value. Or The multivariate discriminant used in the evaluation (for example, discrimination, classification, etc.) is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable. Or a logistic regression equation including at least one of Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable. This makes it possible to use a multivariate discriminant that is particularly useful for evaluating the effectiveness of the therapeutic effect of cancer immunotherapy (for example, discrimination, classification, etc.), and to obtain a discriminant value corresponding to the evaluation result regarding the effectiveness of the treatment for the evaluation target. The evaluation (for example, discrimination, classification, etc.) can be performed with higher accuracy by using the obtained discrimination value.
 また、判別値の取り得る範囲が所定範囲(例えば0.0から1.0までの範囲、0.0から10.0までの範囲、0.0から100.0までの範囲、又は-10.0から10.0までの範囲、など)に収まるように、例えば、判別値に対して任意の値を加減乗除したり、また、判別値を所定の変換手法(例えば、指数変換、対数変換、角変換、平方根変換、プロビット変換、又は逆数変換など)で変換したり、また、判別値に対してこれらの計算を組み合わせて行ったりすることで、判別値を変換してもよい。例えば、判別値を指数としネイピア数を底とする指数関数の値(具体的には、治療が有効である確率pを定義したときの自然対数ln(p/(1-p))が判別値と等しいとした場合におけるp/(1-p)の値)をさらに算出してもよく、また、算出した指数関数の値を1と当該値との和で割った値(具体的には、確率pの値)をさらに算出してもよい。
 また、特定の条件のときの変換後の値が特定の値となるように、判別値を変換してもよい。例えば、感度が80%のときの変換後の値が5.0となり且つ感度が95%のときの変換後の値が8.0となるように判別値を変換してもよい。
 なお、本明細書における判別値は、多変量判別式の値そのものであってもよく、多変量判別式の値を変換した後の値であってもよい。
In addition, the discriminant value can have a predetermined range (for example, a range from 0.0 to 1.0, a range from 0.0 to 10.0, a range from 0.0 to 100.0, or −10. For example, an arbitrary value is added / subtracted / multiplied / divided with respect to the discriminant value so that the discriminant value falls within a range from 0 to 10.0, etc. The discriminant value may be converted by performing conversion by angle conversion, square root conversion, probit conversion, or reciprocal conversion, or by combining these calculations with respect to the discriminant value. For example, the value of the exponential function with the discriminant value as the exponent and the Napier number as the base (specifically, the natural logarithm ln (p / (1-p)) when defining the probability p that the treatment is effective is the discriminant value. (The value of p / (1-p) in the case where it is equal to), or a value obtained by dividing the calculated exponential function value by the sum of 1 and the value (specifically, The value of the probability p) may be further calculated.
Also, the discriminant value may be converted so that the value after conversion under a specific condition becomes a specific value. For example, the discriminant value may be converted so that the value after conversion when the sensitivity is 80% is 5.0 and the value after conversion when the sensitivity is 95% is 8.0.
Note that the discriminant value in this specification may be the value of the multivariate discriminant itself, or may be a value after converting the value of the multivariate discriminant.
 また、アミノ酸濃度データに基づいて、評価対象が受ける治療法の選択を治療前に行ってもよい。換言すると、アミノ酸濃度データに基づいて、評価対象が受ける治療法を治療前に予測してもよい。 Also, based on the amino acid concentration data, the treatment method to be evaluated can be selected before treatment. In other words, based on the amino acid concentration data, a treatment method to be received by the evaluation object may be predicted before treatment.
 ここで、上記した各多変量判別式は、例えば、本出願人による国際出願である国際公開第2004/052191号に記載の方法または本出願人による国際出願である国際公開第2006/098192号に記載の方法で作成してもよい。なお、これら方法で得られた多変量判別式であれば、入力データとしてのアミノ酸濃度データにおけるアミノ酸濃度の単位に因らず、当該多変量判別式を癌免疫療法の治療効果の評価に好適に用いることができる。 Here, each multivariate discriminant described above is described in, for example, the method described in International Publication No. 2004/052191 which is an international application by the present applicant or International Publication No. 2006/098192 which is an international application by the present applicant. You may create by the method of description. If the multivariate discriminant obtained by these methods is used, the multivariate discriminant is suitable for evaluating the therapeutic effect of cancer immunotherapy regardless of the unit of amino acid concentration in the amino acid concentration data as input data. Can be used.
 また、多変量判別式とは、一般に多変量解析で用いられる式の形式を意味し、例えば分数式、重回帰式、多重ロジスティック回帰式、線形判別式、マハラノビス距離、正準判別関数、サポートベクターマシン、決定木などを包含する。また、異なる形式の多変量判別式の和で示されるような式も含まれる。また、重回帰式、多重ロジスティック回帰式、正準判別関数などにおいては各変数に係数および定数項が付加されるが、この場合の係数および定数項は、好ましくは実数であること、より好ましくはデータから判別を行うために得られた係数および定数項の99%信頼区間の範囲に属する値、さらに好ましくはデータから判別を行うために得られた係数および定数項の95%信頼区間の範囲に属する値であればかまわない。また、各係数の値、及びその信頼区間は、それを実数倍したものでもよく、定数項の値、及びその信頼区間は、それに任意の実定数を加減乗除したものでもよい。ロジスティック回帰式、線形判別式、重回帰式などを多変量判別式として用いる場合、線形変換(定数の加算、定数倍)や単調増加(減少)の変換(例えばlogit変換など)は評価性能を変えるものではなく変換前と同等であるので、これらの変換が行われた後のものを用いてもよい。 The multivariate discriminant means a formula format generally used in multivariate analysis. For example, a fractional equation, multiple regression equation, multiple logistic regression equation, linear discriminant, Mahalanobis distance, canonical discriminant function, support vector Includes machines, decision trees, etc. Also included are expressions as indicated by the sum of different forms of multivariate discriminants. In addition, in multiple regression equations, multiple logistic regression equations, canonical discriminant functions, etc., coefficients and constant terms are added to each variable. In this case, the coefficients and constant terms are preferably real numbers, more preferably Values belonging to the 99% confidence interval range of the coefficient and constant term obtained for discrimination from the data, more preferably within the 95% confidence interval range of the coefficient and constant term obtained from the data It doesn't matter if it belongs. Further, the value of each coefficient and its confidence interval may be obtained by multiplying it by a real number, and the value of the constant term and its confidence interval may be obtained by adding / subtracting / multiplying / dividing an arbitrary real constant thereto. When logistic regression, linear discriminant, multiple regression, etc. are used as multivariate discriminants, linear transformation (addition of constants, constant multiplication) or monotonic increase (decrease) conversion (eg logit transformation) changes the evaluation performance. Since these are equivalent to those before conversion, those after these conversions may be used.
 なお、分数式とは、当該分数式の分子がアミノ酸A,B,C,・・・の和で表わされ及び/又は当該分数式の分母がアミノ酸a,b,c,・・・の和で表わされるものである。また、分数式には、このような構成の分数式α,β,γ,・・・の和(例えばα+βのようなもの)も含まれる。また、分数式には、分割された分数式も含まれる。なお、分子や分母に用いられるアミノ酸にはそれぞれ適当な係数がついてもかまわない。また、分子や分母に用いられるアミノ酸は重複してもかまわない。また、各分数式に適当な係数がついてもかまわない。また、各変数の係数の値や定数項の値は、実数であればかまわない。分数式で、分子の変数と分母の変数を入れ替えた組み合わせは、目的変数との相関の正負の符号は概して逆転するが、それらの相関性は保たれるので、判別性では同等と見なせるので、分子の変数と分母の変数を入れ替えた組み合わせも、包含するものである。 The fractional expression means that the numerator of the fractional expression is represented by the sum of amino acids A, B, C,... And / or the denominator of the fractional expression is the sum of amino acids a, b, c,. It is represented by In addition, the fractional expression includes a sum of fractional expressions α, β, γ,. The fractional expression also includes a divided fractional expression. An appropriate coefficient may be added to each amino acid used in the numerator and denominator. In addition, amino acids used in the numerator and denominator may overlap. Moreover, an appropriate coefficient may be attached to each fractional expression. The value of the coefficient of each variable and the value of the constant term may be real numbers. In the fractional expression, the combination of the numerator variable and the denominator variable is generally reversed in the sign of the correlation with the target variable, but since the correlation is maintained, it can be considered equivalent in discriminability. Combinations of swapping numerator and denominator variables are also included.
 そして、第2実施形態では、癌免疫療法の治療効果を評価する際、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。また、第2実施形態では、癌免疫療法の治療効果を評価する際、多変量判別式における変数として、アミノ酸の濃度以外に、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)をさらに用いてもかまわない。 In the second embodiment, when evaluating the therapeutic effect of cancer immunotherapy, in addition to the concentration of amino acids, other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, immunocompetent intracellular cytokine, A delayed excessive reaction (DTH) or the like may be further used. In the second embodiment, when evaluating the therapeutic effect of cancer immunotherapy, as a variable in the multivariate discriminant, in addition to the amino acid concentration, other biological information (eg, tumor marker, blood cytokine, immunocompetent cell) Number, immunocompetent intracellular cytokines, delayed hyperfractionation (DTH), etc.) may be further used.
 ここで、多変量判別式作成処理(工程1~工程4)の概要について詳細に説明する。なお、ここで説明する処理はあくまでも一例であり、多変量判別式の作成方法はこれに限定されない。 Here, the outline of the multivariate discriminant creation process (step 1 to step 4) will be described in detail. Note that the processing described here is merely an example, and the method of creating the multivariate discriminant is not limited to this.
 まず、制御部は、アミノ酸濃度データ(例えば、アミノ酸濃度に関するデータ、または、アミノ酸濃度の変化量に関するデータ、など)と癌の状態を表す指標(例えば、腫瘍の大きさなど)に関する癌状態指標データ(例えば、癌の状態を表す指標に関するデータ(例えば、指標に基づいて得られる治療効果の有無など)、または、癌の状態の指標の変化量に関するデータ(例えば、腫瘍の大きさの差分値など)、など)とを含む記憶部に記憶された癌状態情報から所定の式作成手法に基づいて、多変量判別式の候補である候補多変量判別式(例えば、y=a+a+・・・+a、y:癌状態指標データ、x:アミノ酸濃度データ、a:定数、i=1,2,・・・,n)を作成する(工程1)。なお、事前に、癌状態情報から欠損値や外れ値などを持つデータを除去してもよい。 First, the control unit determines amino acid concentration data (for example, data relating to amino acid concentration or data relating to the amount of change in amino acid concentration) and cancer state index data relating to an index (for example, tumor size) indicating a cancer state. (For example, data related to an index representing a cancer state (for example, presence or absence of a therapeutic effect obtained based on the index), or data related to a change amount of an index of a cancer state (for example, a difference value of a tumor size, etc.) ), Etc.) based on cancer state information stored in the storage unit, a candidate multivariate discriminant that is a candidate for a multivariate discriminant (for example, y = a 1 x 1 + a 2) x 2 +... + a n x n , y: cancer state index data, x i : amino acid concentration data, a i : constant, i = 1, 2,..., n) are created (step 1). Note that data having missing values, outliers, and the like may be removed from the cancer state information in advance.
 なお、工程1において、癌状態情報から、複数の異なる式作成手法(主成分分析や判別分析、サポートベクターマシン、重回帰分析、ロジスティック回帰分析、k-means法、クラスター解析、決定木などの多変量解析に関するものを含む。)を併用して複数の候補多変量判別式を作成してもよい。具体的には、多数の治療開始前群および多数の治療開始後群から得た血液を分析して得たアミノ酸濃度データおよび癌状態指標データから構成される多変量データである癌状態情報に対して、複数の異なるアルゴリズムを利用して複数群の候補多変量判別式を同時並行的に作成してもよい。例えば、異なるアルゴリズムを利用して判別分析およびロジスティック回帰分析を同時に行い、2つの異なる候補多変量判別式を作成してもよい。また、主成分分析を行って作成した候補多変量判別式を利用して癌状態情報を変換し、変換した癌状態情報に対して判別分析を行うことで候補多変量判別式を作成してもよい。これにより、最終的に、評価条件に合った適切な多変量判別式を作成することができる。 In Step 1, a plurality of different formula creation methods (principal component analysis, discriminant analysis, support vector machine, multiple regression analysis, logistic regression analysis, k-means method, cluster analysis, decision tree, etc.) are obtained from cancer status information. A plurality of candidate multivariate discriminants may be created by using the above in combination. Specifically, for cancer status information that is multivariate data composed of amino acid concentration data and cancer status index data obtained by analyzing blood obtained from a large number of pre-treatment groups and a large number of post-treatment groups. Thus, a plurality of groups of candidate multivariate discriminants may be created concurrently using a plurality of different algorithms. For example, two different candidate multivariate discriminants may be created by performing discriminant analysis and logistic regression analysis simultaneously using different algorithms. Also, even if the candidate multivariate discriminant is created by converting the cancer state information using the candidate multivariate discriminant created by performing the principal component analysis and performing the discriminant analysis on the converted cancer state information Good. Thereby, finally, an appropriate multivariate discriminant suitable for the evaluation condition can be created.
 ここで、主成分分析を用いて作成した候補多変量判別式は、全てのアミノ酸濃度データの分散を最大にするような各アミノ酸変数を含む一次式である。また、判別分析を用いて作成した候補多変量判別式は、各群内の分散の和の全てのアミノ酸濃度データの分散に対する比を最小にするような各アミノ酸変数を含む高次式(指数や対数を含む)である。また、サポートベクターマシンを用いて作成した候補多変量判別式は、群間の境界を最大にするような各アミノ酸変数を含む高次式(カーネル関数を含む)である。また、重回帰分析を用いて作成した候補多変量判別式は、全てのアミノ酸濃度データからの距離の和を最小にするような各アミノ酸変数を含む高次式である。ロジスティック回帰分析を用いて作成した候補多変量判別式は、確率の対数オッズを表す線形モデルであり、その確率の尤度を最大にするような各アミノ酸変数を含む一次式である。また、k-means法とは、各アミノ酸濃度データのk個近傍を探索し、近傍点の属する群の中で一番多いものをそのデータの所属群と定義し、入力されたアミノ酸濃度データの属する群と定義された群とが最も合致するようなアミノ酸変数を選択する手法である。また、クラスター解析とは、全てのアミノ酸濃度データの中で最も近い距離にある点同士をクラスタリング(群化)する手法である。また、決定木とは、アミノ酸変数に序列をつけて、序列が上位であるアミノ酸変数の取りうるパターンからアミノ酸濃度データの群を予測する手法である。 Here, the candidate multivariate discriminant prepared using principal component analysis is a linear expression including each amino acid variable that maximizes the variance of all amino acid concentration data. In addition, the candidate multivariate discriminant created using discriminant analysis is a high-order formula (exponential or exponential) including each amino acid variable that minimizes the ratio of the sum of variances within each group to the variance of all amino acid concentration data. Including logarithm). In addition, the candidate multivariate discriminant created using the support vector machine is a higher-order formula (including a kernel function) including each amino acid variable that maximizes the boundary between groups. In addition, the candidate multivariate discriminant created using multiple regression analysis is a high-order expression including each amino acid variable that minimizes the sum of distances from all amino acid concentration data. A candidate multivariate discriminant created using logistic regression analysis is a linear model representing the log odds of probability, and is a linear expression including each amino acid variable that maximizes the likelihood of the probability. The k-means method searches k neighborhoods of each amino acid concentration data, defines the largest group among the groups to which the neighboring points belong as the group to which the data belongs, This is a method of selecting an amino acid variable that best matches the group to which the group belongs. Cluster analysis is a method of clustering (grouping) points that are closest to each other in all amino acid concentration data. The decision tree is a technique for predicting a group of amino acid concentration data based on patterns that can be taken by amino acid variables having higher ranks by adding ranks to amino acid variables.
 多変量判別式作成処理の説明に戻り、制御部は、工程1で作成した候補多変量判別式を、所定の検証手法に基づいて検証(相互検証)する(工程2)。候補多変量判別式の検証は、工程1で作成した各候補多変量判別式に対して行う。 Returning to the description of the multivariate discriminant creation process, the control unit verifies (mutually verifies) the candidate multivariate discriminant created in step 1 based on a predetermined verification method (step 2). The candidate multivariate discriminant is verified for each candidate multivariate discriminant created in step 1.
 なお、工程2において、ランダムサンプリング法、ブートストラップ法、ホールドアウト法、N-フォールド法またはリーブワンアウト法などのうち少なくとも1つに基づいて候補多変量判別式の判別率や感度、特異度、情報量基準、ROC_AUC(受信者特性曲線の曲線下面積)などのうち少なくとも1つに関して検証してもよい。これにより、癌状態情報や診断条件を考慮した予測性または頑健性の高い候補多変量判別式を作成することができる。 In step 2, the discrimination rate, sensitivity, specificity, etc. of the candidate multivariate discriminant based on at least one of random sampling method, bootstrap method, holdout method, N-fold method, leave one out method, etc. The verification may be performed with respect to at least one of the information criterion, ROC_AUC (area under the curve of the receiver characteristic curve), and the like. Thereby, a candidate multivariate discriminant with high predictability or robustness in consideration of cancer state information and diagnostic conditions can be created.
 ここで、判別率とは、本実施形態で評価した治療効果の結果が真の状態として陰性のものを正しく陰性と評価し、真の状態として陽性のものを正しく陽性と評価している割合である。また、感度とは、本実施形態で評価した治療効果の結果が真の状態として陽性のものを正しく陽性と評価している割合である。また、特異度とは、本実施形態で評価した治療効果の結果が真の状態として陰性のものを正しく陰性と評価している割合である。また、情報量基準とは、工程1で作成した候補多変量判別式のアミノ酸変数の数と、本実施形態で評価した治療効果の結果および入力データに記載された治療効果の結果の差異と、を足し合わせたものである。また、ROC_AUC(受信者特性曲線の曲線下面積)は、2次元座標上に(x,y)=(1-特異度,感度)をプロットして作成される曲線である受信者特性曲線(ROC)の曲線下面積として定義され、ROC_AUCの値は完全な判別では1となり、この値が1に近いほど判別性が高いことを示す。また、予測性とは、候補多変量判別式の検証を繰り返すことで得られた判別率や感度、特異性を平均したものである。また、頑健性とは、候補多変量判別式の検証を繰り返すことで得られた判別率や感度、特異性の分散である。 Here, the discrimination rate is a ratio in which the result of the therapeutic effect evaluated in the present embodiment is negative as a true state and is evaluated as negative correctly, and a positive result as a true state is correctly evaluated as positive. is there. Sensitivity is the ratio at which positive results are positively evaluated as positive as the result of the therapeutic effect evaluated in this embodiment. Further, the specificity is a ratio at which negative results are correctly evaluated as negative as the true result of the therapeutic effect evaluated in the present embodiment. The information criterion is the difference between the number of amino acid variables of the candidate multivariate discriminant created in step 1, the result of the treatment effect evaluated in this embodiment and the result of the treatment effect described in the input data, Are added together. ROC_AUC (area under the curve of the receiver characteristic curve) is a receiver characteristic curve (ROC) that is a curve created by plotting (x, y) = (1−specificity, sensitivity) on a two-dimensional coordinate. ), The value of ROC_AUC is 1 in complete discrimination, and the closer this value is to 1, the higher the discriminability. The predictability is an average of the discrimination rate, sensitivity, and specificity obtained by repeating the verification of the candidate multivariate discriminant. Robustness is the variance of discrimination rate, sensitivity, and specificity obtained by repeating verification of candidate multivariate discriminants.
 多変量判別式作成処理の説明に戻り、制御部は、所定の変数選択手法に基づいて候補多変量判別式の変数を選択することで、候補多変量判別式を作成する際に用いる癌状態情報に含まれるアミノ酸濃度データの組み合わせを選択する(工程3)。アミノ酸変数の選択は、工程1で作成した各候補多変量判別式に対して行ってもよい。これにより、候補多変量判別式のアミノ酸変数を適切に選択することができる。そして、工程3で選択したアミノ酸濃度データを含む癌状態情報を用いて再び工程1を実行する。 Returning to the description of the multivariate discriminant creation process, the control unit selects cancer candidate variable variables based on a predetermined variable selection method, and thus cancer state information used when creating a candidate multivariate discriminant A combination of amino acid concentration data contained in is selected (step 3). The selection of amino acid variables may be performed for each candidate multivariate discriminant created in step 1. Thereby, the amino acid variable of a candidate multivariate discriminant can be selected appropriately. Then, Step 1 is executed again using the cancer state information including the amino acid concentration data selected in Step 3.
 なお、工程3において、工程2での検証結果からステップワイズ法、ベストパス法、近傍探索法、遺伝的アルゴリズムのうち少なくとも1つに基づいて候補多変量判別式のアミノ酸変数を選択してもよい。 In step 3, the amino acid variable of the candidate multivariate discriminant may be selected from the verification result in step 2 based on at least one of stepwise method, best path method, neighborhood search method, and genetic algorithm. .
 ここで、ベストパス法とは、候補多変量判別式に含まれるアミノ酸変数を1つずつ順次減らしていき、候補多変量判別式が与える評価指標を最適化することでアミノ酸変数を選択する方法である。 Here, the best path method is a method of selecting amino acid variables by sequentially reducing amino acid variables included in the candidate multivariate discriminant one by one and optimizing the evaluation index given by the candidate multivariate discriminant. is there.
 多変量判別式作成処理の説明に戻り、制御部は、上述した工程1、工程2および工程3を繰り返し実行し、これにより蓄積した検証結果に基づいて、複数の候補多変量判別式の中から多変量判別式として採用する候補多変量判別式を選出することで、多変量判別式を作成する(工程4)。なお、候補多変量判別式の選出には、例えば、同じ式作成手法で作成した候補多変量判別式の中から最適なものを選出する場合と、すべての候補多変量判別式の中から最適なものを選出する場合とがある。 Returning to the description of the multivariate discriminant creation process, the control unit repeatedly executes the above-described step 1, step 2 and step 3, and based on the verification results accumulated thereby, the control unit can select from a plurality of candidate multivariate discriminants. A multivariate discriminant is created by selecting candidate multivariate discriminants to be adopted as the multivariate discriminant (step 4). In selecting candidate multivariate discriminants, for example, selecting the optimal one from among candidate multivariate discriminants created by the same formula creation method, and selecting the optimum from all candidate multivariate discriminants Sometimes there is a choice.
 以上、説明したように、多変量判別式作成処理では、癌状態情報に基づいて、候補多変量判別式の作成、候補多変量判別式の検証および候補多変量判別式の変数の選択に関する処理を一連の流れで体系化(システム化)して実行することにより、癌免疫療法の治療効果の評価に最適な多変量判別式を作成することができる。換言すると、多変量判別式作成処理では、アミノ酸濃度を多変量の統計解析に用い、最適でロバストな変数の組を選択するために変数選択法とクロスバリデーションとを組み合わせて、評価性能の高い多変量判別式を抽出する。多変量判別式としては、ロジスティック回帰、線形判別、サポートベクターマシン、マハラノビス距離法、重回帰分析、クラスター解析、決定木などを用いることができる。 As described above, in the multivariate discriminant creation process, processing related to the creation of a candidate multivariate discriminant, verification of the candidate multivariate discriminant, and selection of a variable of the candidate multivariate discriminant based on the cancer state information. By performing systematization (systematization) in a series of flows, it is possible to create a multivariate discriminant that is optimal for evaluating the therapeutic effect of cancer immunotherapy. In other words, in the multivariate discriminant creation process, the amino acid concentration is used for multivariate statistical analysis, and the variable selection method and cross-validation are combined in order to select the optimal and robust variable set. Extract the variable discriminant. As the multivariate discriminant, logistic regression, linear discrimination, support vector machine, Mahalanobis distance method, multiple regression analysis, cluster analysis, decision tree, and the like can be used.
[2-2.第2実施形態の構成]
 ここでは、第2実施形態にかかる癌免疫療法評価システム(以下では本システムと記す場合がある。)の構成について、図4から図20を参照して説明する。なお、本システムはあくまでも一例であり、本発明はこれに限定されない。
[2-2. Configuration of Second Embodiment]
Here, the configuration of a cancer immunotherapy evaluation system according to the second embodiment (hereinafter sometimes referred to as the present system) will be described with reference to FIGS. 4 to 20. This system is merely an example, and the present invention is not limited to this.
 まず、本システムの全体構成について図4および図5を参照して説明する。図4は本システムの全体構成の一例を示す図である。また、図5は本システムの全体構成の他の一例を示す図である。本システムは、図4に示すように、癌免疫療法による治療を受ける評価対象に対する治療の効果を評価する癌免疫療法評価装置100と、評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供するクライアント装置200(本発明の情報通信端末装置に相当)とを、ネットワーク300を介して通信可能に接続して構成されている。 First, the overall configuration of this system will be described with reference to FIG. 4 and FIG. FIG. 4 is a diagram showing an example of the overall configuration of the present system. FIG. 5 is a diagram showing another example of the overall configuration of the present system. As shown in FIG. 4, the present system includes a cancer immunotherapy evaluation apparatus 100 that evaluates the effect of treatment on an evaluation target that receives treatment by cancer immunotherapy, and a client that provides amino acid concentration data relating to the concentration value of the amino acid to be evaluated. The apparatus 200 (corresponding to the information communication terminal apparatus of the present invention) is configured to be communicably connected via the network 300.
 なお、本システムは、図5に示すように、癌免疫療法評価装置100やクライアント装置200の他に、癌免疫療法評価装置100で多変量判別式を作成する際に用いる癌状態情報や、癌免疫療法の治療効果の評価を行うために用いる多変量判別式などを格納したデータベース装置400を、ネットワーク300を介して通信可能に接続して構成されてもよい。これにより、ネットワーク300を介して、癌免疫療法評価装置100からクライアント装置200やデータベース装置400へ、あるいはクライアント装置200やデータベース装置400から癌免疫療法評価装置100へ、癌免疫療法による治療での癌の状態に関する情報などが提供される。ここで、癌免疫療法による治療での癌の状態に関する情報とは、癌免疫療法による治療でのヒトを含む生物の癌の状態に関する特定の項目について測定した値に関する情報(例えば、治療効果の有無、または腫瘍の大きさの差分値など)である。また、癌免疫療法による治療での癌の状態に関する情報は、癌免疫療法評価装置100やクライアント装置200や他の装置(例えば各種の計測装置等)で生成され、主にデータベース装置400に蓄積される。 In addition to the cancer immunotherapy evaluation apparatus 100 and the client apparatus 200, this system, as shown in FIG. 5, uses cancer status information used when creating a multivariate discriminant in the cancer immunotherapy evaluation apparatus 100, A database apparatus 400 storing a multivariate discriminant used for evaluating the therapeutic effect of immunotherapy may be configured to be communicably connected via the network 300. Accordingly, cancer in the treatment by cancer immunotherapy from the cancer immunotherapy evaluation apparatus 100 to the client apparatus 200 or the database apparatus 400, or from the client apparatus 200 or database apparatus 400 to the cancer immunotherapy evaluation apparatus 100 via the network 300. Information about the state of the is provided. Here, the information on the cancer state in the treatment with cancer immunotherapy is information on the value measured for a specific item related to the cancer state of an organism including humans in the treatment with cancer immunotherapy (for example, presence or absence of therapeutic effect) Or a difference value of tumor size). In addition, information related to cancer status in cancer immunotherapy treatment is generated by the cancer immunotherapy evaluation apparatus 100, the client apparatus 200, and other apparatuses (for example, various measurement apparatuses), and is mainly stored in the database apparatus 400. The
 つぎに、本システムの癌免疫療法評価装置100の構成について図6から図18を参照して説明する。図6は、本システムの癌免疫療法評価装置100の構成の一例を示すブロック図であり、該構成のうち本発明に関係する部分のみを概念的に示している。 Next, the configuration of the cancer immunotherapy evaluation apparatus 100 of this system will be described with reference to FIGS. FIG. 6 is a block diagram showing an example of the configuration of the cancer immunotherapy evaluation apparatus 100 of the present system, and conceptually shows only the portion related to the present invention in the configuration.
 癌免疫療法評価装置100は、当該癌免疫療法評価装置を統括的に制御するCPU等の制御部102と、ルータ等の通信装置および専用線等の有線または無線の通信回線を介して当該癌免疫療法評価装置をネットワーク300に通信可能に接続する通信インターフェース部104と、各種のデータベースやテーブルやファイルなどを格納する記憶部106と、入力装置112や出力装置114に接続する入出力インターフェース部108と、で構成されており、これら各部は任意の通信路を介して通信可能に接続されている。ここで、癌免疫療法評価装置100は、各種の分析装置(例えばアミノ酸アナライザー等)と同一筐体で構成されてもよい。 The cancer immunotherapy evaluation apparatus 100 includes a control unit 102 such as a CPU that comprehensively controls the cancer immunotherapy evaluation apparatus, a communication device such as a router, and a wired or wireless communication line such as a dedicated line. A communication interface unit 104 that connects the therapy evaluation device to the network 300 so as to be communicable, a storage unit 106 that stores various databases, tables, files, and the like, and an input / output interface unit 108 that connects to the input device 112 and the output device 114 These parts are connected to be communicable via an arbitrary communication path. Here, the cancer immunotherapy evaluation apparatus 100 may be configured in the same housing as various analysis apparatuses (for example, an amino acid analyzer or the like).
 記憶部106は、ストレージ手段であり、例えば、RAM・ROM等のメモリ装置や、ハードディスクのような固定ディスク装置、フレキシブルディスク、光ディスク等を用いることができる。記憶部106には、OS(Operating System)と協働してCPUに命令を与え各種処理を行うためのコンピュータプログラムが記録されている。記憶部106は、図示の如く、利用者情報ファイル106aと、アミノ酸濃度データファイル106bと、癌状態情報ファイル106cと、指定癌状態情報ファイル106dと、多変量判別式関連情報データベース106eと、判別値ファイル106fと、評価結果ファイル106gと、を格納する。 The storage unit 106 is a storage means, and for example, a memory device such as a RAM / ROM, a fixed disk device such as a hard disk, a flexible disk, an optical disk, or the like can be used. The storage unit 106 stores a computer program for giving instructions to the CPU and performing various processes in cooperation with an OS (Operating System). As illustrated, the storage unit 106 includes a user information file 106a, an amino acid concentration data file 106b, a cancer state information file 106c, a designated cancer state information file 106d, a multivariate discriminant-related information database 106e, and a discriminant value. A file 106f and an evaluation result file 106g are stored.
 利用者情報ファイル106aは、利用者に関する利用者情報を格納する。図7は、利用者情報ファイル106aに格納される情報の一例を示す図である。利用者情報ファイル106aに格納される情報は、図7に示すように、利用者を一意に識別するための利用者IDと、利用者が正当な者であるか否かの認証を行うための利用者パスワードと、利用者の氏名と、利用者の所属する所属先を一意に識別するための所属先IDと、利用者の所属する所属先の部門を一意に識別するための部門IDと、部門名と、利用者の電子メールアドレスと、を相互に関連付けて構成されている。 The user information file 106a stores user information related to users. FIG. 7 is a diagram illustrating an example of information stored in the user information file 106a. As shown in FIG. 7, the information stored in the user information file 106a includes a user ID for uniquely identifying a user and authentication for whether or not the user is a valid person. A user password, a user name, an affiliation ID for uniquely identifying the affiliation to which the user belongs, a department ID for uniquely identifying the department to which the user belongs, The department name and the user's e-mail address are associated with each other.
 図6に戻り、アミノ酸濃度データファイル106bは、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データを格納する。図8は、アミノ酸濃度データファイル106bに格納される情報の一例を示す図である。アミノ酸濃度データファイル106bに格納される情報は、図8に示すように、評価対象である個体(サンプル)を一意に識別するための個体番号と、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データとを相互に関連付けて構成されている。ここで、図8では、アミノ酸濃度データを数値、すなわち連続尺度として扱っているが、アミノ酸濃度データは名義尺度や順序尺度でもよい。なお、名義尺度や順序尺度の場合は、それぞれの状態に対して任意の数値を与えることで解析してもよい。また、アミノ酸濃度データに、他の生体情報(例えば、腫瘍マーカー、血中サイトカイン、免疫担当細胞数、免疫担当細胞内サイトカイン、遅延型過分反応(DTH)など)を組み合わせてもよい。 Referring back to FIG. 6, the amino acid concentration data file 106b stores amino acid concentration data before starting treatment and amino acid concentration data after starting treatment. FIG. 8 is a diagram showing an example of information stored in the amino acid concentration data file 106b. As shown in FIG. 8, the information stored in the amino acid concentration data file 106b includes an individual number for uniquely identifying an individual (sample) to be evaluated, amino acid concentration data before starting treatment, and amino acid concentration after starting treatment. It is configured to correlate with data. Here, in FIG. 8, the amino acid concentration data is treated as a numerical value, that is, a continuous scale, but the amino acid concentration data may be a nominal scale or an order scale. In the case of a nominal scale or an order scale, analysis may be performed by giving an arbitrary numerical value to each state. In addition, amino acid concentration data may be combined with other biological information (eg, tumor marker, blood cytokine, number of immunocompetent cells, cytokine in immunocompetent cell, delayed excessive reaction (DTH), etc.).
 図6に戻り、癌状態情報ファイル106cは、多変量判別式を作成する際に用いる癌状態情報を格納する。図9は、癌状態情報ファイル106cに格納される情報の一例を示す図である。癌状態情報ファイル106cに格納される情報は、図9に示すように、個体番号と、癌の状態を表す指標(指標T、指標T、指標T・・・)に関する癌状態指標データ(T)と、アミノ酸濃度データと、を相互に関連付けて構成されている。ここで、図9では、癌状態指標データおよびアミノ酸濃度データを数値(すなわち連続尺度)として扱っているが、癌状態指標データおよびアミノ酸濃度データは名義尺度や順序尺度でもよい。なお、名義尺度や順序尺度の場合は、それぞれの状態に対して任意の数値を与えることで解析してもよい。また、癌状態指標データは、癌の状態のマーカーとなる既知の単一の状態指標であってもよく、数値データを用いてもよい。 Returning to FIG. 6, the cancer state information file 106 c stores cancer state information used when creating a multivariate discriminant. FIG. 9 is a diagram illustrating an example of information stored in the cancer state information file 106c. As shown in FIG. 9, the information stored in the cancer state information file 106c includes cancer state index data relating to individual numbers and indices (index T 1 , index T 2 , index T 3 ...) Representing the cancer state. (T) and amino acid concentration data are associated with each other. Here, in FIG. 9, the cancer state index data and the amino acid concentration data are treated as numerical values (that is, a continuous scale), but the cancer state index data and the amino acid concentration data may be a nominal scale or an order scale. In the case of a nominal scale or an order scale, analysis may be performed by giving an arbitrary numerical value to each state. Further, the cancer state index data may be a known single state index serving as a marker of cancer state, or numerical data may be used.
 図6に戻り、指定癌状態情報ファイル106dは、後述する癌状態情報指定部102gで指定した癌状態情報を格納する。図10は、指定癌状態情報ファイル106dに格納される情報の一例を示す図である。指定癌状態情報ファイル106dに格納される情報は、図10に示すように、個体番号と、指定した癌状態指標データと、指定したアミノ酸濃度データと、を相互に関連付けて構成されている。 Referring back to FIG. 6, the designated cancer state information file 106d stores the cancer state information designated by the cancer state information designation unit 102g described later. FIG. 10 is a diagram illustrating an example of information stored in the designated cancer state information file 106d. As shown in FIG. 10, the information stored in the designated cancer state information file 106d is configured by associating individual numbers, designated cancer state index data, and designated amino acid concentration data with each other.
 図6に戻り、多変量判別式関連情報データベース106eは、後述する候補多変量判別式作成部102h1で作成した候補多変量判別式を格納する候補多変量判別式ファイル106e1と、後述する候補多変量判別式検証部102h2での検証結果を格納する検証結果ファイル106e2と、後述する変数選択部102h3で選択したアミノ酸濃度データの組み合わせを含む癌状態情報を格納する選択癌状態情報ファイル106e3と、後述する多変量判別式作成部102hで作成した多変量判別式を格納する多変量判別式ファイル106e4と、で構成される。 Returning to FIG. 6, the multivariate discriminant-related information database 106e includes a candidate multivariate discriminant file 106e1 for storing the candidate multivariate discriminant created by the candidate multivariate discriminant-preparing part 102h1, which will be described later, and a candidate multivariate discriminant described later. A verification result file 106e2 for storing a verification result in the discriminant verification unit 102h2, a selected cancer state information file 106e3 for storing cancer state information including a combination of amino acid concentration data selected by a variable selection unit 102h3 described later, and a later-described A multivariate discriminant file 106e4 that stores the multivariate discriminant created by the multivariate discriminant creation unit 102h.
 候補多変量判別式ファイル106e1は、後述する候補多変量判別式作成部102h1で作成した候補多変量判別式を格納する。図11は、候補多変量判別式ファイル106e1に格納される情報の一例を示す図である。候補多変量判別式ファイル106e1に格納される情報は、図11に示すように、ランクと、候補多変量判別式(図11では、F(Gly,Leu,Phe,・・・)やF(Gly,Leu,Phe,・・・)、F(Gly,Leu,Phe,・・・)など)とを相互に関連付けて構成されている。 The candidate multivariate discriminant file 106e1 stores the candidate multivariate discriminant created by the candidate multivariate discriminant creation unit 102h1 described later. FIG. 11 is a diagram illustrating an example of information stored in the candidate multivariate discriminant file 106e1. As shown in FIG. 11, information stored in the candidate multivariate discriminant file 106e1 includes a rank, a candidate multivariate discriminant (in FIG. 11, F 1 (Gly, Leu, Phe,...)) And F 2. (Gly, Leu, Phe,...), F 3 (Gly, Leu, Phe,...)) Are associated with each other.
 図6に戻り、検証結果ファイル106e2は、後述する候補多変量判別式検証部102h2での検証結果を格納する。図12は、検証結果ファイル106e2に格納される情報の一例を示す図である。検証結果ファイル106e2に格納される情報は、図12に示すように、ランクと、候補多変量判別式(図12では、F(Gly,Leu,Phe,・・・)やF(Gly,Leu,Phe,・・・)、F(Gly,Leu,Phe,・・・)など)と、各候補多変量判別式の検証結果(例えば各候補多変量判別式の評価値)と、を相互に関連付けて構成されている。 Returning to FIG. 6, the verification result file 106e2 stores the verification result in the candidate multivariate discriminant verification unit 102h2 described later. FIG. 12 is a diagram illustrating an example of information stored in the verification result file 106e2. As shown in FIG. 12, the information stored in the verification result file 106e2 includes rank, candidate multivariate discriminant (in FIG. 12, F k (Gly, Leu, Phe,...) And F m (Gly, Le, Phe,...), Fl (Gly, Leu, Phe,...)) And the verification results of each candidate multivariate discriminant (for example, the evaluation value of each candidate multivariate discriminant). They are related to each other.
 図6に戻り、選択癌状態情報ファイル106e3は、後述する変数選択部102h3で選択した変数に対応するアミノ酸濃度データの組み合わせを含む癌状態情報を格納する。図13は、選択癌状態情報ファイル106e3に格納される情報の一例を示す図である。選択癌状態情報ファイル106e3に格納される情報は、図13に示すように、個体番号と、後述する癌状態情報指定部102gで指定した癌状態指標データと、後述する変数選択部102h3で選択したアミノ酸濃度データと、を相互に関連付けて構成されている。 Referring back to FIG. 6, the selected cancer state information file 106e3 stores cancer state information including a combination of amino acid concentration data corresponding to variables selected by the variable selection unit 102h3 described later. FIG. 13 is a diagram illustrating an example of information stored in the selected cancer state information file 106e3. As shown in FIG. 13, the information stored in the selected cancer state information file 106e3 is selected by an individual number, cancer state index data designated by a cancer state information designation unit 102g described later, and a variable selection unit 102h3 described later. The amino acid concentration data is associated with each other.
 図6に戻り、多変量判別式ファイル106e4は、後述する多変量判別式作成部102hで作成した多変量判別式を格納する。図14は、多変量判別式ファイル106e4に格納される情報の一例を示す図である。多変量判別式ファイル106e4に格納される情報は、図14に示すように、ランクと、多変量判別式(図14では、F(Phe,・・・)やF(Gly,Leu,Phe)、F(Gly,Leu,Phe,・・・)など)と、各式作成手法に対応する閾値と、各多変量判別式の検証結果(例えば各多変量判別式の評価値)と、を相互に関連付けて構成されている。 Returning to FIG. 6, the multivariate discriminant file 106e4 stores the multivariate discriminant created by the multivariate discriminant-preparing part 102h described later. FIG. 14 is a diagram illustrating an example of information stored in the multivariate discriminant file 106e4. As shown in FIG. 14, the information stored in the multivariate discriminant file 106e4 includes the rank, the multivariate discriminant (in FIG. 14, F p (Phe,...) And F p (Gly, Leu, Phe). ), F k (Gly, Leu, Phe,...)), A threshold corresponding to each formula creation method, a verification result of each multivariate discriminant (for example, an evaluation value of each multivariate discriminant), Are related to each other.
 図6に戻り、判別値ファイル106fは、後述する判別値算出部102iで算出した判別値を格納する。図15は、判別値ファイル106fに格納される情報の一例を示す図である。判別値ファイル106fに格納される情報は、図15に示すように、評価対象である個体(サンプル)を一意に識別するための個体番号と、ランク(多変量判別式を一意に識別するための番号)と、判別値と、を相互に関連付けて構成されている。 Returning to FIG. 6, the discriminant value file 106f stores the discriminant value calculated by the discriminant value calculator 102i described later. FIG. 15 is a diagram illustrating an example of information stored in the discrimination value file 106f. As shown in FIG. 15, information stored in the discriminant value file 106f includes an individual number for uniquely identifying an individual (sample) to be evaluated and a rank (for uniquely identifying a multivariate discriminant). Number) and the discriminant value are associated with each other.
 図6に戻り、評価結果ファイル106gは、後述する判別値基準評価部102jでの評価結果(具体的には、後述する判別値基準判別部102j1での判別結果・分類結果)を格納する。図16は、評価結果ファイル106gに格納される情報の一例を示す図である。評価結果ファイル106gに格納される情報は、評価対象である個体(サンプル)を一意に識別するための個体番号と、評価対象の複数のアミノ酸濃度データ(治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データ)と、多変量判別式で算出した1つまたは複数の判別値(例えば、治療開始前判別値、治療開始後判別値または治療開始前後判別値など)と、癌免疫療法の治療効果の評価に関する評価結果と、を相互に関連付けて構成されている。 Referring back to FIG. 6, the evaluation result file 106g stores the evaluation result in the discriminant value criterion-evaluating unit 102j described later (specifically, the discrimination result / classification result in the discriminant value criterion-discriminating unit 102j1 described later). FIG. 16 is a diagram illustrating an example of information stored in the evaluation result file 106g. Information stored in the evaluation result file 106g includes an individual number for uniquely identifying an individual (sample) to be evaluated, and a plurality of evaluation target amino acid concentration data (amino acid concentration data before treatment start and amino acid after treatment start). Concentration data), one or more discriminant values calculated by a multivariate discriminant (for example, a discriminant value before treatment start, a discriminant value after treatment start or a discriminant value before and after treatment start), and the therapeutic effect of cancer immunotherapy The evaluation results related to the evaluation are associated with each other.
 図6に戻り、記憶部106には、上述した情報以外にその他情報として、Webサイトをクライアント装置200に提供するための各種のWebデータや、CGIプログラム等が記録されている。Webデータとしては後述する各種のWebページを表示するためのデータ等があり、これらデータは例えばHTMLやXMLで記述されたテキストファイルとして形成されている。また、Webデータを作成するための部品用のファイルや作業用のファイルやその他一時的なファイル等も記憶部106に記憶される。記憶部106には、必要に応じて、クライアント装置200に送信するための音声をWAVE形式やAIFF形式の如き音声ファイルで格納したり、静止画や動画をJPEG形式やMPEG2形式の如き画像ファイルで格納したりすることができる。 Referring back to FIG. 6, the storage unit 106 stores various types of Web data for providing the Web site to the client device 200, CGI programs, and the like as other information in addition to the information described above. The Web data includes data for displaying various Web pages to be described later, and these data are formed as text files described in HTML or XML, for example. In addition, a part file, a work file, and other temporary files for creating Web data are also stored in the storage unit 106. The storage unit 106 stores audio for transmission to the client device 200 as an audio file such as WAVE format or AIFF format, and stores still images or moving images as image files such as JPEG format or MPEG2 format as necessary. Can be stored.
 通信インターフェース部104は、癌免疫療法評価装置100とネットワーク300(またはルータ等の通信装置)との間における通信を媒介する。すなわち、通信インターフェース部104は、他の端末と通信回線を介してデータを通信する機能を有する。 The communication interface unit 104 mediates communication between the cancer immunotherapy evaluation device 100 and the network 300 (or a communication device such as a router). That is, the communication interface unit 104 has a function of communicating data with other terminals via a communication line.
 入出力インターフェース部108は、入力装置112や出力装置114に接続する。ここで、出力装置114には、モニタ(家庭用テレビを含む)の他、スピーカやプリンタを用いることができる(なお、以下では、出力装置114をモニタ114として記載する場合がある。)。入力装置112には、キーボードやマウスやマイクの他、マウスと協働してポインティングデバイス機能を実現するモニタを用いることができる。 The input / output interface unit 108 is connected to the input device 112 and the output device 114. Here, in addition to a monitor (including a home television), a speaker or a printer can be used as the output device 114 (hereinafter, the output device 114 may be described as the monitor 114). As the input device 112, a monitor that realizes a pointing device function in cooperation with a mouse can be used in addition to a keyboard, a mouse, and a microphone.
 制御部102は、OS(Operating System)等の制御プログラム・各種の処理手順等を規定したプログラム・所要データなどを格納するための内部メモリを有し、これらのプログラムに基づいて種々の情報処理を実行する。制御部102は、図示の如く、大別して、要求解釈部102aと閲覧処理部102bと認証処理部102cと電子メール生成部102dとWebページ生成部102eと受信部102fと癌状態情報指定部102gと多変量判別式作成部102hと判別値算出部102iと判別値基準評価部102jと結果出力部102kと送信部102mとを備えている。制御部102は、データベース装置400から送信された癌状態情報やクライアント装置200から送信されたアミノ酸濃度データに対して、欠損値のあるデータの除去・外れ値の多いデータの除去・欠損値のあるデータの多い変数の除去などのデータ処理も行う。 The control unit 102 has an internal memory for storing a control program such as an OS (Operating System), a program defining various processing procedures, and necessary data, and performs various information processing based on these programs. Execute. As shown in the figure, the control unit 102 is roughly divided into a request interpretation unit 102a, a browsing processing unit 102b, an authentication processing unit 102c, an email generation unit 102d, a Web page generation unit 102e, a reception unit 102f, and a cancer state information designation unit 102g. A multivariate discriminant creation unit 102h, a discriminant value calculation unit 102i, a discriminant value criterion evaluation unit 102j, a result output unit 102k, and a transmission unit 102m are provided. The control unit 102 removes data with missing values, removes data with many outliers, and has missing values with respect to the cancer state information transmitted from the database device 400 and the amino acid concentration data transmitted from the client device 200. Data processing such as removal of variables with a lot of data is also performed.
 要求解釈部102aは、クライアント装置200やデータベース装置400からの要求内容を解釈し、その解釈結果に応じて制御部102の各部に処理を受け渡す。閲覧処理部102bは、クライアント装置200からの各種画面の閲覧要求を受けて、これら画面のWebデータの生成や送信を行なう。認証処理部102cは、クライアント装置200やデータベース装置400からの認証要求を受けて、認証判断を行う。電子メール生成部102dは、各種の情報を含んだ電子メールを生成する。Webページ生成部102eは、利用者がクライアント装置200で閲覧するWebページを生成する。 The request interpretation unit 102a interprets the request content from the client device 200 or the database device 400, and passes the processing to each unit of the control unit 102 according to the interpretation result. Upon receiving browsing requests for various screens from the client device 200, the browsing processing unit 102b generates and transmits Web data for these screens. Upon receiving an authentication request from the client device 200 or the database device 400, the authentication processing unit 102c makes an authentication determination. The e-mail generation unit 102d generates an e-mail including various types of information. The web page generation unit 102e generates a web page that the user browses on the client device 200.
 受信部102fは、クライアント装置200やデータベース装置400から送信された情報(具体的には、アミノ酸濃度データや癌状態情報、多変量判別式など)を、ネットワーク300を介して受信する。癌状態情報指定部102gは、多変量判別式を作成するにあたり、対象とする癌状態指標データおよびアミノ酸濃度データを指定する。 The receiving unit 102 f receives information (specifically, amino acid concentration data, cancer state information, multivariate discriminant, etc.) transmitted from the client device 200 or the database device 400 via the network 300. When creating the multivariate discriminant, the cancer state information specifying unit 102g specifies target cancer state index data and amino acid concentration data.
 多変量判別式作成部102hは、受信部102fで受信した癌状態情報や癌状態情報指定部102gで指定した癌状態情報に基づいて多変量判別式を作成する。具体的には、多変量判別式作成部102hは、癌状態情報から、候補多変量判別式作成部102h1、候補多変量判別式検証部102h2および変数選択部102h3を繰り返し実行させることにより蓄積された検証結果に基づいて、複数の候補多変量判別式の中から多変量判別式として採用する候補多変量判別式を選出することで、多変量判別式を作成する。 The multivariate discriminant creating unit 102h creates a multivariate discriminant based on the cancer state information received by the receiving unit 102f and the cancer state information designated by the cancer state information designating unit 102g. Specifically, the multivariate discriminant-preparing part 102h is accumulated by repeatedly executing the candidate multivariate discriminant-preparing part 102h1, the candidate multivariate discriminant-verifying part 102h2, and the variable selecting part 102h3 from the cancer state information. A multivariate discriminant is created by selecting a candidate multivariate discriminant to be adopted as a multivariate discriminant from a plurality of candidate multivariate discriminants based on the verification result.
 なお、多変量判別式が予め記憶部106の所定の記憶領域に格納されている場合には、多変量判別式作成部102hは、記憶部106から所望の多変量判別式を選択することで、多変量判別式を作成してもよい。また、多変量判別式作成部102hは、多変量判別式を予め格納した他のコンピュータ装置(例えばデータベース装置400)から所望の多変量判別式を選択しダウンロードすることで、多変量判別式を作成してもよい。 When the multivariate discriminant is stored in a predetermined storage area of the storage unit 106 in advance, the multivariate discriminant-preparing unit 102h selects a desired multivariate discriminant from the storage unit 106, A multivariate discriminant may be created. In addition, the multivariate discriminant creation unit 102h creates a multivariate discriminant by selecting and downloading a desired multivariate discriminant from another computer device (for example, the database device 400) that stores the multivariate discriminant in advance. May be.
 ここで、多変量判別式作成部102hの構成について図17を参照して説明する。図17は、多変量判別式作成部102hの構成を示すブロック図であり、該構成のうち本発明に関係する部分のみを概念的に示している。多変量判別式作成部102hは、候補多変量判別式作成部102h1と、候補多変量判別式検証部102h2と、変数選択部102h3と、をさらに備えている。候補多変量判別式作成部102h1は、癌状態情報から所定の式作成手法に基づいて多変量判別式の候補である候補多変量判別式を作成する。なお、候補多変量判別式作成部102h1は、癌状態情報から、複数の異なる式作成手法を併用して複数の候補多変量判別式を作成してもよい。候補多変量判別式検証部102h2は、候補多変量判別式作成部102h1で作成した候補多変量判別式を所定の検証手法に基づいて検証する。なお、候補多変量判別式検証部102h2は、ランダムサンプリング法、ブートストラップ法、ホールドアウト法、N-フォールド法またはリーブワンアウト法などのうち少なくとも1つに基づいて候補多変量判別式の判別率、感度、特異度、情報量基準、ROC_AUC(受信者特性曲線の曲線下面積)のうち少なくとも1つに関して検証してもよい。変数選択部102h3は、所定の変数選択手法に基づいて候補多変量判別式の変数を選択することで、候補多変量判別式を作成する際に用いる癌状態情報に含まれるアミノ酸濃度データの組み合わせを選択する。なお、変数選択部102h3は、検証結果からステップワイズ法、ベストパス法、近傍探索法、遺伝的アルゴリズムのうち少なくとも1つに基づいて候補多変量判別式の変数を選択してもよい。 Here, the configuration of the multivariate discriminant-preparing part 102h will be described with reference to FIG. FIG. 17 is a block diagram showing the configuration of the multivariate discriminant-preparing part 102h, and conceptually shows only the part related to the present invention. The multivariate discriminant creation unit 102h further includes a candidate multivariate discriminant creation unit 102h1, a candidate multivariate discriminant verification unit 102h2, and a variable selection unit 102h3. The candidate multivariate discriminant creation unit 102h1 creates a candidate multivariate discriminant that is a candidate for the multivariate discriminant based on a predetermined formula creation method from the cancer state information. Note that the candidate multivariate discriminant-preparing part 102h1 may create a plurality of candidate multivariate discriminants from the cancer state information by using a plurality of different formula creation methods. The candidate multivariate discriminant verification unit 102h2 verifies the candidate multivariate discriminant created by the candidate multivariate discriminant creation unit 102h1 based on a predetermined verification method. The candidate multivariate discriminant verification unit 102h2 determines the discrimination rate of the candidate multivariate discriminant based on at least one of a random sampling method, a bootstrap method, a holdout method, an N-fold method, or a leave one out method. , Sensitivity, specificity, information criterion, and ROC_AUC (area under the receiver characteristic curve) may be verified. The variable selection unit 102h3 selects a variable of the candidate multivariate discriminant based on a predetermined variable selection method, so that a combination of amino acid concentration data included in the cancer state information used when creating the candidate multivariate discriminant is selected. select. Note that the variable selection unit 102h3 may select a variable of the candidate multivariate discriminant from the verification result based on at least one of the stepwise method, the best path method, the neighborhood search method, and the genetic algorithm.
 図6に戻り、判別値算出部102iは、多変量判別式作成部102hで作成した多変量判別式、および受信部102fで受信した評価対象のアミノ酸濃度データ(具体的には、治療開始前アミノ酸濃度データ、治療開始後アミノ酸濃度データなど)に基づいて、当該多変量判別式の値である1つまたは複数の判別値(具体的には、治療開始前判別値、治療開始後判別値、治療開始前後判別値など)を算出する。なお、多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つでもよい。 Returning to FIG. 6, the discriminant value calculation unit 102 i includes the multivariate discriminant created by the multivariate discriminant creation unit 102 h and the evaluation target amino acid concentration data received by the receiving unit 102 f (specifically, the amino acid concentration before starting treatment). Based on the concentration data, amino acid concentration data after treatment start, etc., one or more discriminant values that are the values of the multivariate discriminant (specifically, discriminant value before treatment start, discriminant value after treatment start, treatment Calculate the discriminant value before and after the start. Multivariate discriminants are logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created by support vector machine, formula created by Mahalanobis distance method, formula created by canonical discriminant analysis. Any one of the expressions created by the decision tree may be used.
 ここで、判別値算出部102iは、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、1つまたは複数の判別値を算出してもよい。具体的には、後述する判別値基準判別部102j1で評価対象に対して癌免疫療法による治療が有効であるか否かを判別したり、また、当該治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりする場合には、判別値算出部102iは、治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、1つまたは複数の判別値を算出してもよい。なお、多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを変数として含む分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを変数として含むロジスティック回帰式でもよい。 Here, the discriminant value calculation unit 102i is configured to include the Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, and the like included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. At least one concentration value among Thr, Met, Lys, Arg, Gly, Cys2, Pro, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, One or more discriminant values may be calculated based on a multivariate discriminant including at least one of Lys, Arg, Gly, Cys2, and Pro as a variable. Specifically, the discriminant value criterion discriminating unit 102j1 to be described later discriminates whether or not the cancer immunotherapy treatment is effective for the evaluation target, and considers the degree of possibility that the treatment is effective. When the evaluation target is classified into any one of the plurality of categories (ranks) defined as above, the discriminant value calculation unit 102i uses the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment. At least one of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro, and Val , Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gl , Cys2, based on the multivariate discriminant containing at least one as a variable of Pro, may calculate the one or more discriminant value. The multivariate discriminant is a fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as a variable, or Trp, Thr, His, Arg, Ile, A logistic regression equation including at least one of Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu as a variable may be used.
 図6に戻り、判別値基準評価部102jは、判別値算出部102iで算出した1つまたは複数の判別値に基づいて、評価対象に対する癌免疫療法による治療の効果を評価する。判別値基準評価部102jは、例えば、評価対象に対して治療が有効であるかを評価する。判別値基準評価部102jは、判別値基準判別部102j1をさらに備えている。ここで、判別値基準評価部102jの構成について図18を参照して説明する。図18は、判別値基準評価部102jの構成を示すブロック図であり、該構成のうち本発明に関係する部分のみを概念的に示している。 Referring back to FIG. 6, the discriminant value criterion-evaluating unit 102j evaluates the effect of treatment by cancer immunotherapy on the evaluation target based on one or more discriminant values calculated by the discriminant value calculating unit 102i. The discriminant value criterion-evaluating unit 102j evaluates, for example, whether the treatment is effective for the evaluation target. The discrimination value criterion evaluation unit 102j further includes a discrimination value criterion discrimination unit 102j1. Here, the configuration of the discriminant value criterion-evaluating unit 102j will be described with reference to FIG. FIG. 18 is a block diagram showing the configuration of the discriminant value criterion-evaluating unit 102j, and conceptually shows only the portion related to the present invention.
 判別値基準判別部102j1は、判別値算出部102iで算出した1つまたは複数の判別値に基づいて、評価対象に対して癌免疫療法による治療が有効であるか否かを判別したり、治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりする。具体的には、判別値基準判別部102j1は、判別値算出部102iで算出した1つまたは複数の判別値と1つまたは複数の閾値(カットオフ値)とを比較することで、評価対象に対して癌免疫療法による治療が有効であるか否かを判別したり、治療が有効である可能性の程度を考慮して定義された複数の区分(ランク)のうちのどれか1つに評価対象を分類したりする。 The discriminant value criterion discriminating unit 102j1 discriminates whether or not the cancer immunotherapy treatment is effective for the evaluation target based on the one or more discriminant values calculated by the discriminant value calculating unit 102i. The evaluation target is classified into any one of a plurality of categories (ranks) defined in consideration of the possibility of being effective. Specifically, the discriminant value criterion discriminating unit 102j1 compares the one or more discriminant values calculated by the discriminant value calculating unit 102i with one or more threshold values (cut-off values) to be evaluated. In contrast, whether cancer immunotherapy treatment is effective or not, and evaluate to any one of multiple categories (ranks) defined taking into consideration the degree of possibility that the treatment is effective Or classify objects.
 図6に戻り、結果出力部102kは、制御部102の各処理部での処理結果(判別値基準評価部102jでの評価結果(具体的には判別値基準判別部102j1での判別結果または分類結果)を含む)等を出力装置114に出力する。 Returning to FIG. 6, the result output unit 102k outputs the processing results in each processing unit of the control unit 102 (evaluation results in the discrimination value criterion evaluation unit 102j (specifically, discrimination results or classification in the discrimination value criterion discrimination unit 102j1). Including the result) is output to the output device 114.
 送信部102mは、評価対象のアミノ酸濃度データの送信元のクライアント装置200に対して、例えば、判別値、評価結果(例えば判別結果、分類結果など)などを送信したり、データベース装置400に対して、例えば、癌免疫療法評価装置100で作成した多変量判別式や評価結果(例えば、判別結果、分類結果など)などを送信したりする。 The transmission unit 102m transmits, for example, a discrimination value, an evaluation result (eg, a discrimination result, a classification result, etc.) to the client device 200 that is a transmission source of the amino acid concentration data to be evaluated, or a database device 400. For example, a multivariate discriminant created by the cancer immunotherapy evaluation apparatus 100, an evaluation result (for example, a discrimination result, a classification result, etc.), etc. are transmitted.
 つぎに、本システムのクライアント装置200の構成について図19を参照して説明する。図19は、本システムのクライアント装置200の構成の一例を示すブロック図であり、該構成のうち本発明に関係する部分のみを概念的に示している。 Next, the configuration of the client device 200 of this system will be described with reference to FIG. FIG. 19 is a block diagram showing an example of the configuration of the client apparatus 200 of the present system, and conceptually shows only the portion related to the present invention in the configuration.
 クライアント装置200は、制御部210とROM220とHD230とRAM240と入力装置250と出力装置260と入出力IF270と通信IF280とで構成されており、これら各部は任意の通信路を介して通信可能に接続されている。 The client device 200 includes a control unit 210, a ROM 220, an HD 230, a RAM 240, an input device 250, an output device 260, an input / output IF 270, and a communication IF 280. These units are communicably connected via an arbitrary communication path. Has been.
 制御部210は、Webブラウザ211、電子メーラ212、受信部213、送信部214を備えている。Webブラウザ211は、Webデータを解釈し、解釈したWebデータを後述するモニタ261に表示するブラウズ処理を行う。なお、Webブラウザ211には、ストリーム映像の受信・表示・フィードバック等を行う機能を備えたストリームプレイヤ等の各種のソフトウェアをプラグインしてもよい。電子メーラ212は、所定の通信規約(例えば、SMTP(Simple Mail Transfer Protocol)やPOP3(Post Office Protocol version 3)等)に従って電子メールの送受信を行う。受信部213(本発明の結果取得手段の一例に相当)は、通信IF280を介して、癌免疫療法評価装置100から送信された、判別値、評価結果(例えば判別結果、分類結果など)などの各種情報を受信する。要するに、クライアント装置は、判別値や評価結果などの各種情報を取得する機能を有する。送信部214は、通信IF280を介して、評価対象のアミノ酸濃度データ(具体的には、治療開始前アミノ酸濃度データ、治療開始後アミノ酸濃度データなど)などの各種情報を癌免疫療法評価装置100へ送信する。 The control unit 210 includes a web browser 211, an electronic mailer 212, a reception unit 213, and a transmission unit 214. The web browser 211 performs browse processing for interpreting the web data and displaying the interpreted web data on a monitor 261 described later. The Web browser 211 may be plugged in with various software such as a stream player having a function of receiving, displaying, and feeding back a stream video. The electronic mailer 212 transmits and receives electronic mail according to a predetermined communication protocol (for example, SMTP (Simple Mail Transfer Protocol), POP3 (Post Office Protocol version 3), etc.). The receiving unit 213 (corresponding to an example of the result acquisition unit of the present invention), such as a discrimination value and an evaluation result (for example, a discrimination result, a classification result, etc.) transmitted from the cancer immunotherapy evaluation apparatus 100 via the communication IF 280 Receive various information. In short, the client device has a function of acquiring various information such as a discrimination value and an evaluation result. The transmission unit 214 sends various information such as amino acid concentration data to be evaluated (specifically, pre-treatment amino acid concentration data, post-treatment amino acid concentration data, etc.) to the cancer immunotherapy evaluation apparatus 100 via the communication IF 280. Send.
 入力装置250はキーボードやマウスやマイク等である。なお、後述するモニタ261もマウスと協働してポインティングデバイス機能を実現する。出力装置260は、通信IF280を介して受信した情報を出力する出力手段であり、モニタ(家庭用テレビを含む)261およびプリンタ262を含む。この他、出力装置260にスピーカ等を設けてもよい。入出力IF270は入力装置250や出力装置260に接続する。 The input device 250 is a keyboard, a mouse, a microphone, or the like. A monitor 261, which will be described later, also realizes a pointing device function in cooperation with the mouse. The output device 260 is an output unit that outputs information received via the communication IF 280, and includes a monitor (including a home television) 261 and a printer 262. In addition, the output device 260 may be provided with a speaker or the like. The input / output IF 270 is connected to the input device 250 and the output device 260.
 通信IF280は、クライアント装置200とネットワーク300(またはルータ等の通信装置)とを通信可能に接続する。換言すると、クライアント装置200は、モデムやTAやルータなどの通信装置および電話回線を介して、または専用線を介してネットワーク300に接続される。これにより、クライアント装置200は、所定の通信規約に従って癌免疫療法評価装置100にアクセスすることができる。 The communication IF 280 connects the client device 200 and the network 300 (or a communication device such as a router) so that they can communicate with each other. In other words, the client device 200 is connected to the network 300 via a communication device such as a modem, TA, or router and a telephone line, or via a dedicated line. Thereby, the client apparatus 200 can access the cancer immunotherapy evaluation apparatus 100 according to a predetermined communication protocol.
 ここで、プリンタ・モニタ・イメージスキャナ等の周辺装置を必要に応じて接続した情報処理装置(例えば、既知のパーソナルコンピュータ・ワークステーション・家庭用ゲーム装置・インターネットTV・PHS端末・携帯端末・移動体通信端末・PDA等の情報処理端末など)に、Webデータのブラウジング機能や電子メール機能を実現させるソフトウェア(プログラム、データ等を含む)を実装することにより、クライアント装置200を実現してもよい。 Here, an information processing device (for example, a known personal computer, workstation, home game device, Internet TV, PHS terminal, portable terminal, mobile object) connected with peripheral devices such as a printer, a monitor, and an image scanner as necessary. The client device 200 may be realized by installing software (including programs, data, and the like) that realizes a Web data browsing function and an e-mail function in a communication terminal / information processing terminal such as a PDA).
 また、クライアント装置200の制御部210は、制御部210で行う処理の全部または任意の一部を、CPUおよび当該CPUにて解釈して実行するプログラムで実現してもよい。ROM220またはHD230には、OS(Operating System)と協働してCPUに命令を与え、各種処理を行うためのコンピュータプログラムが記録されている。当該コンピュータプログラムは、RAM240にロードされることで実行され、CPUと協働して制御部210を構成する。また、当該コンピュータプログラムは、クライアント装置200と任意のネットワークを介して接続されるアプリケーションプログラムサーバに記録されてもよく、クライアント装置200は、必要に応じてその全部または一部をダウンロードしてもよい。また、制御部210で行う処理の全部または任意の一部を、ワイヤードロジック等によるハードウェアで実現してもよい。 Further, the control unit 210 of the client device 200 may be realized by a CPU and a program that is interpreted and executed by the CPU and all or any part of the processing performed by the control unit 210. The ROM 220 or the HD 230 stores computer programs for giving instructions to the CPU and performing various processes in cooperation with an OS (Operating System). The computer program is executed by being loaded into the RAM 240, and constitutes the control unit 210 in cooperation with the CPU. Further, the computer program may be recorded in an application program server connected to the client apparatus 200 via an arbitrary network, and the client apparatus 200 may download all or a part thereof as necessary. . In addition, all or any part of the processing performed by the control unit 210 may be realized by hardware such as wired logic.
 つぎに、本システムのネットワーク300について図4、図5を参照して説明する。ネットワーク300は、癌免疫療法評価装置100とクライアント装置200とデータベース装置400とを相互に通信可能に接続する機能を有し、例えばインターネットやイントラネットやLAN(有線/無線の双方を含む)等である。なお、ネットワーク300は、VANや、パソコン通信網や、公衆電話網(アナログ/デジタルの双方を含む)や、専用回線網(アナログ/デジタルの双方を含む)や、CATV網や、携帯回線交換網または携帯パケット交換網(IMT2000方式、GSM(登録商標)方式またはPDC/PDC-P方式等を含む)や、無線呼出網や、Bluetooth(登録商標)等の局所無線網や、PHS網や、衛星通信網(CS、BSまたはISDB等を含む)等でもよい。 Next, the network 300 of this system will be described with reference to FIGS. The network 300 has a function of connecting the cancer immunotherapy evaluation apparatus 100, the client apparatus 200, and the database apparatus 400 so that they can communicate with each other, such as the Internet, an intranet, or a LAN (including both wired and wireless). . The network 300 includes a VAN, a personal computer communication network, a public telephone network (including both analog / digital), a dedicated line network (including both analog / digital), a CATV network, and a mobile line switching network. Or mobile packet switching network (including IMT2000 system, GSM (registered trademark) system or PDC / PDC-P system), wireless paging network, local wireless network such as Bluetooth (registered trademark), PHS network, satellite A communication network (including CS, BS or ISDB) may be used.
 つぎに、本システムのデータベース装置400の構成について図20を参照して説明する。図20は、本システムのデータベース装置400の構成の一例を示すブロック図であり、該構成のうち本発明に関係する部分のみを概念的に示している。 Next, the configuration of the database apparatus 400 of this system will be described with reference to FIG. FIG. 20 is a block diagram showing an example of the configuration of the database apparatus 400 of this system, and conceptually shows only the portion related to the present invention in the configuration.
 データベース装置400は、癌免疫療法評価装置100または当該データベース装置で多変量判別式を作成する際に用いる癌状態情報や、癌免疫療法評価装置100で作成した多変量判別式、癌免疫療法評価装置100で得られた評価結果(具体的には判別結果)などを格納する機能を有する。図20に示すように、データベース装置400は、当該データベース装置を統括的に制御するCPU等の制御部402と、ルータ等の通信装置および専用線等の有線または無線の通信回路を介して当該データベース装置をネットワーク300に通信可能に接続する通信インターフェース部404と、各種のデータベースやテーブルやファイル(例えばWebページ用ファイル)などを格納する記憶部406と、入力装置412や出力装置414に接続する入出力インターフェース部408と、で構成されており、これら各部は任意の通信路を介して通信可能に接続されている。 The database device 400 is a cancer immunotherapy evaluation device 100 or cancer state information used when creating a multivariate discriminant in the database device, a multivariate discriminant created in the cancer immunotherapy evaluation device 100, and a cancer immunotherapy evaluation device. 100 has a function of storing an evaluation result (specifically, a discrimination result) obtained in 100. As shown in FIG. 20, the database device 400 includes a control unit 402 such as a CPU that comprehensively controls the database device, a communication device such as a router, and a wired or wireless communication circuit such as a dedicated line. A communication interface unit 404 that connects the apparatus to the network 300 to be communicable, a storage unit 406 that stores various databases, tables, and files (for example, files for Web pages), and an input unit that connects to the input unit 412 and the output unit 414. And an output interface unit 408. These units are communicably connected via an arbitrary communication path.
 記憶部406は、ストレージ手段であり、例えば、RAM・ROM等のメモリ装置や、ハードディスクのような固定ディスク装置や、フレキシブルディスクや、光ディスク等を用いることができる。記憶部406には、各種処理に用いる各種プログラム等を格納する。通信インターフェース部404は、データベース装置400とネットワーク300(またはルータ等の通信装置)との間における通信を媒介する。すなわち、通信インターフェース部404は、他の端末と通信回線を介してデータを通信する機能を有する。入出力インターフェース部408は、入力装置412や出力装置414に接続する。ここで、出力装置414には、モニタ(家庭用テレビを含む)の他、スピーカやプリンタを用いることができる(なお、以下で、出力装置414をモニタ414として記載する場合がある。)。また、入力装置412には、キーボードやマウスやマイクの他、マウスと協働してポインティングデバイス機能を実現するモニタを用いることができる。 The storage unit 406 is a storage means, and for example, a memory device such as a RAM / ROM, a fixed disk device such as a hard disk, a flexible disk, an optical disk, or the like can be used. The storage unit 406 stores various programs used for various processes. The communication interface unit 404 mediates communication between the database device 400 and the network 300 (or a communication device such as a router). That is, the communication interface unit 404 has a function of communicating data with other terminals via a communication line. The input / output interface unit 408 is connected to the input device 412 and the output device 414. Here, in addition to a monitor (including a home TV), a speaker or a printer can be used as the output device 414 (hereinafter, the output device 414 may be described as the monitor 414). In addition to the keyboard, mouse, and microphone, the input device 412 can be a monitor that realizes a pointing device function in cooperation with the mouse.
 制御部402は、OS(Operating System)等の制御プログラム・各種の処理手順等を規定したプログラム・所要データなどを格納するための内部メモリを有し、これらのプログラムに基づいて種々の情報処理を実行する。制御部402は、図示の如く、大別して、要求解釈部402aと閲覧処理部402bと認証処理部402cと電子メール生成部402dとWebページ生成部402eと送信部402fとを備えている。 The control unit 402 has an internal memory for storing a control program such as an OS (Operating System), a program that defines various processing procedures, and necessary data, and performs various information processing based on these programs. Execute. As shown in the figure, the control unit 402 is roughly divided into a request interpreting unit 402a, a browsing processing unit 402b, an authentication processing unit 402c, an e-mail generating unit 402d, a Web page generating unit 402e, and a transmitting unit 402f.
 要求解釈部402aは、癌免疫療法評価装置100からの要求内容を解釈し、その解釈結果に応じて制御部402の各部に処理を受け渡す。閲覧処理部402bは、癌免疫療法評価装置100からの各種画面の閲覧要求を受けて、これら画面のWebデータの生成や送信を行う。認証処理部402cは、癌免疫療法評価装置100からの認証要求を受けて、認証判断を行う。電子メール生成部402dは、各種の情報を含んだ電子メールを生成する。Webページ生成部402eは、利用者がクライアント装置200で閲覧するWebページを生成する。送信部402fは、癌状態情報や多変量判別式などの各種情報を、癌免疫療法評価装置100へ送信する。 The request interpretation unit 402a interprets the request content from the cancer immunotherapy evaluation apparatus 100, and passes the processing to each unit of the control unit 402 according to the interpretation result. Upon receiving browsing requests for various screens from the cancer immunotherapy evaluation apparatus 100, the browsing processing unit 402b generates and transmits Web data for these screens. The authentication processing unit 402c receives an authentication request from the cancer immunotherapy evaluation device 100 and makes an authentication determination. The e-mail generation unit 402d generates an e-mail including various types of information. The web page generation unit 402e generates a web page that the user browses on the client device 200. The transmission unit 402f transmits various types of information such as cancer state information and multivariate discriminants to the cancer immunotherapy evaluation apparatus 100.
[2-3.第2実施形態の具体例]
 ここでは、第2実施形態の具体例について図21を参照して説明する。図21は、第2実施形態にかかる癌免疫療法評価サービス処理の一例を示すフローチャートである。
[2-3. Specific Example of Second Embodiment]
Here, a specific example of the second embodiment will be described with reference to FIG. FIG. 21 is a flowchart illustrating an example of a cancer immunotherapy evaluation service process according to the second embodiment.
 なお、本処理で用いるアミノ酸濃度データは、例えば動物やヒトなどの個体から予め採取した血液(例えば血漿、血清などを含む)を、以下の(A)または(B)などの測定方法で専門業者が分析又は独自に分析して得たアミノ酸の濃度値に関するものである。ここで、アミノ酸濃度の単位は、例えばモル濃度や重量濃度、これらの濃度に任意の定数を加減乗除することで得られるものでもよい。
(A)採取した血液サンプルを遠心することにより血液から血漿を分離した。全ての血漿サンプルは、アミノ酸濃度の測定時まで-80℃で凍結保存した。アミノ酸濃度測定時には、アセトニトリルを添加し除蛋白処理を行った後、標識試薬(3-アミノピリジル-N-ヒドロキシスクシンイミジルカルバメート)を用いてプレカラム誘導体化を行い、そして、液体クロマトグラフ質量分析計(LC/MS)によりアミノ酸濃度を分析した(国際公開第2003/069328号、国際公開第2005/116629号を参照)。
(B)採取した血液サンプルを遠心することにより血液から血漿を分離した。全ての血漿サンプルは、アミノ酸濃度の測定時まで-80℃で凍結保存した。アミノ酸濃度測定時には、スルホサリチル酸を添加し除蛋白処理を行った後、ニンヒドリン試薬を用いたポストカラム誘導体化法を原理としたアミノ酸分析計によりアミノ酸濃度を分析した。
In addition, the amino acid concentration data used in this processing is a specialist who uses blood (including plasma, serum, etc.) collected in advance from an individual such as an animal or human, for example, using a measurement method such as (A) or (B) below. Is related to the concentration value of amino acids obtained by analysis or independent analysis. Here, the unit of amino acid concentration may be obtained by, for example, molar concentration, weight concentration, or by adding / subtracting / subtracting an arbitrary constant to / from these concentrations.
(A) Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at −80 ° C. until the measurement of amino acid concentration. For amino acid concentration measurement, acetonitrile was added to remove protein, followed by precolumn derivatization using a labeling reagent (3-aminopyridyl-N-hydroxysuccinimidyl carbamate), and liquid chromatography mass spectrometry The amino acid concentration was analyzed by a total (LC / MS) (see International Publication No. 2003/069328 and International Publication No. 2005/116629).
(B) Plasma was separated from blood by centrifuging the collected blood sample. All plasma samples were stored frozen at −80 ° C. until the measurement of amino acid concentration. When measuring the amino acid concentration, sulfosalicylic acid was added to remove the protein, and then the amino acid concentration was analyzed by an amino acid analyzer based on the post-column derivatization method using a ninhydrin reagent.
 まず、Webブラウザ211を表示した画面上で利用者が入力装置250を介して癌免疫療法評価装置100が提供するWebサイトのアドレス(URLなど)を指定すると、クライアント装置200は癌免疫療法評価装置100へアクセスする。具体的には、利用者がクライアント装置200のWebブラウザ211の画面更新を指示すると、Webブラウザ211は、癌免疫療法評価装置100が提供するWebサイトのアドレスを所定の通信規約で癌免疫療法評価装置100へ送信することで、アミノ酸濃度データ送信画面に対応するWebページの送信要求を、当該アドレスに基づくルーティングで癌免疫療法評価装置100へ行う。 First, when a user designates an address (such as URL) of a Web site provided by the cancer immunotherapy evaluation apparatus 100 via the input device 250 on the screen displaying the Web browser 211, the client apparatus 200 causes the cancer immunotherapy evaluation apparatus to be displayed. 100 is accessed. Specifically, when the user instructs to update the screen of the Web browser 211 of the client device 200, the Web browser 211 uses the predetermined communication protocol to evaluate the cancer immunotherapy evaluation according to a predetermined communication protocol. By transmitting to the apparatus 100, a transmission request for a Web page corresponding to the amino acid concentration data transmission screen is made to the cancer immunotherapy evaluation apparatus 100 by routing based on the address.
 つぎに、癌免疫療法評価装置100は、要求解釈部102aで、クライアント装置200からの送信を受け、当該送信の内容を解析し、解析結果に応じて制御部102の各部に処理を移す。具体的には、送信の内容がアミノ酸濃度データ送信画面に対応するWebページの送信要求であった場合、癌免疫療法評価装置100は、主として閲覧処理部102bで、記憶部106の所定の記憶領域に格納されている当該Webページを表示するためのWebデータを取得し、取得したWebデータをクライアント装置200へ送信する。より具体的には、利用者からアミノ酸濃度データ送信画面に対応するWebページの送信要求があった場合、癌免疫療法評価装置100は、まず、制御部102で、利用者IDや利用者パスワードの入力を利用者に対して求める。そして、利用者IDやパスワードが入力されると、癌免疫療法評価装置100は、認証処理部102cで、入力された利用者IDやパスワードと利用者情報ファイル106aに格納されている利用者IDや利用者パスワードとの認証判断を行う。そして、癌免疫療法評価装置100は、認証可の場合にのみ、閲覧処理部102bで、アミノ酸濃度データ送信画面に対応するWebページを表示するためのWebデータをクライアント装置200へ送信する。なお、クライアント装置200の特定は、クライアント装置200から送信要求と共に送信されたIPアドレスで行う。 Next, the cancer immunotherapy evaluation device 100 receives the transmission from the client device 200 at the request interpretation unit 102a, analyzes the content of the transmission, and moves the processing to each unit of the control unit 102 according to the analysis result. Specifically, when the content of the transmission is a transmission request for a Web page corresponding to the amino acid concentration data transmission screen, the cancer immunotherapy evaluation apparatus 100 is a predetermined storage area of the storage unit 106 mainly in the browsing processing unit 102b. Web data for displaying the Web page stored in is acquired, and the acquired Web data is transmitted to the client device 200. More specifically, when there is a transmission request for a Web page corresponding to the amino acid concentration data transmission screen from the user, the cancer immunotherapy evaluation device 100 first uses the control unit 102 to check the user ID and the user password. Ask the user for input. Then, when the user ID and password are input, the cancer immunotherapy evaluation device 100 causes the authentication processing unit 102c to input the input user ID and password and the user ID stored in the user information file 106a. Make authentication with user password. The cancer immunotherapy evaluation apparatus 100 transmits Web data for displaying a Web page corresponding to the amino acid concentration data transmission screen to the client apparatus 200 by the browsing processing unit 102b only when authentication is possible. The client device 200 is identified by the IP address transmitted from the client device 200 together with the transmission request.
 つぎに、クライアント装置200は、癌免疫療法評価装置100から送信されたWebデータ(アミノ酸濃度データ送信画面に対応するWebページを表示するためのもの)を受信部213で受信し、受信したWebデータをWebブラウザ211で解釈し、モニタ261にアミノ酸濃度データ送信画面を表示する。 Next, the client device 200 receives the Web data (for displaying a Web page corresponding to the amino acid concentration data transmission screen) transmitted from the cancer immunotherapy evaluation device 100 by the receiving unit 213, and receives the received Web data. Is interpreted by the Web browser 211, and the amino acid concentration data transmission screen is displayed on the monitor 261.
 つぎに、モニタ261に表示されたアミノ酸濃度データ送信画面に対し利用者が入力装置250を介して個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データを入力・選択すると、クライアント装置200は、送信部214で、入力情報や選択事項を特定するための識別子を癌免疫療法評価装置100へ送信することで、個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データを癌免疫療法評価装置100へ送信する(ステップSA21)。なお、ステップSA21におけるアミノ酸濃度データの送信は、FTP等の既存のファイル転送技術等により実現してもよい。 Next, when the user inputs / selects the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data of the individual via the input device 250 on the amino acid concentration data transmission screen displayed on the monitor 261, the client device 200 The transmitting unit 214 transmits input information and an identifier for specifying selection items to the cancer immunotherapy evaluation apparatus 100, so that the amino acid concentration data before starting treatment and the amino acid concentration data after starting treatment are evaluated for cancer immunotherapy. It transmits to the apparatus 100 (step SA21). The transmission of amino acid concentration data in step SA21 may be realized by an existing file transfer technique such as FTP.
 つぎに、癌免疫療法評価装置100は、要求解釈部102aで、クライアント装置200から送信された識別子を解釈することによりクライアント装置200の要求内容を解釈し、癌免疫療法による治療が有効であるか否かの判別用の多変量判別式の送信要求をデータベース装置400へ行う。 Next, the cancer immunotherapy evaluation device 100 interprets the request contents of the client device 200 by interpreting the identifier transmitted from the client device 200 by the request interpretation unit 102a, and whether the cancer immunotherapy treatment is effective. A request for transmission of a multivariate discriminant for determining whether or not is sent to the database apparatus 400.
 つぎに、データベース装置400は、要求解釈部402aで、癌免疫療法評価装置100からの送信要求を解釈し、記憶部406の所定の記憶領域に格納した多変量判別式(例えばアップデートされた最新のもの)を癌免疫療法評価装置100へ送信する(ステップSA22)。具体的には、ステップSA22では、Val,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式を癌免疫療法評価装置100へ送信する。 Next, the database apparatus 400 interprets the transmission request from the cancer immunotherapy evaluation apparatus 100 by the request interpretation unit 402a and stores the multivariate discriminant (for example, the updated latest data) stored in a predetermined storage area of the storage unit 406. Is transmitted to the cancer immunotherapy evaluation apparatus 100 (step SA22). Specifically, in step SA22, at least one of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, and Pro. A multivariate discriminant including one as a variable is transmitted to the cancer immunotherapy evaluation apparatus 100.
 つぎに、癌免疫療法評価装置100は、受信部102fで、クライアント装置200から送信された個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データと、データベース装置400から送信された多変量判別式を受信し、受信した治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データをアミノ酸濃度データファイル106bの所定の記憶領域に格納すると共に、受信した多変量判別式を多変量判別式ファイル106e4の所定の記憶領域に格納する(ステップSA23)。 Next, the cancer immunotherapy evaluation apparatus 100 uses the receiving unit 102f to determine the pre-treatment amino acid concentration data and post-treatment amino acid concentration data transmitted from the client device 200, and the multivariate discrimination transmitted from the database device 400. The received pre-treatment amino acid concentration data and post-treatment amino acid concentration data are stored in a predetermined storage area of the amino acid concentration data file 106b, and the received multivariate discriminant is stored in the multivariate discriminant file 106e4. The data is stored in a predetermined storage area (step SA23).
 つぎに、癌免疫療法評価装置100は、制御部102で、ステップSA23で受信した個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データから欠損値や外れ値などのデータを除去する(ステップSA24)。 Next, in the cancer immunotherapy evaluation apparatus 100, the controller 102 removes data such as missing values and outliers from the pre-treatment amino acid concentration data and post-treatment amino acid concentration data of the individual received in step SA23 (step S23). SA24).
 つぎに、癌免疫療法評価装置100は、判別値算出部102iで、ステップSA24で欠損値や外れ値などのデータが除去された個体の治療開始前アミノ酸濃度データおよび治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの濃度値、およびステップSA23で受信したVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを変数として含む多変量判別式に基づいて、1つまたは複数の判別値(例えば、治療開始前アミノ酸濃度データを多変量判別式に代入して得られる判別値(治療開始前判別値)、治療開始後アミノ酸濃度データを多変量判別式に代入して得られる判別値(治療開始後判別値)、または、治療開始前アミノ酸濃度データと治療開始後アミノ酸濃度データとの差分または比などに関する変化量データを多変量判別式に代入して得られる判別値(治療開始前後判別値)、など)を算出する(ステップSA25)。 Next, the cancer immunotherapy evaluation apparatus 100 includes the discriminant value calculation unit 102i in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data of the individual from which data such as a missing value and an outlier have been removed in step SA24. At least one concentration value of Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro, and in step SA23 Multivariate including at least one of received Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Lys, Arg, Gly, Cys2, Pro as a variable Based on the discriminant, one or more discriminant values (eg, pre-treatment start Discriminant value obtained by substituting noic acid concentration data into a multivariate discriminant (discriminant value before treatment start), discriminant value obtained by substituting amino acid concentration data after treatment start into a multivariate discriminant (discriminant value after treatment start) ), Or discriminant values obtained by substituting change data related to the difference or ratio between the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data into the multivariate discriminant (discrimination value before and after treatment start, etc.) Is calculated (step SA25).
 つぎに、癌免疫療法評価装置100は、判別値基準判別部102j1で、ステップSA25で算出した判別値と予め設定された閾値とを比較することで、個体に対して癌免疫療法による治療が有効であるか否かを判別し、その判別結果を評価結果ファイル106gの所定の記憶領域に格納する(ステップSA26)。 Next, the cancer immunotherapy evaluation apparatus 100 compares the discriminant value calculated in step SA25 with a preset threshold value in the discriminant value criterion discriminating unit 102j1, so that treatment by cancer immunotherapy is effective for the individual. And the determination result is stored in a predetermined storage area of the evaluation result file 106g (step SA26).
 つぎに、癌免疫療法評価装置100は、送信部102mで、ステップSA26で得た判別結果(ステップSA25で算出した判別値を含めてもよい)を、アミノ酸濃度データの送信元のクライアント装置200とデータベース装置400とへ送信する(ステップSA27)。具体的には、まず、癌免疫療法評価装置100は、Webページ生成部102eで、判別結果を表示するためのWebページを作成し、作成したWebページに対応するWebデータを記憶部106の所定の記憶領域に格納する。ついで、利用者がクライアント装置200のWebブラウザ211に入力装置250を介して所定のURLを入力し上述した認証を経た後、クライアント装置200は、当該Webページの閲覧要求を癌免疫療法評価装置100へ送信する。ついで、癌免疫療法評価装置100は、閲覧処理部102bで、クライアント装置200から送信された閲覧要求を解釈し、判別結果を表示するためのWebページに対応するWebデータを記憶部106の所定の記憶領域から読み出す。そして、癌免疫療法評価装置100は、送信部102mで、読み出したWebデータをクライアント装置200へ送信すると共に、当該Webデータ又は判別結果をデータベース装置400へ送信する。 Next, the cancer immunotherapy evaluation apparatus 100 uses the transmission unit 102m to send the determination result obtained in step SA26 (which may include the determination value calculated in step SA25) to the client apparatus 200 that is the transmission source of amino acid concentration data. The data is transmitted to the database device 400 (step SA27). Specifically, first, the cancer immunotherapy evaluation apparatus 100 creates a web page for displaying the discrimination result in the web page generation unit 102e, and stores Web data corresponding to the created web page in the storage unit 106. Stored in the storage area. Next, after the user inputs a predetermined URL to the Web browser 211 of the client device 200 via the input device 250 and performs the above-described authentication, the client device 200 issues a request for browsing the Web page to the cancer immunotherapy evaluation device 100. Send to. Next, in the cancer immunotherapy evaluation device 100, the browsing processing unit 102b interprets the browsing request transmitted from the client device 200, and stores Web data corresponding to the Web page for displaying the determination result in the storage unit 106. Read from storage area. Then, the cancer immunotherapy evaluation device 100 transmits the read Web data to the client device 200 and transmits the Web data or the determination result to the database device 400 by the transmission unit 102m.
 ここで、ステップSA27において、癌免疫療法評価装置100は、制御部102で、判別結果を電子メールで利用者のクライアント装置200へ通知してもよい。具体的には、まず、癌免疫療法評価装置100は、電子メール生成部102dで、利用者IDなどを基にして利用者情報ファイル106aに格納されている利用者情報を送信タイミングに従って参照し、利用者の電子メールアドレスを取得する。ついで、癌免疫療法評価装置100は、電子メール生成部102dで、取得した電子メールアドレスを宛て先とし利用者の氏名および判別結果を含む電子メールに関するデータを生成する。ついで、癌免疫療法評価装置100は、送信部102mで、生成した当該データを利用者のクライアント装置200へ送信する。 Here, in step SA27, the cancer immunotherapy evaluation apparatus 100 may notify the user client apparatus 200 of the determination result by e-mail at the control unit 102. Specifically, the cancer immunotherapy evaluation apparatus 100 first refers to the user information stored in the user information file 106a based on the user ID or the like in the e-mail generation unit 102d according to the transmission timing, Get the user's email address. Next, the cancer immunotherapy evaluation apparatus 100 uses the e-mail generation unit 102d to generate data related to the e-mail including the name and discrimination result of the user with the acquired e-mail address as the destination. Next, the cancer immunotherapy evaluation apparatus 100 transmits the generated data to the user client apparatus 200 by the transmission unit 102m.
 また、ステップSA27において、癌免疫療法評価装置100は、FTP等の既存のファイル転送技術等で、判別結果を利用者のクライアント装置200へ送信してもよい。 In step SA27, the cancer immunotherapy evaluation apparatus 100 may transmit the determination result to the user client apparatus 200 using an existing file transfer technique such as FTP.
 図21の説明に戻り、データベース装置400は、制御部402で、癌免疫療法評価装置100から送信された判別結果またはWebデータを受信し、受信した判別結果またはWebデータを記憶部406の所定の記憶領域に保存(蓄積)する(ステップSA28)。 Returning to the description of FIG. 21, in the database device 400, the control unit 402 receives the discrimination result or Web data transmitted from the cancer immunotherapy evaluation device 100, and stores the received discrimination result or Web data in a predetermined unit of the storage unit 406. Save (accumulate) in the storage area (step SA28).
 また、クライアント装置200は、受信部213で、癌免疫療法評価装置100から送信されたWebデータを受信し、受信したWebデータをWebブラウザ211で解釈し、個体の判別結果が記されたWebページの画面をモニタ261に表示する(ステップSA29)。なお、判別結果が癌免疫療法評価装置100から電子メールで送信された場合には、クライアント装置200は、電子メーラ212の公知の機能で、癌免疫療法評価装置100から送信された電子メールを任意のタイミングで受信し、受信した電子メールをモニタ261に表示する。 In addition, the client device 200 receives the Web data transmitted from the cancer immunotherapy evaluation device 100 by the receiving unit 213, interprets the received Web data by the Web browser 211, and the Web page on which the individual determination result is written. Is displayed on the monitor 261 (step SA29). When the discrimination result is transmitted from the cancer immunotherapy evaluation apparatus 100 by e-mail, the client apparatus 200 arbitrarily selects the e-mail transmitted from the cancer immunotherapy evaluation apparatus 100 by a known function of the e-mailer 212. The received e-mail is displayed on the monitor 261.
 以上により、利用者は、モニタ261に表示されたWebページを閲覧することで、判別結果を確認することができる。なお、利用者は、モニタ261に表示されたWebページの表示内容をプリンタ262で印刷してもよい。 As described above, the user can check the determination result by browsing the Web page displayed on the monitor 261. Note that the user may print the display content of the Web page displayed on the monitor 261 with the printer 262.
 また、判別結果が癌免疫療法評価装置100から電子メールで送信された場合には、利用者は、モニタ261に表示された電子メールを閲覧することで、判別結果を確認することができる。利用者は、モニタ261に表示された電子メールの表示内容をプリンタ262で印刷してもよい。 In addition, when the discrimination result is transmitted from the cancer immunotherapy evaluation apparatus 100 by e-mail, the user can check the discrimination result by browsing the e-mail displayed on the monitor 261. The user may print the content of the e-mail displayed on the monitor 261 with the printer 262.
 これにて、癌免疫療法評価サービス処理の説明を終了する。 This completes the explanation of the cancer immunotherapy evaluation service process.
[2-4.他の実施形態]
 本発明にかかる癌免疫療法評価装置、癌免疫療法評価方法、癌免疫療法評価プログラム、記録媒体、癌免疫療法評価システム、および情報通信端末装置は、上述した第2実施形態以外にも、特許請求の範囲に記載した技術的思想の範囲内において種々の異なる実施形態にて実施されてよいものである。
[2-4. Other Embodiments]
A cancer immunotherapy evaluation device, a cancer immunotherapy evaluation method, a cancer immunotherapy evaluation program, a recording medium, a cancer immunotherapy evaluation system, and an information communication terminal device according to the present invention are claimed in addition to the second embodiment described above. The present invention may be implemented in various different embodiments within the scope of the technical idea described in the above.
 また、第2実施形態において説明した各処理のうち、自動的に行われるものとして説明した処理の全部または一部を手動的に行うこともでき、あるいは、手動的に行われるものとして説明した処理の全部または一部を公知の方法で自動的に行うこともできる。 In addition, among the processes described in the second embodiment, all or part of the processes described as being performed automatically can be performed manually, or the processes described as being performed manually All or a part of the above can be automatically performed by a known method.
 このほか、上記文献中や図面中で示した処理手順、制御手順、具体的名称、各処理の登録データや検索条件等のパラメータを含む情報、画面例、データベース構成については、特記する場合を除いて任意に変更することができる。 In addition, unless otherwise specified, the processing procedures, control procedures, specific names, information including registration data for each processing, parameters such as search conditions, screen examples, and database configurations shown in the above documents and drawings Can be changed arbitrarily.
 また、癌免疫療法評価装置100に関して、図示の各構成要素は機能概念的なものであり、必ずしも物理的に図示の如く構成されていることを要しない。 In addition, regarding the cancer immunotherapy evaluation apparatus 100, each illustrated component is functionally conceptual and does not necessarily need to be physically configured as illustrated.
 例えば、癌免疫療法評価装置100が備える処理機能、特に制御部102にて行われる各処理機能については、その全部または任意の一部を、CPU(Central Processing Unit)および当該CPUにて解釈実行されるプログラムにて実現してもよく、また、ワイヤードロジックによるハードウェアとして実現してもよい。尚、プログラムは、情報処理装置に本発明にかかる癌免疫療法評価方法を実行させるためのプログラム化された命令を含む一時的でないコンピュータ読み取り可能な記録媒体に記録されており、必要に応じて癌免疫療法評価装置100に機械的に読み取られる。すなわち、ROMまたはHDDなどの記憶部106などには、OS(Operating System)と協働してCPUに命令を与え、各種処理を行うためのコンピュータプログラムが記録されている。このコンピュータプログラムは、RAMにロードされることによって実行され、CPUと協働して制御部を構成する。 For example, the processing functions provided in the cancer immunotherapy evaluation apparatus 100, in particular, each processing function performed by the control unit 102, is interpreted and executed by a CPU (Central Processing Unit) and the CPU. It may be realized by a program or hardware based on wired logic. The program is recorded on a non-transitory computer-readable recording medium including programmed instructions for causing the information processing apparatus to execute the cancer immunotherapy evaluation method according to the present invention. It is mechanically read by the immunotherapy evaluation apparatus 100. That is, in the storage unit 106 such as a ROM or an HDD, computer programs for performing various processes by giving instructions to the CPU in cooperation with an OS (Operating System) are recorded. This computer program is executed by being loaded into the RAM, and constitutes a control unit in cooperation with the CPU.
 また、このコンピュータプログラムは、癌免疫療法評価装置100に対して任意のネットワークを介して接続されたアプリケーションプログラムサーバに記憶されていてもよく、必要に応じてその全部または一部をダウンロードすることも可能である。 The computer program may be stored in an application program server connected to the cancer immunotherapy evaluation apparatus 100 via an arbitrary network, and may be downloaded in whole or in part as necessary. Is possible.
 また、本発明にかかる癌免疫療法評価プログラムを、一時的でないコンピュータ読み取り可能な記録媒体に格納してもよく、また、プログラム製品として構成することもできる。ここで、この「記録媒体」とは、メモリーカード、USBメモリ、SDカード、フレキシブルディスク、光磁気ディスク、ROM、EPROM、EEPROM(登録商標)、CD-ROM、MO、DVD、および、Blu-ray(登録商標) Disc等の任意の「可搬用の物理媒体」を含むものとする。 Further, the cancer immunotherapy evaluation program according to the present invention may be stored in a non-temporary computer-readable recording medium, or may be configured as a program product. Here, the “recording medium” means a memory card, USB memory, SD card, flexible disk, magneto-optical disk, ROM, EPROM, EEPROM (registered trademark), CD-ROM, MO, DVD, and Blu-ray. (Registered trademark) It shall include any “portable physical medium” such as Disc.
 また、「プログラム」とは、任意の言語または記述方法にて記述されたデータ処理方法であり、ソースコードまたはバイナリコード等の形式を問わない。なお、「プログラム」は必ずしも単一的に構成されるものに限られず、複数のモジュールやライブラリとして分散構成されるものや、OS(Operating System)に代表される別個のプログラムと協働してその機能を達成するものをも含む。なお、実施形態に示した各装置において記録媒体を読み取るための具体的な構成および読み取り手順ならびに読み取り後のインストール手順等については、周知の構成や手順を用いることができる。 Also, the “program” is a data processing method described in an arbitrary language or description method, and may be in the form of source code or binary code. Note that the “program” is not necessarily limited to a single configuration, but is distributed in the form of a plurality of modules and libraries, or in cooperation with a separate program typified by an OS (Operating System). Including those that achieve the function. In addition, a well-known structure and procedure can be used about the specific structure and reading procedure for reading a recording medium in each apparatus shown to embodiment, the installation procedure after reading, etc.
 記憶部106に格納される各種のデータベース等は、RAM、ROM等のメモリ装置、ハードディスク等の固定ディスク装置、フレキシブルディスク、および、光ディスク等のストレージ手段であり、各種処理やウェブサイト提供に用いる各種のプログラム、テーブル、データベース、および、ウェブページ用ファイル等を格納する。 Various databases and the like stored in the storage unit 106 are storage devices such as a memory device such as a RAM and a ROM, a fixed disk device such as a hard disk, a flexible disk, and an optical disk. Programs, tables, databases, web page files, and the like.
 また、癌免疫療法評価装置100は、既知のパーソナルコンピュータまたはワークステーション等の情報処理装置として構成してもよく、また、任意の周辺装置が接続された当該情報処理装置として構成してもよい。また、癌免疫療法評価装置100は、当該情報処理装置に本発明の癌免疫療法評価方法を実現させるソフトウェア(プログラムまたはデータ等を含む)を実装することにより実現してもよい。 Further, the cancer immunotherapy evaluation apparatus 100 may be configured as an information processing apparatus such as a known personal computer or workstation, or may be configured as the information processing apparatus connected to an arbitrary peripheral device. The cancer immunotherapy evaluation apparatus 100 may be realized by installing software (including a program or data) that realizes the cancer immunotherapy evaluation method of the present invention in the information processing apparatus.
 更に、装置の分散・統合の具体的形態は図示するものに限られず、その全部または一部を、各種の付加等に応じてまたは機能負荷に応じて、任意の単位で機能的または物理的に分散・統合して構成することができる。すなわち、上述した実施形態を任意に組み合わせて実施してもよく、実施形態を選択的に実施してもよい。 Furthermore, the specific form of distribution / integration of the devices is not limited to that shown in the figure, and all or a part of them may be functionally or physically in arbitrary units according to various additions or according to functional loads. It can be configured to be distributed and integrated. That is, the above-described embodiments may be arbitrarily combined and may be selectively implemented.
 最後に、癌免疫療法評価装置100で行う多変量判別式作成処理の一例について図22を参照して詳細に説明する。なお、ここで説明する処理はあくまでも一例であり、多変量判別式の作成方法はこれに限定されない。図22は多変量判別式作成処理の一例を示すフローチャートである。なお、当該多変量判別式作成処理は、癌状態情報を管理するデータベース装置400で行ってもよい。 Finally, an example of multivariate discriminant creation processing performed by the cancer immunotherapy evaluation apparatus 100 will be described in detail with reference to FIG. Note that the processing described here is merely an example, and the method of creating the multivariate discriminant is not limited to this. FIG. 22 is a flowchart illustrating an example of multivariate discriminant creation processing. The multivariate discriminant creation process may be performed by the database apparatus 400 that manages cancer state information.
 なお、本説明では、癌免疫療法評価装置100は、データベース装置400から事前に取得した癌状態情報を、癌状態情報ファイル106cの所定の記憶領域に格納しているものとする。また、癌免疫療法評価装置100は、癌状態情報指定部102gで事前に指定した癌状態指標データ(例えば、癌の状態を表す指標に関するデータ(例えば、指標に基づいて得られる治療効果の有無など)、または、癌の状態の指標の変化量に関するデータ(例えば、腫瘍の大きさの差分値など)、など)およびアミノ酸濃度データ(例えば、アミノ酸濃度に関するデータ、または、アミノ酸濃度の変化量に関するデータ、など)を含む癌状態情報を、指定癌状態情報ファイル106dの所定の記憶領域に格納しているものとする。 In this description, it is assumed that the cancer immunotherapy evaluation device 100 stores cancer state information acquired in advance from the database device 400 in a predetermined storage area of the cancer state information file 106c. In addition, the cancer immunotherapy evaluation apparatus 100 uses the cancer state index data specified in advance by the cancer state information specifying unit 102g (for example, data related to an index indicating a cancer state (for example, presence or absence of a therapeutic effect obtained based on the index). ), Or data relating to the amount of change in an indicator of cancer status (for example, a difference in tumor size) and amino acid concentration data (eg, data relating to amino acid concentration or data relating to the amount of change in amino acid concentration) , Etc.) is stored in a predetermined storage area of the designated cancer state information file 106d.
 まず、多変量判別式作成部102hは、候補多変量判別式作成部102h1で、指定癌状態情報ファイル106dの所定の記憶領域に格納されている癌状態情報から所定の式作成手法に基づいて候補多変量判別式を作成し、作成した候補多変量判別式を候補多変量判別式ファイル106e1の所定の記憶領域に格納する(ステップSB21)。具体的には、まず、多変量判別式作成部102hは、候補多変量判別式作成部102h1で、複数の異なる式作成手法(主成分分析や判別分析、サポートベクターマシン、重回帰分析、ロジスティック回帰分析、k-means法、クラスター解析、決定木などの多変量解析に関するものを含む。)の中から所望のものを1つ選択し、選択した式作成手法に基づいて、作成する候補多変量判別式の形(式の形)を決定する。つぎに、多変量判別式作成部102hは、候補多変量判別式作成部102h1で、癌状態情報に基づいて、選択した式選択手法に対応する種々(例えば平均や分散など)の計算を実行する。つぎに、多変量判別式作成部102hは、候補多変量判別式作成部102h1で、計算結果および決定した候補多変量判別式のパラメータを決定する。これにより、選択した式作成手法に基づいて候補多変量判別式が作成される。なお、複数の異なる式作成手法を併用して候補多変量判別式を同時並行(並列)的に作成する場合は、選択した式作成手法ごとに上記の処理を並行して実行すればよい。また、複数の異なる式作成手法を併用して候補多変量判別式を直列的に作成する場合は、例えば、主成分分析を行って作成した候補多変量判別式を利用して癌状態情報を変換し、変換した癌状態情報に対して判別分析を行うことで候補多変量判別式を作成してもよい。 First, the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1 based on a predetermined formula creation method from cancer state information stored in a predetermined storage area of the designated cancer state information file 106d. A multivariate discriminant is created, and the created candidate multivariate discriminant is stored in a predetermined storage area of the candidate multivariate discriminant file 106e1 (step SB21). Specifically, first, the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1, and a plurality of different formula creation methods (principal component analysis, discriminant analysis, support vector machine, multiple regression analysis, logistic regression) Analysis, k-means method, cluster analysis, decision tree, etc. related to multivariate analysis.) Select a desired one from among them, and create candidate multivariate discrimination based on the selected formula creation method Determine the form of the expression (form of the expression). Next, the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-preparing part 102h1, and executes various calculations (for example, average and variance) corresponding to the selected formula selection method based on the cancer state information. . Next, the multivariate discriminant-preparing part 102h determines the calculation result and parameters of the determined candidate multivariate discriminant-expression in the candidate multivariate discriminant-preparing part 102h1. Thereby, a candidate multivariate discriminant is created based on the selected formula creation method. In addition, when a candidate multivariate discriminant is created in parallel and in parallel by using a plurality of different formula creation methods, the above-described processing may be executed in parallel for each selected formula creation method. In addition, when creating a candidate multivariate discriminant serially using a combination of multiple different formula creation methods, for example, transform cancer status information using a candidate multivariate discriminant created by performing principal component analysis. Then, the candidate multivariate discriminant may be created by performing discriminant analysis on the converted cancer state information.
 つぎに、多変量判別式作成部102hは、候補多変量判別式検証部102h2で、ステップSB21で作成した候補多変量判別式を所定の検証手法に基づいて検証(相互検証)し、検証結果を検証結果ファイル106e2の所定の記憶領域に格納する(ステップSB22)。具体的には、多変量判別式作成部102hは、候補多変量判別式検証部102h2で、指定癌状態情報ファイル106dの所定の記憶領域に格納されている癌状態情報に基づいて候補多変量判別式を検証する際に用いる検証用データを作成し、作成した検証用データに基づいて候補多変量判別式を検証する。なお、ステップSB21で複数の異なる式作成手法を併用して候補多変量判別式を複数作成した場合には、多変量判別式作成部102hは、候補多変量判別式検証部102h2で、各式作成手法に対応する候補多変量判別式ごとに所定の検証手法に基づいて検証する。ここで、ステップSB22において、ランダムサンプリング法、ブートストラップ法、ホールドアウト法、N-フォールド法またはリーブワンアウト法などのうち少なくとも1つに基づいて候補多変量判別式の判別率や感度、特異度、情報量基準、ROC_AUC(受信者特性曲線の曲線下面積)などのうち少なくとも1つに関して検証してもよい。これにより、癌状態情報や診断条件を考慮した予測性または頑健性の高い候補指標式を選択することができる。 Next, the multivariate discriminant-preparing part 102h verifies (mutually verifies) the candidate multivariate discriminant created in step SB21 with the candidate multivariate discriminant-verifying part 102h2, and verifies the verification result. The result is stored in a predetermined storage area of the verification result file 106e2 (step SB22). Specifically, the multivariate discriminant-preparing part 102h is a candidate multivariate discriminant-verifying part 102h2, based on the cancer state information stored in a predetermined storage area of the designated cancer state information file 106d. The verification data used when verifying the formula is created, and the candidate multivariate discriminant is verified based on the created verification data. If a plurality of candidate multivariate discriminants are created in combination with a plurality of different formula creation methods in step SB21, the multivariate discriminant creation unit 102h creates each formula in the candidate multivariate discriminant verification unit 102h2. Each candidate multivariate discriminant corresponding to the method is verified based on a predetermined verification method. Here, in step SB22, the discrimination rate, sensitivity, and specificity of the candidate multivariate discriminant based on at least one of the random sampling method, the bootstrap method, the holdout method, the N-fold method, the leave one-out method, etc. , Information criteria, ROC_AUC (area under the receiver characteristic curve), etc. Thereby, it is possible to select a candidate index formula having high predictability or robustness in consideration of cancer state information and diagnosis conditions.
 つぎに、多変量判別式作成部102hは、変数選択部102h3で、所定の変数選択手法に基づいて、候補多変量判別式の変数を選択することで、候補多変量判別式を作成する際に用いる癌状態情報に含まれるアミノ酸濃度データの組み合わせを選択し、選択したアミノ酸濃度データの組み合わせを含む癌状態情報を選択癌状態情報ファイル106e3の所定の記憶領域に格納する(ステップSB23)。なお、ステップSB21で複数の異なる式作成手法を併用して候補多変量判別式を複数作成し、ステップSB22で各式作成手法に対応する候補多変量判別式ごとに所定の検証手法に基づいて検証した場合には、ステップSB23において、多変量判別式作成部102hは、変数選択部102h3で、候補多変量判別式ごとに所定の変数選択手法に基づいて候補多変量判別式の変数を選択してもよい。ここで、ステップSB23において、検証結果からステップワイズ法、ベストパス法、近傍探索法、遺伝的アルゴリズムのうち少なくとも1つに基づいて候補多変量判別式の変数を選択してもよい。なお、ベストパス法とは、候補多変量判別式に含まれる変数を1つずつ順次減らしていき、候補多変量判別式が与える評価指標を最適化することで変数を選択する方法である。また、ステップSB23において、多変量判別式作成部102hは、変数選択部102h3で、指定癌状態情報ファイル106dの所定の記憶領域に格納されている癌状態情報に基づいてアミノ酸濃度データの組み合わせを選択してもよい。 Next, when the multivariate discriminant-preparing part 102h creates a candidate multivariate discriminant by selecting a variable of the candidate multivariate discriminant based on a predetermined variable selection method in the variable selector 102h3. A combination of amino acid concentration data included in the cancer state information to be used is selected, and cancer state information including the selected combination of amino acid concentration data is stored in a predetermined storage area of the selected cancer state information file 106e3 (step SB23). In step SB21, a plurality of candidate multivariate discriminants are created in combination with a plurality of different formula creation methods, and in step SB22, each candidate multivariate discriminant corresponding to each formula creation method is verified based on a predetermined verification method. In this case, in step SB23, the multivariate discriminant-preparing part 102h selects a variable of the candidate multivariate discriminant based on a predetermined variable selection method for each candidate multivariate discriminant in the variable selector 102h3. Also good. Here, in step SB23, the variable of the candidate multivariate discriminant may be selected based on at least one of the stepwise method, the best path method, the neighborhood search method, and the genetic algorithm from the verification result. The best path method is a method of selecting variables by sequentially reducing the variables included in the candidate multivariate discriminant one by one and optimizing the evaluation index given by the candidate multivariate discriminant. In step SB23, the multivariate discriminant-preparing part 102h selects a combination of amino acid concentration data based on the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d by the variable selection part 102h3. May be.
 つぎに、多変量判別式作成部102hは、指定癌状態情報ファイル106dの所定の記憶領域に格納されている癌状態情報に含まれるアミノ酸濃度データの全ての組み合わせが終了したか否かを判定し、判定結果が「終了」であった場合(ステップSB24:Yes)には次のステップ(ステップSB25)へ進み、判定結果が「終了」でなかった場合(ステップSB24:No)にはステップSB21へ戻る。なお、多変量判別式作成部102hは、予め設定した回数が終了したか否かを判定し、判定結果が「終了」であった場合には(ステップSB24:Yes)次のステップ(ステップSB25)へ進み、判定結果が「終了」でなかった場合(ステップSB24:No)にはステップSB21へ戻ってもよい。また、多変量判別式作成部102hは、ステップSB23で選択したアミノ酸濃度データの組み合わせが、指定癌状態情報ファイル106dの所定の記憶領域に格納されている癌状態情報に含まれるアミノ酸濃度データの組み合わせまたは前回のステップSB23で選択したアミノ酸濃度データの組み合わせと同じであるか否かを判定し、判定結果が「同じ」であった場合(ステップSB24:Yes)には次のステップ(ステップSB25)へ進み、判定結果が「同じ」でなかった場合(ステップSB24:No)にはステップSB21へ戻ってもよい。また、多変量判別式作成部102hは、検証結果が具体的には各候補多変量判別式に関する評価値である場合には、当該評価値と各式作成手法に対応する所定の閾値との比較結果に基づいて、ステップSB25へ進むかステップSB21へ戻るかを判定してもよい。 Next, the multivariate discriminant-preparing part 102h determines whether or not all combinations of amino acid concentration data included in the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d have been completed. If the determination result is “end” (step SB24: Yes), the process proceeds to the next step (step SB25). If the determination result is not “end” (step SB24: No), the process proceeds to step SB21. Return. The multivariate discriminant-preparing part 102h determines whether or not the preset number of times has ended, and if the determination result is “end” (step SB24: Yes), the next step (step SB25). If the determination result is not “end” (step SB24: No), the process may return to step SB21. In addition, the multivariate discriminant-preparing part 102h has a combination of amino acid concentration data included in the cancer state information stored in the predetermined storage area of the designated cancer state information file 106d as the combination of the amino acid concentration data selected in step SB23. Alternatively, it is determined whether or not the combination of the amino acid concentration data selected in the previous step SB23 is the same, and when the determination result is “same” (step SB24: Yes), the process proceeds to the next step (step SB25). If the determination result is not “same” (step SB24: No), the process may return to step SB21. Further, when the verification result is specifically an evaluation value related to each candidate multivariate discriminant, the multivariate discriminant creation unit 102h compares the evaluation value with a predetermined threshold corresponding to each formula creation method. Based on the result, it may be determined whether to proceed to step SB25 or to return to step SB21.
 つぎに、多変量判別式作成部102hは、検証結果に基づいて、複数の候補多変量判別式の中から多変量判別式として採用する候補多変量判別式を選出することで多変量判別式を決定し、決定した多変量判別式(選出した候補多変量判別式)を多変量判別式ファイル106e4の所定の記憶領域に格納する(ステップSB25)。ここで、ステップSB25において、例えば、同じ式作成手法で作成した候補多変量判別式の中から最適なものを選出する場合と、すべての候補多変量判別式の中から最適なものを選出する場合とがある。 Next, the multivariate discriminant-preparing part 102h selects a multivariate discriminant by selecting a candidate multivariate discriminant to be adopted as a multivariate discriminant from a plurality of candidate multivariate discriminants based on the verification result. The determined multivariate discriminant (selected candidate multivariate discriminant) is stored in a predetermined storage area of the multivariate discriminant file 106e4 (step SB25). Here, in step SB25, for example, when the optimum one is selected from candidate multivariate discriminants created by the same formula creation method, and when the optimum one is selected from all candidate multivariate discriminants There is.
 これにて、多変量判別式作成処理の説明を終了する。 This completes the explanation of the multivariate discriminant creation process.
 C57BL/6J(WT)マウス10匹およびNudeマウス10匹に3LL細胞を皮下移植(1×10 cells/100μl)し、腫瘍サイズが100-150mmになった時点で低用量5FU(50mg/kg)の腹腔内投与を行なった。 Ten C57BL / 6J (WT) mice and 10 Nude mice were transplanted with 3LL cells subcutaneously (1 × 10 6 cells / 100 μl), and when the tumor size reached 100-150 mm 2 , a low dose of 5FU (50 mg / kg) was obtained. ) Was administered intraperitoneally.
 5FUの投与前(治療開始前,pre)と投与後6日目(治療開始後(治療中),post)で採血を行なった(図23)。 Blood was collected before administration of 5FU (before treatment start, pre) and 6 days after administration (after treatment start (during treatment), post) (FIG. 23).
 腫瘍サイズの測定は2もしくは3日に1回行った。腫瘍サイズの経日変化を図24および図25に示す。図24および図25に示すグラフの横軸は5FUの投与日をday0とした日数を表し、縦軸は腫瘍サイズ(mm)の平均値を表す。図24はWT(レスポンダー)、図25はNude(ノンレスポンダー)のデータである。図24および図25には、それぞれ治療あり群と治療なし群が示されている。 Tumor size was measured once every 2 or 3 days. The daily changes in tumor size are shown in FIGS. The horizontal axis of the graphs shown in FIG. 24 and FIG. 25 represents the number of days when the day of 5FU administration is day 0, and the vertical axis represents the average value of tumor size (mm 2 ). FIG. 24 shows WT (responder) data and FIG. 25 shows Nude (non-responder) data. 24 and 25 show a group with treatment and a group without treatment, respectively.
 WTでは5FU投与後に有意な腫瘍増殖抑制が認められたが、一方で、Nudeでは5FU投与後に腫瘍増殖抑制は認めなかった。この結果は、5FU投与によりMDSC(myeloid-derived suppressing cell)を除去するとT細胞が軸となる抗腫瘍応答が惹起されるが、T細胞の欠損しているNudeマウスでは抗腫瘍効果が得られないことに因る。 WT showed significant tumor growth inhibition after 5FU administration, while Nude did not show tumor growth inhibition after 5FU administration. This result shows that removal of MDSC (myloid-derived suppression cell) by 5FU induces an antitumor response centered on T cells, but antitumor effects are not obtained in Nude mice lacking T cells. It depends on.
 血漿中のアミノ酸濃度の測定を、上述した実施形態で説明した(A)の測定方法で行った。 Measurement of amino acid concentration in plasma was performed by the measurement method (A) described in the above embodiment.
 WTマウスの血漿中アミノ酸濃度のデータを図26および図27に示す。図26および図27において、横軸は治療開始前(pre)と治療開始後(post)とを表し、縦軸は各アミノ酸濃度(μM)の平均値を表す。Student’s t-testの結果、治療開始前に比べて治療開始後では、Val,Leu,Ile,His,Phe,Trp,Gln,Asp,Ornが有意に低下していた(*:p<0.05,***:p<0.001,****:p<0.0001)。 FIG. 26 and FIG. 27 show data on amino acid concentrations in plasma of WT mice. 26 and 27, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration (μM). As a result of Student's t-test, Val, Leu, Ile, His, Phe, Trp, Gln, Asp, and Orn significantly decreased after the start of treatment compared to before the start of treatment (*: p <0 .05, ***: p <0.001, ***: p <0.0001).
 Nudeマウスの血漿中アミノ酸濃度のデータを図28および図29に示す。図28および図29において、横軸は治療開始前(pre)と治療開始後(post)とを表し、縦軸は各アミノ酸濃度(μM)の平均値を表す。Student’s t-testの結果、治療開始前に比べて治療開始後では、Ala,Thr,Lys,Proが有意に増加していた(*:p<0.05,**:p<0.01)。 FIG. 28 and FIG. 29 show data on amino acid concentrations in plasma of Nude mice. 28 and 29, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration (μM). As a result of Student's t-test, Ala, Thr, Lys, and Pro were significantly increased after the start of treatment compared to before the start of treatment (*: p <0.05, **: p <0. 01).
 WT(レスポンダー)についての、治療開始前の各アミノ酸を100%としたときの治療開始後の各アミノ酸の分布を示すレーダーチャートを、図30に示す。Nude(ノンレスポンダー)についての、治療開始前の各アミノ酸を100%としたときの治療開始後の各アミノ酸の分布を示すレーダーチャートを、図31に示す。WTのアミノ酸プロファイル変化とNudeでのアミノ酸プロファイル変化は異なっており、抗腫瘍効果が得られる場合に特有の血漿中アミノ酸プロファイル変化が明らかとなった。WTの治療開始前後で有意差があったアミノ酸変数Val,Leu,Ile,His,Phe,Trp,Gln,Asp,Ornは癌免疫療法の効果の判別能を持つことが判明した。また、Nudeの治療開始前後で有意差があったアミノ酸変数Ala,Thr,Lys,Proについても癌免疫療法の効果判別能を持つことが判明した。 FIG. 30 shows a radar chart showing the distribution of each amino acid after the start of treatment, assuming that each amino acid before the start of treatment is 100% for WT (responder). FIG. 31 shows a radar chart showing the distribution of each amino acid after the start of treatment when Nude (non-responder) is 100% of each amino acid before the start of treatment. The amino acid profile change in WT and the amino acid profile change in Nude were different, and a characteristic amino acid profile change in plasma was clarified when an antitumor effect was obtained. The amino acid variables Val, Leu, Ile, His, Phe, Trp, Gln, Asp, and Orn that were significantly different before and after the start of WT treatment were found to have the ability to discriminate the effects of cancer immunotherapy. It was also found that the amino acid variables Ala, Thr, Lys, and Pro that had a significant difference before and after the start of Nude treatment also had the ability to discriminate cancer immunotherapy effects.
 さらに、WTマウスにおける各アミノ酸変数による治療開始前と治療開始後の2群判別の判別性能を、ROC曲線の曲線下面積(ROC_AUC)で評価した。その結果、アミノ酸変数Val,Leu,Ile,Lys,His,Phe,Trp,Gln,Asp,OrnについてAUCが0.7より大きい値を示した(図32および図33)。これにより、アミノ酸変数Val,Leu,Ile,Lys,His,Phe,Trp,Gln,Asp,Ornが、癌免疫療法の効果の判別能を持つことが判明した。 Furthermore, the discrimination performance of 2-group discrimination before and after the start of treatment with each amino acid variable in WT mice was evaluated by the area under the ROC curve (ROC_AUC). As a result, the AUC was greater than 0.7 for the amino acid variables Val, Leu, Ile, Lys, His, Phe, Trp, Gln, Asp, and Orn (FIGS. 32 and 33). As a result, the amino acid variables Val, Leu, Ile, Lys, His, Phe, Trp, Gln, Asp, and Orn were found to have the ability to discriminate the effects of cancer immunotherapy.
 実施例1と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post/pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 1, a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 国際公開第2004/052191号に記載の方法を用いて、多変量判別式としての分数式に含める変数(4個以下)の組み合わせを下記22種類のアミノ酸から探索し、そしてクロスバリデーションとしてブートストラップ法を採用して、癌免疫療法の効果の判別性能を最大化する分数式を探索した。
 ここで、22種類のアミノ酸は、Ala,Arg,Asn,Asp,Cit,Cys2,Gln,Glu,Gly,His,Ile,Leu,Lys,Met,Orn,Phe,Pro,Ser,Thr,Trp,Tyr,Valである。
Using the method described in International Publication No. 2004/052191, a combination of variables (4 or less) to be included in a fractional expression as a multivariate discriminant is searched from the following 22 types of amino acids, and the bootstrap method is used as cross validation Was used to search for a fractional expression that maximizes the ability to discriminate the effects of cancer immunotherapy.
Here, 22 kinds of amino acids are Ala, Arg, Asn, Asp, Cit, Cys2, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Orn, Phe, Pro, Ser, Thr, Trp, Tyr. , Val.
 ROC_AUCの値が1であるという良好な判別能を有する分数式の一覧を図34に示す。図34に含まれる式における変数の出現頻度を図35に示す。出現頻度が多い順に10位までのアミノ酸変数は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 FIG. 34 shows a list of fractional expressions having a good discrimination ability that the value of ROC_AUC is 1. FIG. 35 shows the frequency of appearance of variables in the formula included in FIG. The amino acid variables up to position 10 in the order of appearance frequency were Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, and His. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 多変量判別式としてのロジスティック回帰式に含める変数(4個以下)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (4 or less) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids, and adopt a random sampling method as cross-validation to maximize the discriminating performance of cancer immunotherapy effects We searched for logistic regression equations.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図36、図37および図38に示す。ここで、図36、図37および図38には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図36、図37および図38に含まれる式における変数の出現頻度を図39に示す。出現頻度が多い順に10位までのアミノ酸変数は、Trp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lysであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 36, 37, and 38 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC. Here, FIG. 36, FIG. 37, and FIG. 38 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 39 shows the appearance frequency of variables in the expressions included in FIGS. 36, 37, and 38. The amino acid variables up to position 10 in the order of appearance frequency were Trp, Thr, His, Arg, Ile, Pro, Phe, Met, Ala, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例1と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で差し引いた差分(post-pre)を計算し、当該差分に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 1, a difference (post-pre) was calculated by subtracting each plasma amino acid concentration after the start of treatment from each plasma amino acid concentration before the start of the treatment, and a change related to the difference. Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(4個以下)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (4 or less) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids, and adopt a random sampling method as cross-validation to maximize the discriminating performance of cancer immunotherapy effects We searched for logistic regression equations.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図40、図41および図42に示す。ここで、図40、図41および図42には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図40、図41および図42に含まれる式における変数の出現頻度を図43に示す。出現頻度が多い順に10位までのアミノ酸変数は、Trp,Asp,Ile,Thr,Arg,Pro,Ser,Leu,Met,Lysであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 40, FIG. 41 and FIG. 42 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC. Here, FIG. 40, FIG. 41, and FIG. 42 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 43 shows the appearance frequency of variables in the expressions included in FIGS. 40, 41, and 42. The amino acid variables up to position 10 in the order of appearance frequency were Trp, Asp, Ile, Thr, Arg, Pro, Ser, Leu, Met, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例1と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post/pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 1, a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(3~6個)の組み合わせを下記19種のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。
 ここで、19種類のアミノ酸は、Ala,Arg,Asn,Cit,Gln,Gly,His,Ile,Leu,Lys,Met,Orn,Phe,Pro,Ser,Thr,Trp,Tyr,Valである。
Search for combinations of variables (3 to 6) to be included in the logistic regression equation as a multivariate discriminant from the following 19 types of amino acids and adopt a random sampling method as cross-validation to determine the discriminating ability of cancer immunotherapy effects The logistic regression equation to maximize was searched.
Here, 19 kinds of amino acids are Ala, Arg, Asn, Cit, Gln, Gly, His, Ile, Leu, Lys, Met, Orn, Phe, Pro, Ser, Thr, Trp, Tyr, Val.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図44-48に示す。ここで、図44-48には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図44-48に含まれる式における変数の出現頻度を図49に示す。出現頻度が多い順に10位までのアミノ酸変数は、His,Thr,Lys,Phe,Arg,Ile,Met,Ser,Val,Asnであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 44-48 shows a list of logistic regression equations with equally good discriminating ability evaluated by the value of ROC_AUC. Here, FIGS. 44 to 48 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 49 shows the frequency of occurrence of variables in the expressions included in FIGS. The amino acid variables up to position 10 in the order of appearance frequency were His, Thr, Lys, Phe, Arg, Ile, Met, Ser, Val, Asn. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例1と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で差し引いた差分(post-pre)を計算し、当該差分に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 1, a difference (post-pre) was calculated by subtracting each plasma amino acid concentration after the start of treatment from each plasma amino acid concentration before the start of the treatment, and a change related to the difference. Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(3~6個)の組み合わせを上記19種のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (3 to 6) to be included in the logistic regression equation as a multivariate discriminant from the above 19 amino acids and adopt a random sampling method as cross validation to improve the discriminating performance of cancer immunotherapy effects The logistic regression equation to maximize was searched.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図50-54に示す。ここで、図50-54には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図50-54に含まれる式における変数の出現頻度を図55に示す。出現頻度が多い順に10位までのアミノ酸変数は、Lys,Gln,Thr,Phe,Met,Pro,Ser,Ala,Asn,Valであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 Figure 50-54 shows a list of logistic regression equations with equally good discriminating ability evaluated by the value of ROC_AUC. 50-54 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 55 shows the appearance frequency of variables in the expressions included in FIGS. The amino acid variables up to position 10 in the order of appearance frequency were Lys, Gln, Thr, Phe, Met, Pro, Ser, Ala, Asn, and Val. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 4T1担癌マウスの脾臓からT細胞を単離し、抗CD3抗体、抗CD28抗体およびIL-2存在下で活性化・増殖させることにより抗4T1活性化T細胞を作製した。 T cells were isolated from the spleen of 4T1 tumor-bearing mice, and anti-4T1 activated T cells were prepared by activating and proliferating in the presence of anti-CD3 antibody, anti-CD28 antibody and IL-2.
 4T1細胞あるいはCT26細胞をそれぞれBalb/cマウス10匹に皮下移植(1x10 cells/100μl)し、移植後10日目に前述の抗4T1活性化T細胞を尾静脈より移植した。 4T1 cells or CT26 cells were subcutaneously transplanted (1 × 10 6 cells / 100 μl) into 10 Balb / c mice, respectively, and the above-mentioned anti-4T1 activated T cells were transplanted from the tail vein 10 days after the transplantation.
 抗4T1活性化T細胞移植の前(治療前,pre)と移植後15日目(治療後,post(治療中,施術中ということ))に採血を行なった(図56)。 Blood was collected before anti-4T1-activated T cell transplantation (before treatment, pre) and on the 15th day after transplantation (after treatment, post (during treatment, during treatment)) (FIG. 56).
 移植後18日目に4T1担癌マウスならびにCT26担癌マウスの腫瘍組織を切除・凍結切片を作製した後、H/E染色と抗CD4抗体、抗CD8抗体ならびに抗F4/80抗体(マクロファージのマーカー)による蛍光免疫染色を行った(図57)。 On day 18 after transplantation, tumor tissues of 4T1 tumor-bearing mice and CT26 tumor-bearing mice were excised and frozen sections were prepared, and then H / E staining, anti-CD4 antibody, anti-CD8 antibody and anti-F4 / 80 antibody (macrophage marker) ) Fluorescence immunostaining was performed (FIG. 57).
 4T1担癌マウスの腫瘍組織中には広範囲にわたってネクローシスが認められたが、CT26担癌マウスの腫瘍組織中では認められなかった。また、4T1担癌マウスの腫瘍組織中にはCD4T細胞ならびにCD8T細胞の浸潤が認められた(図57中の、丸で囲まれた箇所を参照)が、CT26担癌マウスの腫瘍組織中ではこれらT細胞の浸潤は認められなかった。マクロファージの浸潤は両担癌マウスの腫瘍組織で認めた(図57中の、丸で囲まれた箇所を参照)。 Necrosis was observed over a wide range in the tumor tissue of 4T1 tumor-bearing mice, but not in the tumor tissue of CT26-bearing mice. In addition, infiltration of CD4 + T cells and CD8 + T cells was observed in the tumor tissue of 4T1 tumor-bearing mice (see the circled portion in FIG. 57), but tumors of CT26-bearing mice were observed. Infiltration of these T cells was not observed in the tissue. Macrophage infiltration was observed in the tumor tissues of both cancer-bearing mice (see the circled area in FIG. 57).
 以上の結果から抗4T1活性化T細胞を移入した4T1担癌マウスをレスポンダー、抗4T1活性化T細胞を移入したCT26担癌マウスをノンレスポンダーとした。 Based on the above results, 4T1 tumor-bearing mice transferred with anti-4T1 activated T cells were used as responders, and CT26 tumor-bearing mice transferred with anti-4T1 activated T cells were used as non-responders.
 血漿中のアミノ酸濃度の測定を、上述した実施形態で説明した(A)の測定方法で行った。 Measurement of amino acid concentration in plasma was performed by the measurement method (A) described in the above embodiment.
 4T1担癌マウスの血漿中アミノ酸濃度のデータを図58および図59に示す。図58および図59において、横軸は治療開始前(pre)と治療開始後(post)とを表し、縦軸は各アミノ酸濃度(μM)の平均値を表す。Student’s t-testの結果、治療開始前に比べて治療開始後では、Arg,His,Met,Cys2が有意に低下し、Glu,Aspは有意に増加していた(*:p<0.05,**:p<0.01,****:p<0.0001)。 Data of amino acid concentrations in plasma of 4T1 tumor-bearing mice are shown in FIG. 58 and FIG. 58 and 59, the horizontal axis represents the pre-treatment start (pre) and the post-treatment start (post), and the vertical axis represents the average value of each amino acid concentration (μM). As a result of Student's t-test, Arg, His, Met, and Cys2 were significantly decreased and Glu and Asp were significantly increased (*: p <0. 05, **: p <0.01, ***: p <0.0001).
 同様にCT26担癌マウスの血漿中アミノ酸濃度のデータを図60および図61に示す。Student’s t-testの結果、治療開始前に比べて治療開始後では、Alaが有意に低下し、Thr,Pro,Glu,Aspは有意に増加していた(*:p<0.05,**:p<0.01)。 Similarly, data of amino acid concentrations in plasma of CT26-bearing mice are shown in FIG. 60 and FIG. As a result of Student's t-test, Ala was significantly decreased and Thr, Pro, Glu, and Asp were significantly increased after the start of treatment compared to before the start of treatment (*: p <0.05, **: p <0.01).
 また、治療開始前の各アミノ酸を100%としたときの治療開始後の各アミノ酸の分布を示すレーダーチャートを、図62および図63に示す。4T1担癌マウス(レスポンダー)とCT26担癌(ノンレスポンダー)を比べるとアミノ酸プロファイルのパターンが異なった。以上の結果、抗腫瘍効果の有無によって血漿中アミノ酸プロファイル変化のパターンが異なることが明らかとなった。4T1担癌マウスの治療開始前後で有意差があったアミノ酸変数のうちArg,His,Met,Cys2は癌免疫療法の効果の判別能を持つことが判明した。また、CT26担癌マウスの治療開始前後で有意差があったアミノ酸変数のうちAla,Thr,Proについても癌免疫療法の効果判別能を持つことが判明した。 Further, radar charts showing the distribution of each amino acid after the start of treatment when each amino acid before the start of treatment is taken as 100% are shown in FIGS. When comparing 4T1 tumor-bearing mice (responders) and CT26 cancer-bearing (non-responders), the pattern of amino acid profiles was different. As a result, it became clear that the pattern of amino acid profile change in plasma differs depending on the presence or absence of antitumor effects. Of the amino acid variables that differed significantly before and after the start of treatment of 4T1 tumor-bearing mice, it was found that Arg, His, Met, and Cys2 have the ability to discriminate the effects of cancer immunotherapy. It was also found that Ala, Thr, Pro among the amino acid variables that had a significant difference before and after the start of treatment of CT26 tumor-bearing mice also had the ability to discriminate cancer immunotherapy effects.
 更に、4T1担癌マウスにおける各アミノ酸変数による治療開始前と治療開始後の2群判別性能を、ROC曲線の曲線下面積(ROC_AUC)で評価した。その結果、アミノ酸変数Thr,Val,Ile,Lys,Arg,His,Phe,Met,Cys2,Gln,Glu,Asp,CitについてAUCが0.7より大きい値を示した(図64および図65)。これにより、アミノ酸変数Thr,Val,Ile,Lys,Arg,His,Phe,Met,Cys2,Gln,Glu,Asp,Citが、癌免疫療法の効果の判別能を持つことが判明した。 Furthermore, the 2-group discrimination performance before and after the start of treatment by each amino acid variable in 4T1 tumor-bearing mice was evaluated by the area under the curve (ROC_AUC) of the ROC curve. As a result, AUC was greater than 0.7 for amino acid variables Thr, Val, Ile, Lys, Arg, His, Phe, Met, Cys2, Gln, Glu, Asp, and Cit (FIGS. 64 and 65). This revealed that the amino acid variables Thr, Val, Ile, Lys, Arg, His, Phe, Met, Cys2, Gln, Glu, Asp, and Cit have the ability to discriminate the effects of cancer immunotherapy.
 実施例6と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post/pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 国際公開第2004/052191号に記載の方法を用いて、多変量判別式としての分数式に含める変数(4個以下)の組み合わせを上記22種類のアミノ酸から探索し、そしてクロスバリデーションとしてブートストラップ法を採用して、癌免疫療法の効果の判別性能を最大化する分数式を探索した。 Using the method described in International Publication No. 2004/052191, a combination of variables (4 or less) to be included in a fractional expression as a multivariate discriminant is searched from the above 22 kinds of amino acids, and a bootstrap method is used as a cross validation Was used to search for a fractional expression that maximizes the ability to discriminate the effects of cancer immunotherapy.
 ROC_AUCの値が1であるという良好な判別能を有する分数式の一覧を図66に示す。図66に含まれる式における変数の出現頻度を図67に示す。出現頻度が多い順に10位までのアミノ酸変数は、Pro,Glu,Orn,Arg,Gly,Ile,Thr,Trp,Ser,Tyrであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 FIG. 66 shows a list of fractional expressions having good discrimination ability that the value of ROC_AUC is 1. FIG. 67 shows the frequency of occurrence of variables in the formula included in FIG. The amino acid variables up to position 10 in the order of appearance frequency were Pro, Glu, Orn, Arg, Gly, Ile, Thr, Trp, Ser, Tyr. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例6と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post/pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(4~6個)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (4-6) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids and adopt a random sampling method as cross-validation to improve the discriminating performance of cancer immunotherapy effects The logistic regression equation to maximize was searched.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図68、図69および図70に示す。ここで、図68、図69および図70には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図68、図69および図70に含まれる式における変数の出現頻度を図71に示す。出現頻度が多い順に10位までのアミノ酸変数は、Trp,His,Thr,Ile,Pro,Phe,Asn,Arg,Cys2,Lysであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 68, FIG. 69, and FIG. 70 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC. Here, FIG. 68, FIG. 69, and FIG. 70 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 71 shows the frequency of occurrence of variables in the expressions included in FIGS. The amino acid variables up to position 10 in the order of appearance frequency were Trp, His, Thr, Ile, Pro, Phe, Asn, Arg, Cys2, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例6と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post-pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post-pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and a change related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(4~6個)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (4-6) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids and adopt a random sampling method as cross-validation to improve the discriminating performance of cancer immunotherapy effects The logistic regression equation to maximize was searched.
 ROC_AUCの値で評価した判別能が同等に良好なロジスティック回帰式の一覧を図72、図73および図74に示す。ここで、図72、図73および図74には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。図72、図73および図74に含まれる式における変数の出現頻度を図75に示す。出現頻度が多い順に10位までのアミノ酸変数は、Trp,His,Thr,Ile,Pro,Phe,Asn,Arg,Cys2,Lysであった。これにより、これらのアミノ酸変数が、癌免疫療法の効果の判別能を持つことが判明した。 72, 73, and 74 show a list of logistic regression equations with equally good discrimination ability evaluated by the value of ROC_AUC. Here, FIG. 72, FIG. 73, and FIG. 74 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation. FIG. 75 shows the appearance frequency of variables in the expressions included in FIGS. 72, 73, and 74. The amino acid variables up to position 10 in the order of appearance frequency were Trp, His, Thr, Ile, Pro, Phe, Asn, Arg, Cys2, and Lys. This revealed that these amino acid variables have the ability to discriminate the effects of cancer immunotherapy.
 実施例6と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post/pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post / pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and changes related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(2個)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (2) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids, and adopt a random sampling method as cross-validation to maximize the discriminating performance of cancer immunotherapy effects We searched for a logistic regression equation.
 ROC_AUCの値で評価した判別能が0.7以上であるロジスティック回帰式の一覧を図76-80に示す。ここで、図76-80には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。 A list of logistic regression equations with discriminability evaluated by ROC_AUC value of 0.7 or more is shown in Figure 76-80. Here, FIG. 76-80 shows a logistic regression equation (including variable combinations, coefficients, and constants) and a value of ROC_AUC with cross validation.
 実施例6と同じ血漿中アミノ酸濃度データに基づいて、治療開始後の各血漿中アミノ酸濃度を治療開始前の各血漿中アミノ酸濃度で割った比(post-pre)を計算し、当該比に関する変化量データを得た。得られた変化量データを用いて、癌免疫療法効果の判別に有効な、血漿中アミノ酸濃度を変数に持つ癌免疫療法の効果を判別するための多変量判別式(多変量関数)を求めた。 Based on the same plasma amino acid concentration data as in Example 6, a ratio (post-pre) obtained by dividing each plasma amino acid concentration after the start of treatment by each plasma amino acid concentration before the start of treatment was calculated, and a change related to the ratio Quantity data was obtained. Using the obtained variation data, a multivariate discriminant (multivariate function) for determining the effect of cancer immunotherapy with the amino acid concentration in the plasma as a variable was determined, which was effective in determining the cancer immunotherapy effect. .
 多変量判別式としてのロジスティック回帰式に含める変数(2個)の組み合わせを上記22種類のアミノ酸から探索し、クロスバリデーションとしてランダムサンプリング法を採用して、癌免疫療法の効果の判別性能を最大化するロジスティック回帰式を探索した。 Search for combinations of variables (2) to be included in the logistic regression equation as a multivariate discriminant from the above 22 types of amino acids, and adopt a random sampling method as cross-validation to maximize the discriminating performance of cancer immunotherapy effects We searched for a logistic regression equation.
 ROC_AUCの値で評価した判別能が0.7以上であるロジスティック回帰式の一覧を図81-85に示す。ここで、図81-85には、ロジスティック回帰式(変数の組み合わせと係数と定数を含む)と、クロスバリデーション有りでのROC_AUCの値とが示されている。 81-85 shows a list of logistic regression equations with discriminability evaluated by ROC_AUC value of 0.7 or more. Here, FIGS. 81 to 85 show the logistic regression equation (including variable combinations, coefficients, and constants) and the value of ROC_AUC with cross validation.
 以上のように、本発明にかかる癌免疫療法の評価方法などは、産業上の多くの分野、特に医薬品や食品、医療などの分野で広く実施することができ、特に、癌免疫療法の治療効果の評価を行うバイオインフォマティクス分野において極めて有用である。 As described above, the cancer immunotherapy evaluation method and the like according to the present invention can be widely implemented in many industrial fields, particularly in the fields of pharmaceuticals, foods, and medicine, and in particular, the therapeutic effect of cancer immunotherapy. It is extremely useful in the field of bioinformatics for evaluating the above.
 100 癌免疫療法評価装置
 102 制御部
 102a 要求解釈部
 102b 閲覧処理部
 102c 認証処理部
 102d 電子メール生成部
 102e Webページ生成部
 102f 受信部
 102g 癌状態情報指定部
 102h 多変量判別式作成部
 102h1 候補多変量判別式作成部
 102h2 候補多変量判別式検証部
 102h3 変数選択部
 102i 判別値算出部
 102j 判別値基準評価部
 102j1 判別値基準判別部
 102k 結果出力部
 102m 送信部
 104 通信インターフェース部
 106 記憶部
 106a 利用者情報ファイル
 106b アミノ酸濃度データファイル
 106c 癌状態情報ファイル
 106d 指定癌状態情報ファイル
 106e 多変量判別式関連情報データベース
 106e1 候補多変量判別式ファイル
 106e2 検証結果ファイル
 106e3 選択癌状態情報ファイル
 106e4 多変量判別式ファイル
 106f 判別値ファイル
 106g 評価結果ファイル
 108 入出力インターフェース部
 112 入力装置
 114 出力装置
 200 クライアント装置(情報通信端末装置)
 300 ネットワーク
 400 データベース装置
DESCRIPTION OF SYMBOLS 100 Cancer immunotherapy evaluation apparatus 102 Control part 102a Request interpretation part 102b Browsing process part 102c Authentication process part 102d E-mail production | generation part 102e Web page production | generation part 102f Reception part 102g Cancer state information designation | designated part 102h Multivariate discriminant preparation part 102h1 Many candidates Variable discriminant creation unit 102h2 Candidate multivariate discriminant verification unit 102h3 Variable selection unit 102i Discriminant value calculation unit 102j Discriminant value criterion evaluation unit 102j1 Discriminant value criterion discriminator 102k Result output unit 102m Transmitter 104 Communication interface unit 106 Storage unit 106a Person information file 106b Amino acid concentration data file 106c Cancer status information file 106d Designated cancer status information file 106e Multivariate discriminant-related information database 106e1 Candidate multivariate discriminant file 106e2 Testimony result file 106e3 selection cancer state information file 106e4 Multivariate discriminant file 106f discriminant value file 106g Evaluation result file 108 output interface unit 112 input unit 114 output unit 200 the client device (information communication terminal apparatus)
300 network 400 database device

Claims (22)

  1.  癌免疫療法による治療を受ける評価対象から前記治療が開始される前に採取された血液中のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記評価対象から前記治療が開始された後に採取された血液中のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを取得する取得ステップと、
     前記取得ステップで取得した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに基づいて、前記評価対象に対する前記治療の効果を評価する濃度値基準評価ステップと
     を含むことを特徴とする癌免疫療法の評価方法。
    Pre-treatment amino acid concentration data regarding amino acid concentration values collected before the start of treatment from an evaluation subject receiving treatment by cancer immunotherapy, and after the treatment is started from the evaluation subject An acquisition step of acquiring amino acid concentration data after the start of treatment related to the concentration value of amino acids in the blood;
    A concentration value reference evaluation step for evaluating the effect of the treatment on the evaluation target based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data acquired in the acquisition step. Evaluation method of immunotherapy.
  2.  前記濃度値基準評価ステップは、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの前記濃度値に基づいて、前記評価対象に対する前記治療の効果を評価すること、
     を特徴とする請求項1に記載の癌免疫療法の評価方法。
    The concentration value reference evaluation step includes Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. Evaluating the effect of the treatment on the evaluation target based on the concentration value of at least one of Thr, Met, Lys, Arg, Gly, Cys2, and Pro;
    The method for evaluating cancer immunotherapy according to claim 1.
  3.  前記濃度値基準評価ステップは、前記濃度値に基づいて、前記評価対象に対して前記治療が有効であるかを評価すること、
     を特徴とする請求項2に記載の癌免疫療法の評価方法。
    The concentration value reference evaluation step evaluates whether the treatment is effective for the evaluation object based on the concentration value;
    The method for evaluating cancer immunotherapy according to claim 2.
  4.  前記濃度値基準評価ステップは、
     前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む予め設定された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップ
     をさらに含むこと、
     を特徴とする請求項1に記載の癌免疫療法の評価方法。
    The density value reference evaluation step includes:
    Based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data, and a preset multivariate discriminant including the amino acid concentration as a variable, the value of the multivariate discriminant and A discriminant value calculating step of calculating a discriminant value corresponding to the evaluation result relating to the effect of the treatment,
    The method for evaluating cancer immunotherapy according to claim 1.
  5.  前記多変量判別式は、ロジスティック回帰式、分数式、線形判別式、重回帰式、サポートベクターマシンで作成された式、マハラノビス距離法で作成された式、正準判別分析で作成された式、決定木で作成された式のいずれか1つであること、
     を特徴とする請求項4に記載の癌免疫療法の評価方法。
    The multivariate discriminant is a logistic regression formula, fractional formula, linear discriminant formula, multiple regression formula, formula created with support vector machine, formula created with Mahalanobis distance method, formula created with canonical discriminant analysis, Be one of the formulas created in the decision tree,
    The method for evaluating cancer immunotherapy according to claim 4.
  6.  前記判別値算出ステップは、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データに含まれるVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つの前記濃度値、ならびにVal,Ile,Leu,His,Phe,Trp,Gln,Glu,Asp,Orn,Ala,Ser,Thr,Met,Lys,Arg,Gly,Cys2,Proのうち少なくとも1つを前記変数として含む前記多変量判別式に基づいて、前記判別値を算出すること、
     を特徴とする請求項4または5に記載の癌免疫療法の評価方法。
    The discriminant value calculating step includes Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr included in the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data. , Met, Lys, Arg, Gly, Cys2, Pro, and at least one of the concentration values, and Val, Ile, Leu, His, Phe, Trp, Gln, Glu, Asp, Orn, Ala, Ser, Thr, Met, Calculating the discriminant value based on the multivariate discriminant including at least one of Lys, Arg, Gly, Cys2, and Pro as the variable;
    The method for evaluating cancer immunotherapy according to claim 4 or 5.
  7.  前記多変量判別式は、Glu,Cys2,Trp,Asp,Orn,Phe,Val,Ile,Gly,Hisのうち少なくとも1つを前記変数として含む前記分数式、またはTrp,Thr,His,Arg,Ile,Pro,Phe,Met,Ala,Lys,Asp,Ser,Leuのうち少なくとも1つを前記変数として含む前記ロジスティック回帰式であること、
     を特徴とする請求項6に記載の癌免疫療法の評価方法。
    The multivariate discriminant is the fractional expression including at least one of Glu, Cys2, Trp, Asp, Orn, Phe, Val, Ile, Gly, His as the variable, or Trp, Thr, His, Arg, Ile. , Pro, Phe, Met, Ala, Lys, Asp, Ser, and Leu, the logistic regression equation including at least one of the variables,
    The method for evaluating cancer immunotherapy according to claim 6.
  8.  前記濃度値基準評価ステップは、
     前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップ
     をさらに含むこと、
     を特徴とする請求項4から7のいずれか1つに記載の癌免疫療法の評価方法。
    The density value reference evaluation step includes:
    A discriminant value criterion evaluation step for evaluating the effect of the treatment on the evaluation target based on the discriminant value calculated in the discriminant value calculating step;
    The method for evaluating cancer immunotherapy according to any one of claims 4 to 7, wherein:
  9.  制御部と記憶部とを備えた癌免疫療法評価装置であって、
     前記制御部は、
     癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出手段
     を備えたこと、
     を特徴とする癌免疫療法評価装置。
    A cancer immunotherapy evaluation apparatus comprising a control unit and a storage unit,
    The controller is
    Before the start of treatment by cancer immunotherapy, the amino acid concentration data before starting treatment regarding the concentration value of the evaluation target amino acid to receive the treatment, and the start of treatment regarding the concentration value of the target amino acid after the start of the treatment Based on the post-amino acid concentration data and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the multivariate discriminant is the value of the multivariate discriminant and the evaluation result regarding the effect of the treatment on the evaluation target Provided with a discriminant value calculating means for calculating the corresponding discriminant value;
    A cancer immunotherapy evaluation apparatus characterized by the above.
  10.  前記制御部は、
     前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段
     をさらに備えたこと、
     を特徴とする請求項9に記載の癌免疫療法評価装置。
    The controller is
    A discriminant value criterion evaluation unit for evaluating the effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation unit;
    The cancer immunotherapy evaluation apparatus according to claim 9.
  11.  制御部と記憶部とを備えた情報処理装置において実行される癌免疫療法評価方法であって、
     前記制御部において実行される、
     癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップ
     を含むこと、
     を特徴とする癌免疫療法評価方法。
    A cancer immunotherapy evaluation method executed in an information processing device including a control unit and a storage unit,
    Executed in the control unit,
    Before the start of treatment by cancer immunotherapy, the amino acid concentration data before starting treatment regarding the concentration value of the evaluation target amino acid to receive the treatment, and the start of treatment regarding the concentration value of the target amino acid after the start of the treatment Based on the post-amino acid concentration data and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the multivariate discriminant is the value of the multivariate discriminant and the evaluation result regarding the effect of the treatment on the evaluation target Including a discriminant value calculating step for calculating a corresponding discriminant value,
    A cancer immunotherapy evaluation method characterized by the above.
  12.  前記制御部において実行される、
     前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップ
     をさらに含むこと、
     を特徴とする請求項11に記載の癌免疫療法評価方法。
    Executed in the control unit,
    A discriminant value criterion evaluation step for evaluating the effect of the treatment on the evaluation target based on the discriminant value calculated in the discriminant value calculating step;
    The method for evaluating cancer immunotherapy according to claim 11.
  13.  制御部と記憶部とを備えた情報処理装置において実行させるための癌免疫療法評価プログラムであって、
     前記制御部において実行させるための、
     癌免疫療法による治療が開始される前の、前記治療を受ける評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、およびアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出ステップ
     を含むこと、
     を特徴とする癌免疫療法評価プログラム。
    A cancer immunotherapy evaluation program for execution in an information processing apparatus including a control unit and a storage unit,
    For executing in the control unit,
    Before the start of treatment by cancer immunotherapy, the amino acid concentration data before starting treatment regarding the concentration value of the evaluation target amino acid to receive the treatment, and the start of treatment regarding the concentration value of the target amino acid after the start of the treatment Based on the post-amino acid concentration data and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable, the multivariate discriminant is the value of the multivariate discriminant and the evaluation result regarding the effect of the treatment on the evaluation target Including a discriminant value calculating step for calculating a corresponding discriminant value,
    A cancer immunotherapy evaluation program characterized by
  14.  前記制御部において実行させるための、
     前記判別値算出ステップで算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価ステップ
     をさらに含むこと、
     を特徴とする請求項13に記載の癌免疫療法評価プログラム。
    For executing in the control unit,
    A discriminant value criterion evaluation step for evaluating the effect of the treatment on the evaluation target based on the discriminant value calculated in the discriminant value calculating step;
    The cancer immunotherapy evaluation program according to claim 13.
  15.  制御部と記憶部とを備えた癌免疫療法評価装置と、制御部を備え、癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置とを、ネットワークを介して通信可能に接続して構成された癌免疫療法評価システムであって、
     前記情報通信端末装置の前記制御部は、
     前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを、前記癌免疫療法評価装置へ送信するアミノ酸濃度データ送信手段と、
     前記癌免疫療法評価装置から送信された、アミノ酸の濃度を変数として含む多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を受信する結果受信手段と
     を備え、
     前記癌免疫療法評価装置の前記制御部は、
     前記情報通信端末装置から送信された前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データを受信するアミノ酸濃度データ受信手段と、
     前記アミノ酸濃度データ受信手段で受信した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびに前記記憶部に記憶された前記多変量判別式に基づいて、前記判別値を算出する判別値算出手段と、
     前記判別値算出手段で算出した前記判別値を前記情報通信端末装置へ送信する結果送信手段と、
     を備えたこと、
     を特徴とする癌免疫療法評価システム。
    A cancer immunotherapy evaluation device including a control unit and a storage unit, and an information communication terminal device including a control unit and providing amino acid concentration data relating to the concentration value of an amino acid to be evaluated that is treated by cancer immunotherapy, A cancer immunotherapy evaluation system configured to be communicably connected via
    The control unit of the information communication terminal device,
    Amino acid concentration data before treatment start related to the concentration value of the amino acid to be evaluated before the treatment is started, and amino acid concentration data after treatment start related to the concentration value of the amino acid to be evaluated after the treatment has been started, Amino acid concentration data transmitting means for transmitting to the cancer immunotherapy evaluation device;
    A result receiving means for receiving a discriminant value, which is a value of a multivariate discriminant that includes an amino acid concentration as a variable and that is transmitted from the cancer immunotherapy evaluation device and that corresponds to an evaluation result relating to the effect of the treatment on the evaluation target; With
    The control unit of the cancer immunotherapy evaluation apparatus comprises:
    Amino acid concentration data receiving means for receiving the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data transmitted from the information communication terminal device;
    Discriminant value for calculating the discriminant value based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data received by the amino acid concentration data receiving means, and the multivariate discriminant stored in the storage unit A calculation means;
    A result transmitting means for transmitting the discriminant value calculated by the discriminant value calculating means to the information communication terminal device;
    Having
    A cancer immunotherapy evaluation system characterized by.
  16.  前記癌免疫療法評価装置の前記制御部は、前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段をさらに備え、
     前記結果送信手段は、前記判別値基準評価手段で得られた前記評価対象の前記評価結果を前記情報通信端末装置へ送信し、
     前記結果受信手段は、前記癌免疫療法評価装置から送信された前記評価対象の前記評価結果を受信すること、
     を特徴とする請求項15に記載の癌免疫療法評価システム。
    The control unit of the cancer immunotherapy evaluation apparatus further includes a discriminant value reference evaluation unit that evaluates the effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation unit,
    The result transmission means transmits the evaluation result of the evaluation target obtained by the discriminant value criterion evaluation means to the information communication terminal device,
    The result receiving means receives the evaluation result of the evaluation object transmitted from the cancer immunotherapy evaluation device;
    The cancer immunotherapy evaluation system according to claim 15.
  17.  制御部を備え、癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置であって、
     前記制御部は、
     アミノ酸の濃度を変数として含む多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を取得する結果取得手段
     を備え、
     前記判別値は、前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データ、および前記多変量判別式に基づいて算出したものであること、
     を特徴とする情報通信端末装置。
    An information communication terminal device comprising a control unit and providing amino acid concentration data relating to a concentration value of an amino acid to be evaluated for treatment by cancer immunotherapy,
    The controller is
    A result obtaining means for obtaining a discriminant value corresponding to an evaluation result relating to the effect of the treatment on the evaluation target, which is a value of a multivariate discriminant including a concentration of amino acid as a variable;
    The discriminant value is a pre-treatment amino acid concentration data related to the concentration value of the evaluation target amino acid before the start of the treatment, and a post-treatment start amino acid related to the evaluation target amino acid concentration value after the start of the treatment. Calculated based on concentration data and the multivariate discriminant,
    An information communication terminal device.
  18.  前記結果取得手段は、前記評価対象に対する前記治療の効果に関する前記評価結果を取得し、
     前記評価結果は、前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価して得られた結果であること、
     を特徴とする請求項17に記載の情報通信端末装置。
    The result acquisition means acquires the evaluation result related to the effect of the treatment on the evaluation target,
    The evaluation result is a result obtained by evaluating the effect of the treatment on the evaluation target based on the discriminant value;
    The information communication terminal device according to claim 17.
  19.  前記多変量判別式を記憶し、前記判別値を算出する癌免疫療法評価装置とネットワークを介して通信可能に接続されており、
     前記制御部は、前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データを、前記癌免疫療法評価装置へ送信するアミノ酸濃度データ送信手段をさらに備え、
     前記結果取得手段は、前記癌免疫療法評価装置から送信された前記判別値を受信すること、
     を特徴とする請求項17に記載の情報通信端末装置。
    The multivariate discriminant is stored and connected to a cancer immunotherapy evaluation apparatus that calculates the discriminant value through a network,
    The control unit further comprises amino acid concentration data transmission means for transmitting the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data to the cancer immunotherapy evaluation device,
    The result acquisition means receives the discriminant value transmitted from the cancer immunotherapy evaluation device;
    The information communication terminal device according to claim 17.
  20.  前記癌免疫療法評価装置は、前記治療の効果をさらに評価し、
     前記結果取得手段は、前記癌免疫療法評価装置から送信された、前記評価対象に対する前記治療の効果に関する前記評価結果を受信すること、
     を特徴とする請求項19に記載の情報通信端末装置。
    The cancer immunotherapy evaluation device further evaluates the effect of the treatment,
    The result acquisition means receives the evaluation result relating to the effect of the treatment on the evaluation target transmitted from the cancer immunotherapy evaluation device;
    The information communication terminal device according to claim 19.
  21.  癌免疫療法による治療を受ける評価対象のアミノ酸の濃度値に関するアミノ酸濃度データを提供する情報通信端末装置とネットワークを介して通信可能に接続された、制御部と記憶部とを備えた癌免疫療法評価装置であって、
     前記制御部は、
     前記情報通信端末装置から送信された、前記治療が開始される前の前記評価対象のアミノ酸の濃度値に関する治療開始前アミノ酸濃度データ、および前記治療が開始された後の前記評価対象のアミノ酸の濃度値に関する治療開始後アミノ酸濃度データを受信するアミノ酸濃度データ受信手段と、
     前記アミノ酸濃度データ受信手段で受信した前記治療開始前アミノ酸濃度データおよび前記治療開始後アミノ酸濃度データ、ならびにアミノ酸の濃度を変数として含む前記記憶部に記憶された多変量判別式に基づいて、当該多変量判別式の値であって前記評価対象に対する前記治療の効果に関する評価結果に相当する判別値を算出する判別値算出手段と、
     前記判別値算出手段で算出した前記判別値を前記情報通信端末装置へ送信する結果送信手段と、
     を備えたこと、
     を特徴とする癌免疫療法評価装置。
    Cancer immunotherapy evaluation comprising a control unit and a storage unit that are communicably connected via an information communication terminal device that provides amino acid concentration data relating to the concentration value of an amino acid to be evaluated for treatment by cancer immunotherapy A device,
    The controller is
    The pre-treatment amino acid concentration data relating to the concentration value of the evaluation target amino acid before the start of the treatment and the concentration of the evaluation target amino acid after the start of the treatment transmitted from the information communication terminal device Amino acid concentration data receiving means for receiving amino acid concentration data after the start of treatment regarding the value;
    Based on the pre-treatment amino acid concentration data and the post-treatment amino acid concentration data received by the amino acid concentration data receiving means, and the multivariate discriminant stored in the storage unit including the amino acid concentration as a variable. A discriminant value calculating means for calculating a discriminant value corresponding to an evaluation result relating to an effect of the treatment on the evaluation object, which is a value of a variable discriminant;
    A result transmitting means for transmitting the discriminant value calculated by the discriminant value calculating means to the information communication terminal device;
    Having
    A cancer immunotherapy evaluation apparatus characterized by the above.
  22.  前記制御部は、前記判別値算出手段で算出した前記判別値に基づいて、前記評価対象に対する前記治療の効果を評価する判別値基準評価手段をさらに備え、
     前記結果送信手段は、前記判別値基準評価手段で得られた前記評価対象の前記評価結果を前記情報通信端末装置へ送信すること、
     を特徴とする請求項21に記載の癌免疫療法評価装置。
    The control unit further includes a discriminant value reference evaluation unit that evaluates the effect of the treatment on the evaluation target based on the discriminant value calculated by the discriminant value calculation unit,
    The result transmitting means transmits the evaluation result of the evaluation object obtained by the discriminant value criterion evaluating means to the information communication terminal device;
    The cancer immunotherapy evaluation apparatus according to claim 21, wherein:
PCT/JP2013/061807 2012-05-11 2013-04-22 Cancer immunotherapy evaluation method, cancer immunotherapy evaluation device, cancer immunotherapy evaluation program, cancer immunotherapy evaluation system, and information communication terminal device WO2013168550A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2014514434A JP6375947B2 (en) 2012-05-11 2013-04-22 Acquisition method, evaluation device, evaluation method, evaluation program, evaluation system, and terminal device

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2012-109920 2012-05-11
JP2012109920 2012-05-11

Publications (1)

Publication Number Publication Date
WO2013168550A1 true WO2013168550A1 (en) 2013-11-14

Family

ID=49550601

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2013/061807 WO2013168550A1 (en) 2012-05-11 2013-04-22 Cancer immunotherapy evaluation method, cancer immunotherapy evaluation device, cancer immunotherapy evaluation program, cancer immunotherapy evaluation system, and information communication terminal device

Country Status (2)

Country Link
JP (1) JP6375947B2 (en)
WO (1) WO2013168550A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190089895A (en) 2016-12-01 2019-07-31 아지노모토 가부시키가이샤 A method of monitoring cancer, a calculation method, an evaluation apparatus, a calculation apparatus, an evaluation program, a calculation program, an evaluation system,
CN110763806A (en) * 2019-10-25 2020-02-07 三只松鼠股份有限公司 Method for evaluating spicy grade of duck neck
KR20220093122A (en) 2019-11-08 2022-07-05 아지노모토 가부시키가이샤 Evaluation method, calculation method, evaluation device, calculation device, evaluation program, calculation program, recording medium, evaluation system and terminal device for pharmacological action of an immune checkpoint inhibitor
JP7103220B2 (en) 2016-06-30 2022-07-20 味の素株式会社 Evaluation method of postpartum ketosis

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007523346A (en) * 2004-02-19 2007-08-16 イェール ユニバーシティ Identification of oncoprotein biomarkers using proteomic technology
WO2008075664A1 (en) * 2006-12-21 2008-06-26 Ajinomoto Co., Inc. Method for evaluation of cancer, cancer evaluation apparatus, cancer evaluation method, cancer evaluation system, cancer evaluation program, and recording medium
WO2010119003A1 (en) * 2009-04-17 2010-10-21 Transgene Sa Biomarker for monitoring patients
JP2011058949A (en) * 2009-09-09 2011-03-24 Hitachi Ltd Marker factor and application of atherosclerosis

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4581994B2 (en) * 2002-12-09 2010-11-17 味の素株式会社 Biological state information processing apparatus, biological state information processing method, biological state information management system, program, and recording medium

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2007523346A (en) * 2004-02-19 2007-08-16 イェール ユニバーシティ Identification of oncoprotein biomarkers using proteomic technology
WO2008075664A1 (en) * 2006-12-21 2008-06-26 Ajinomoto Co., Inc. Method for evaluation of cancer, cancer evaluation apparatus, cancer evaluation method, cancer evaluation system, cancer evaluation program, and recording medium
WO2010119003A1 (en) * 2009-04-17 2010-10-21 Transgene Sa Biomarker for monitoring patients
JP2011058949A (en) * 2009-09-09 2011-03-24 Hitachi Ltd Marker factor and application of atherosclerosis

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7103220B2 (en) 2016-06-30 2022-07-20 味の素株式会社 Evaluation method of postpartum ketosis
KR20190089895A (en) 2016-12-01 2019-07-31 아지노모토 가부시키가이샤 A method of monitoring cancer, a calculation method, an evaluation apparatus, a calculation apparatus, an evaluation program, a calculation program, an evaluation system,
CN110763806A (en) * 2019-10-25 2020-02-07 三只松鼠股份有限公司 Method for evaluating spicy grade of duck neck
KR20220093122A (en) 2019-11-08 2022-07-05 아지노모토 가부시키가이샤 Evaluation method, calculation method, evaluation device, calculation device, evaluation program, calculation program, recording medium, evaluation system and terminal device for pharmacological action of an immune checkpoint inhibitor

Also Published As

Publication number Publication date
JPWO2013168550A1 (en) 2016-01-07
JP6375947B2 (en) 2018-08-22

Similar Documents

Publication Publication Date Title
JP5470848B2 (en) Lung cancer evaluation method, lung cancer evaluation device, lung cancer evaluation method, lung cancer evaluation system, lung cancer evaluation program, recording medium, and information communication terminal device
JP5746811B2 (en) Colorectal cancer evaluation method, colorectal cancer evaluation device, colorectal cancer evaluation method, colorectal cancer evaluation system, colorectal cancer evaluation program, and recording medium
KR101272207B1 (en) Method of evaluating gastric cancer, gastric cancer evaluation apparatus, gastric cancer evaluation method, gastric cancer evaluation system, gastric cancer evaluation program and recording medium
JP5754136B2 (en) Female genital cancer evaluation method, female genital cancer evaluation device, female genital cancer evaluation method, female genital cancer evaluation program, recording medium, female genital cancer evaluation system, and information communication terminal device
JP2014025946A (en) Evaluation method for cancer, cancer evaluation device, cancer evaluation method, cancer evaluation system, cancer evaluation program, and recording medium
JP6299063B2 (en) Method for evaluating pancreatic cancer risk disease, pancreatic cancer risk disease evaluation device, pancreatic cancer risk disease evaluation method, pancreatic cancer risk disease evaluation program and pancreatic cancer risk disease evaluation system
US20210287802A1 (en) Method for evaluating pancreatic cancer, pancreatic cancer evaluating apparatus, pancreatic cancer evaluating method, pancreatic cancer evaluating program product, pancreatic cancer evaluating system and information communication terminal apparatus
JP6375947B2 (en) Acquisition method, evaluation device, evaluation method, evaluation program, evaluation system, and terminal device
WO2013011919A1 (en) Method for evaluating nash, device for evaluating nash, program for evaluating nash, system for evaluating nash, information-communication terminal device, and method for searching for substance used to prevent or improve nash
WO2010095682A1 (en) Obesity evaluation method
JP2014002174A (en) Method for evaluating breast cancer, breast cancer evaluation apparatus, breast cancer evaluation method, breast cancer evaluation system, breast cancer evaluation program and recording medium
JPWO2018066621A1 (en) Evaluation method of colon cancer, evaluation device, evaluation program, evaluation system, and terminal device
JP7337018B2 (en) Acquisition method, calculation method, evaluation device, calculation device, evaluation program, calculation program, evaluation system
JP6614215B2 (en) Acquisition method, calculation method, stroke evaluation device, calculation device, stroke evaluation program, calculation program, and stroke evaluation system
WO2008016101A1 (en) Method for evaluation of stress, stress evaluation apparatus, stress evaluation method, stress evaluation system, stress evaluation program, and recording medium
WO2013002381A1 (en) Method for evaluating fatty liver disease, device for evaluating fatty liver disease, method for evaluating fatty liver disease, program for evaluating fatty liver disease, system for evaluating fatty liver disease, information-communication terminal device, and method for searching for substance used to prevent or improve fatty-liver-disease
WO2013115283A1 (en) Method for evaluating early stage nephropathy, early stage nephropathy evaluation device, early stage nephropathy evaluation method, early stage nephropathy evaluation program, early stage nephropathy evaluation system and information communication terminal device
JP6270257B2 (en) Acquisition method, cardiovascular event evaluation device, cardiovascular event evaluation program, cardiovascular event evaluation system, and terminal device
WO2021090941A1 (en) Evaluation method, calculation method, evaluation device, calculation device, evaluation program, calculation program, recording medium, evaluation system, and terminal device for pharmacological action of immune check point inhibitor
JPWO2018066620A1 (en) Evaluation method for pancreatic cancer, evaluation device, evaluation program, evaluation system, and terminal device
JP2011080957A (en) Fibromyalgia and depression evaluation method, fibromyalgia and depression evaluation apparatus, and fibromyalgia and depression evaluation method
KR102477319B1 (en) Cancer monitoring method, calculation method, evaluation device, calculation device, evaluation program, calculation program, evaluation system, and terminal device

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: 13787646

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2014514434

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13787646

Country of ref document: EP

Kind code of ref document: A1