WO2023074711A1 - T cell evaluation method and composition for t cell evaluation - Google Patents

T cell evaluation method and composition for t cell evaluation Download PDF

Info

Publication number
WO2023074711A1
WO2023074711A1 PCT/JP2022/039806 JP2022039806W WO2023074711A1 WO 2023074711 A1 WO2023074711 A1 WO 2023074711A1 JP 2022039806 W JP2022039806 W JP 2022039806W WO 2023074711 A1 WO2023074711 A1 WO 2023074711A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
cells
acid sequence
gene
polypeptide
Prior art date
Application number
PCT/JP2022/039806
Other languages
French (fr)
Japanese (ja)
Inventor
新 金子
洋平 河合
佳奈 山口
勇一 熊木
達 二見
Original Assignee
国立大学法人京都大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人京都大学 filed Critical 国立大学法人京都大学
Publication of WO2023074711A1 publication Critical patent/WO2023074711A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M1/00Apparatus for enzymology or microbiology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M1/00Apparatus for enzymology or microbiology
    • C12M1/34Measuring or testing with condition measuring or sensing means, e.g. colony counters
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor

Definitions

  • the present invention provides a method for evaluating T cells, a composition for evaluating T cells, a pharmaceutical composition containing high-quality T cells obtained by the method for evaluating T cells, and cancer prevention or treatment using the pharmaceutical composition. It relates to treatment methods and the like.
  • T cells antigen-specific cytotoxic T-lymphocytes (CTL) related to the disease
  • CTL cytotoxic T-lymphocytes
  • T cells unmodified T cells
  • Expectations are high for cancer immunotherapy, which treats cancer by returning cells to the original patient.
  • Patent Document 1 describes a method of inducing CD8 ⁇ + ⁇ + CTL by culturing CD4 + CD8 + T cells in a medium containing interleukin 7 and a TCR (T cell receptor) activator.
  • T cell quality evaluation items include, for example, analysis of differentiation markers expressed at each stage of T cells, analysis of cytokines produced after TCR stimulation, proliferation ability, cytotoxic activity, animal experiments and cancer cell-suppressing activity.
  • the quality of T cells is comprehensively evaluated by these items, but such evaluations are often complicated and require a long period of time. Furthermore, since the cost of evaluation is high, there is a demand for a method that can evaluate the quality of T cells more simply and in a short period of time.
  • Non-Patent Document 1 As a method for evaluating the quality of such T cells, for example, it has been found that there is a relationship between the expression level of PD-1, lag3, etc. and the exhaustion of T cells (Non-Patent Document 1). , using these proteins as T cell exhaustion markers, a method of evaluating T cell exhaustion based on the expression level thereof.
  • the present invention has been made in view of the above-mentioned problems of the prior art, and provides a T cell evaluation method capable of evaluating the quality of relatively young T cells using a new index, and a T cell evaluation reagent used therein. for the purpose.
  • secretome secretome
  • high-quality T cells high-quality T cells
  • low-quality T cells T cells with relatively low proliferative potential
  • PCA principal component analysis
  • genes with a high principal component loading value of the first principal component are expressed in cells with a high PC1 score, that is, high-quality T cells, and genes with a low principal component loading value of the first principal component are expressed in It was found to be expressed in cells with low PC1 score, ie low quality T cells.
  • the present inventors used genes with a large absolute value of the principal component loading with respect to the first principal component as markers for quality evaluation of T cells, and used their expression as an index to determine the quality of T cells. We found that quality can be evaluated. Furthermore, since these genes are genes that are expressed relatively early after TCR stimulation of T cells, it was found that the quality of relatively young T cells can also be evaluated by using their expression as an index, and the present invention has been made. I came to complete it.
  • a method for evaluating T cells comprising: (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  • the detection step detects the expression product using a probe molecule that specifically binds to the expression product of at least one gene selected from the group consisting of the genes (I) to (III)
  • the method according to [1] which is a step.
  • the expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule specifically binds to the polypeptide
  • the method of [2] which is an antibody and/or an aptamer.
  • At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The method according to any one of [1] to [4], which is a gene.
  • At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The method according to any one of [1] to [5], which is a gene.
  • the expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule specifically binds to the polypeptide
  • the reagent of [6] which is an antibody and/or an aptamer.
  • At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III')
  • At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III')
  • [15] A method of treating or preventing cancer, comprising administering the pharmaceutical composition of [12] to a subject who has or is at risk of having cancer.
  • a method for treating or preventing cancer comprising administering T cells to a subject having cancer or at risk of having cancer, wherein A detection step of detecting the expression of at least one gene selected from the group consisting of the genes of ⁇ (III); an evaluation step of evaluating the quality of T cells using the expression of the gene as an index; A method, including (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  • a method for producing T cells comprising a detection step of detecting expression of at least one gene selected from the group consisting of the following genes (I) to (III) in the T cells; an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
  • a method of producing a T cell comprising: (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  • the present invention it is possible to provide a T cell evaluation method that can evaluate the quality of T cells using a new index, and a T cell evaluation reagent used for the method.
  • FIG. 3 is a scatter plot obtained by performing principal component analysis (PCA) on data for all 37,912 genes in 23 T cell lines.
  • FIG. 3 is a diagram showing the relationship between the rank of proliferation rate (proliferation rank) and the rank of PC1 score (PC1 rank) of 23 types of T cell lines.
  • the method for evaluating T cells of the present invention comprises a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in T cells; an evaluation step of evaluating the quality of T cells using the expression of the gene as an index; (hereinafter sometimes simply referred to as "the method of the present invention").
  • the T cell in the present invention means a cell expressing an antigen receptor called T cell receptor (TCR) on the surface, alpha beta ( ⁇ ) T cells and gamma delta ( ⁇ ) containing T cells.
  • TCR T cell receptor
  • T cells in the present invention include, for example, helper T cells, cytotoxic T cells, regulatory T cells, natural killer T cells, naive T cells, memory T cells (e.g., stem cell memory T cells (TSCM), central memory T cells). cells (TCM), effector memory T cells, etc.), antigen-stimulated memory T cells, or terminal effector T cells, combinations thereof, or subpopulations thereof.
  • helper T cells cytotoxic T cells
  • regulatory T cells e.g., regulatory T cells, natural killer T cells, naive T cells
  • memory T cells e.g., stem cell memory T cells (TSCM), central memory T cells).
  • TCM stem cell memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • TCM central memory T cells
  • Helper T cells are CD4-positive cells, and are further classified into Th1 cells, Th2 cells, Th17 cells, etc. according to the cytokines they express (e.g., J Allergy Clin. Immunol., 135(3): 626-635, 2012).
  • Th1 cells include, for example, cells expressing IFN- ⁇ , IL-2, TNF- ⁇ , and the like.
  • Th2 cells include, for example, cells expressing IL-4, IL-5, IL-6, IL-10, IL-13, and the like.
  • Th17 cells include, for example, cells expressing IL-17 or IL-6.
  • Cytotoxic T cells are CD8-positive cells, and like helper T cells, are further classified into Tc1 cells, Tc2 cells, etc. according to the cytokines they express.
  • Tc1 cells include cells expressing IFN- ⁇ , IL-2, TNF- ⁇ , and the like.
  • Tc2 cells include cells that express IL-4, IL-5, IL-6, IL-10, IL-13, and the like.
  • Regulatory T cells preferably include, for example, CD4(+) CD25(+) FoxP3(+) cells.
  • Naive T cells include, for example, CD4 (+) CD45RA (+) CD62L (+) CCR7 (+) cells, CD8 (+) CD45RA (+) CD62L (+) CCR7 (+) cells, CD4 (+) CCR7 ( +)CD45RA(+)CD95(-)CD45RO(-) cells or CD8(+)CCR7(+)CD45RA(+)CD95(-)CD45RO(-) cells are preferred.
  • Stem cell memory T cells include, for example, CD4 (+) CD45RA (+) CD62L (+) CCR7 (+) CD95 (+) cells, CD8 (+) CD45RA (+) CD62L (+) CCR7 (+) CD95 (+) ) cells, CD4(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+) cells or CD8(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+) cells are preferred. be done.
  • Central memory T cells include, for example, CD4 (+) CD45RA (-) CD62L (+) CCR7 (+) CD95 (+) cells, CD8 (+) CD45RA (-) CD62L (+) CCR7 (+) CD95 (+) ) cells, CD4(+)CCR7(+)CD45RA(-)CD45RO(+) cells, CD8(+)CCR7(+)CD45RA(-)CD45RO(+) cells, and the like are preferable.
  • effector memory T cells include preferably CD4(+) CCR7(-) CD45RA(-) CD45RO(+) cells or CD8(+) CCR7(-) CD45RA(-) CD45RO(+) cells.
  • Terminal effector T cells preferably include, for example, CD4(+)CD45RA(+)CD62L(-) cells or CD8(+)CD45RA(+)CD62L(-) cells.
  • T cells differentiated from pluripotent stem cells such as induced pluripotent stem cells (iPS cells) and embryonic stem cells (ES cells); T cells differentiated from somatic stem cells such as hematopoietic stem cells; It may be a T cell.
  • pluripotent stem cells such as induced pluripotent stem cells (iPS cells) and embryonic stem cells (ES cells)
  • ES cells differentiated from somatic stem cells such as hematopoietic stem cells; It may be a T cell.
  • T cells are isolated from peripheral blood mononuclear cells (PBMC: Peripheral Blood Monoclear Cells) isolated from peripheral blood using conventionally known methods or commercially available kits. It can be isolated.
  • PBMC peripheral blood mononuclear Cells
  • T cells include T cells that have not been artificially genetically modified (hereinafter sometimes referred to as “unmodified T cells”), as well as artificial genes in the unmodified T cells. Also included are “genetically modified T cells” that have been modified.
  • artificial genetic modification includes modification of T cell functions by gene transfer to T cells or gene editing of T cells. The artificial genetic modification may be modification of the gene itself possessed by T cells, deletion of the gene itself possessed by T cells, or modification resulting from the introduction of a foreign gene. A combination of two or more types may also be used.
  • the method of artificial genetic modification includes conventionally known methods and methods based thereon, and is not particularly limited.
  • Gene introduction into T cells includes, for example, the gene itself (DNA, mRNA, miRNA, antagomir, ODN (oligodeoxyribonucleotide), etc.) that modifies the function of T cells, or a vector into which the gene is inserted (lentiviral vector , ⁇ - or ⁇ -retroviral vectors, adenoviral vectors, adeno-associated viral vectors, herpes viral vectors, equine encephalitis viral vectors, non-viral vectors such as transposon vector piggyBac (registered trademark) to T cells
  • Gene editing of T cells includes, for example, gene editing of T cells using site-specific nucleases (meganuclease, zinc finger nuclease, TALEN, PPR (Pentatricpeptide repeat), CRISPR-Cas, etc.) (genome editing).
  • Examples of the gene that modifies the function of T cells include proteins secreted from T cells; fusion proteins containing proteins expressed on the surface of T cells and at least one intracellular signaling domain; proteins expressed on the surface of T cells; Fusion proteins comprising at least one co-stimulatory domain and at least one intracellular signaling domain include, more specifically, cell surface enzymes, cell adhesion factors, receptors (e.g., chimeric antigen receptors), and these and subunits that make up the
  • Detection process (target gene)
  • the expression of at least one gene selected from the group consisting of genes (I) to (III) below is detected in T cells.
  • a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added
  • the gene (I), the gene (II), and the gene (III) are independently of each other. Two or more types may be used.
  • the polypeptide encoded by any one of the genes (I) to (III) is preferably contained in the T cell-derived secretome.
  • secretome refers to proteins that are secreted from cells or that are exposed outside the cells through cell membranes, and include embodiments in which all or part of these proteins are endogenous to cells.
  • the gene whose expression is to be detected (hereinafter sometimes referred to as "target gene") is, if it is human-derived, typically "(I) any one of SEQ ID NOS: 1 to 77 A gene encoding a polypeptide comprising the amino acid sequence described in .
  • Tables 1 to 5 below show the sequence numbers and GenBank Accession Nos. of each amino acid sequence for the amino acid sequences set forth in SEQ ID NOS: 1 to 77. (GenBank No.); SEQ ID NO of a typical nucleotide sequence of a gene encoding a polypeptide consisting of each amino acid sequence, its open reading frame (ORF position), and GenBank Accession No.
  • Tables 1 to 5 show the origins from which each gene was obtained in the examples below, namely the following (a) to (d): (a) Genes mapped by transcriptome analysis in day0 samples (transcriptome day0) (b) genes mapped by transcriptome analysis in day6 samples (transcriptome day6); (c) genes mapped by transcriptome analysis in day14 samples (transcriptome day14); (d) genes encoding proteins identified by secretome analysis (secretome) are shown together with the "Origin data" column. Furthermore, the principal component loading of the first principal component of the expression level of each gene obtained in (a) to (d) above by the principal component analysis (PCA) of the following example is also included. show.
  • PCA principal component analysis
  • the present inventors have found that in high-quality T cells and low-quality T cells, there is a high correlation between the PC1 score obtained by principal component analysis (PCA) for gene expression levels and the quality of T cells.
  • PCA principal component analysis
  • Genes with high values of principal component loading of one principal component are expressed in cells with high PC1 score, i.e. high-quality T cells, and genes with low values of principal component loading of the first principal component have low PC1 scores. cells, namely low quality T cells.
  • the described genes encoding polypeptides consisting of amino acid sequences represented by SEQ ID NOs: 26 to 77 have a high contribution rate to the first principal component (specifically, the main The absolute value of the component load is 0.9 or more) and encodes a protein other than a transmembrane protein.
  • homologous genes e.g., counterpart genes in organisms other than humans
  • (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77
  • the target gene can also be the target gene.
  • the nucleotide sequence of a gene can be mutated in nature (that is, non-artificially) due to mutation thereof, etc., such natural mutants can also be used as the target gene in the present invention. can.
  • a person skilled in the art can modify the nucleotide sequence and modify the function, although it is different from the amino acid sequence that codes for it. Since proteins can also be prepared, such variants may also be used as the target gene.
  • amino acid sequence in which amino acid residues are substituted, deleted, inserted, and/or added means an amino acid sequence in which amino acid residues are substituted, deleted, inserted, or added, or It shows that the amino acid sequence is a combination of two or more of Further, “plurality” means, for example, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 is preferably an integer of, but not limited to.
  • One or more amino acid residues is preferably 1 to 10 amino acid residues, more preferably 1 to 5, still more preferably 1 to 3, particularly preferably 2 or less. is.
  • (IV) hybridizes under stringent conditions with the complementary strand of the nucleotide sequence encoding the amino acid sequence set forth in any one of SEQ ID NOS: 1 to 77 Also included are “nucleotide sequences”. Hybridization reactions are typically performed under stringent conditions to isolate homologous genes.
  • stringent conditions means that the washing operation of the membrane after hybridization is performed at a high temperature in a low-salt solution. 2 ⁇ SSC) in 0.5% (w/v) SDS solution at 60° C. for 20 minutes.
  • hybridization can be carried out, for example, according to the method described in the instructions attached to the known ECL direct DNA/RNA labeling/detection system (manufactured by GE Healthcare Bioscience).
  • ECL direct DNA/RNA labeling/detection system manufactured by GE Healthcare Bioscience.
  • these conditions are only examples, and the required stringency can be achieved by appropriately combining DNA concentration, DNA length, hybridization reaction time, and the like.
  • proteins encoded by homologous genes obtained by such methods usually have a high degree of identity with the amino acid sequence set forth in any one of SEQ ID NOS: 1-77. Therefore, in the embodiment of the target gene according to the present invention, "(III) encodes a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77 Also includes “genes”. Amino acid sequence identity can be determined, for example, using the BLASTP program (Altschul et al., J. Mol. Biol., 215, 1990, p. 403-410).
  • identity with the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77 is typically preferably 70% or more, more preferably 75% or more, 80% or more, 85% or more. % or more, 90% or more, 95% or more (eg, 96% or more, 97% or more, 98% or more, 99% or more).
  • the target gene is preferably at least one gene selected from the group consisting of the following genes (I') to (III') among the genes listed in Tables 1 to 5. Since the proteins expressed from these genes are exposed outside the membrane as membrane proteins of T cells, after detection, the membrane proteins are used to produce high-quality (or low-quality) T cells. This is because capture, separation, or purification is also possible.
  • (I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27) (II') ) Substitution or deletion of one or more amino acid residues in the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27, and 44 (more preferably SEQ ID NOs: 5, 15, and 27) Gene (III') SEQ ID NOS: 5, 7, 15, 27, and 44 (more preferably SEQ ID NOS: 5, 15, and 27) encoding a polypeptide comprising deleted, inserted, and/or added amino acid sequences
  • a gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence described in any one of the above, wherein in the group consisting of the genes (I') to (III'), (I The gene of '), the gene of (II'), and the gene of (III') may be each independently of one type or two
  • the target gene according to the present invention from the viewpoint of being able to evaluate the quality of younger T cells (less days after TCR stimulation), among the genes shown in Tables 1 to 5 above, "derived data" Genes whose column is "transcriptome day 0" are preferred.
  • detecting gene expression includes both detection of the presence or absence of gene expression and detection of the degree of expression.
  • the expression of the target gene is detected.
  • the gene expression level can be grasped as an absolute amount or as a relative amount. When grasping the relative amount, for example, it can be judged by comparing with the gene expression level of a prepared standard sample.
  • a "standard sample” refers to a sample for which it has been specified in advance whether or not the target gene is expressed, and if so, the amount of expression. For example, T cells that have been previously identified as having good or bad quality can be used as the standard sample.
  • “expression of a gene” means both transcription and translation of a gene. Therefore, “detection of gene expression” in the present invention includes detection at the transcription level (mRNA level) and detection at the translation level (protein level) (that is, detection of the polypeptide encoded by the gene). both are included.
  • splicing occurs in which introns in the pre-mRNA are removed and the exons before and after are recombined (splicing). In some cases, this produces different mature mRNAs. In turn, various proteins are translated thereby. Various mRNAs and proteins resulting from such splicing differences are called "splicing variants". Therefore, detection of gene expression in the present invention includes detection of the splicing variant as long as it is specifically recognized by the following probe molecule.
  • the detection step according to the method of the present invention at least one selected from the group consisting of the genes (I) to (III) (preferably the genes (I') to (III'))
  • the expression product of a gene is detected using a probe molecule that specifically binds to said expression product.
  • the expression product of the target gene is mRNA transcribed from the target gene (hereinafter sometimes referred to as "target polynucleotide").
  • the target polynucleotide also includes cDNA using the mRNA as a template.
  • Detection methods include, for example, Northern blotting, dot blotting, and RNase protection assay using oligonucleotide probes designed to hybridize to appropriate positions in the base sequence of the target polynucleotide as the probe molecule. , DNA microarray analysis, in situ hybridization, and the like to detect the target polynucleotide.
  • the oligonucleotide probe one labeled with a labeling substance is used, a signal corresponding to the labeling substance is detected, and the detected signal amount is the amount of the target polynucleotide (i.e., expression level of the target gene).
  • labeling substances at this time include FITC (fluorescein isothiocyanate), FAM (fluorescein amidite), DEAC (7-(diethylamino)coumarin), R6G (rhodamine 6G), Texas Red, Cy5, and BODIPY FL (trade name).
  • luminescence substances such as luminol, luciferin, and lucigenin.
  • the "signal” includes coloration (coloration), reflected light, luminescence, quenching, fluorescence, radiation from a radioactive isotope, and the like, and in addition to those that can be confirmed with the naked eye, depending on the type of signal It also includes those that can be confirmed by measuring methods and equipment.
  • oligonucleotide primers designed to sandwich appropriate positions in the target polynucleotide are used, PCR method, RT-PCR method, TRC (Transcription Reverse Transcription Concerned) method, NASBA (Nucleic Acid Sequence-Based Amplification) method, TMA (Transcription-Mediated Amplification) method, or the like may also be used to amplify and detect the target polynucleotide.
  • a signal (fluorescence) obtained by intercalating the obtained amplification product with a fluorescent dye e.g., ethidium bromide, SYBR Green (trade name), SYTO63 (trade name), etc.
  • a fluorescent dye e.g., ethidium bromide, SYBR Green (trade name), SYTO63 (trade name), etc.
  • the detected signal amount fluorescence intensity
  • detection may be performed in combination with the oligonucleotide probe (double dye probe method, etc.).
  • the amount of the target polynucleotide may be determined by directly subjecting a sample containing the target polynucleotide to a sequencer for analysis.
  • oligonucleotide probes and oligonucleotide primers can be designed by a person skilled in the art based on the sequence of the target polynucleotide by a known method or a method based thereon.
  • the length of the oligonucleotide probe and oligonucleotide primer are each independently preferably at least 15 bases, usually 15 to 100 bases, preferably 17 to 30 bases, more preferably 20-25 bases.
  • the oligonucleotide probes and oligonucleotide primers can be synthesized by, for example, a commercially available oligonucleotide synthesizer.
  • nucleotides deoxyribonucleotides and/or ribonucleotides
  • PNA polyamide nucleic acid
  • LNA locked nucleic acid
  • ENA registered trademark, 2'- O,4'-C-Ethylene-bridged nucleic acids
  • the expression product of the target gene is a polypeptide encoded by the target gene (hereinafter sometimes referred to as "target polypeptide"). More specifically, it is a polypeptide encoded by any one of the genes (I) to (III), that is, selected from the group consisting of the following polypeptides (i) to (iii) is a polypeptide that is (i) a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 1-77; (ii) one or more amino acid residues in the amino acid sequence set forth in any one of SEQ ID NOS: 1-77; is a substituted, deleted, inserted, and / or added amino acid sequence (iii) an amino acid sequence having 70% or more identity with the amino acid sequence set forth in any one of SEQ ID NOS: 1 to 77
  • polypeptides (i) to (iii) are each independently one species alone, may be two or more.
  • polypeptides (i) to (iii) include polypeptides encoded by any one of the genes (I') to (III'), that is, the following (i' ) to (iii') are preferably selected from the group consisting of polypeptides.
  • polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27);
  • SEQ ID NO: 5 , 7, 15, 27, and 44 (more preferably SEQ ID NOS: 5, 15, and 27), wherein one or more amino acid residues are substituted, deleted, inserted, and/or an amino acid according to any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27), comprising a polypeptide (iii') comprising an added amino acid sequence
  • a polypeptide comprising an amino acid sequence having 70% or more identity with a sequence.
  • polypeptides (i') to (iii') each independently It may be one type alone or two or more types.
  • these target polypeptides to be detected are preferably the above secretomes (T cell-derived secretomes).
  • detection methods for detection at the translation level include detection using anti-polypeptide antibodies and/or aptamers, and detection using labeled amino acids.
  • Detection methods for detection at the translation level include, for example, antibodies that specifically bind to the target polypeptide (hereinafter sometimes referred to as “anti-polypeptide antibodies”) and/or aptamers (hereinafter , In some cases, simply referred to as "aptamer”), immune cell staining method, imaging cytometry, flow cytometry, ELISA (Enzyme-Linked Immuno Sorbent Assay) method, radioimmunoassay, immunoprecipitation method, immunoblotting (Western blotting method etc.), methods of detection using antibodies such as antibody arrays and in vivo imaging (immunological techniques); methods of detection using aptamers instead of the antibodies.
  • the immunological method uses an enzyme immunoassay device such as AIA-900 (manufactured by Tosoh Corporation) or a chemiluminescent enzyme immunoassay device such as AIA-CL2400 (manufactured by Tosoh Corporation) after preparing necessary reagents. can be done automatically.
  • AIA-900 manufactured by Tosoh Corporation
  • AIA-CL2400 manufactured by Tosoh Corporation
  • an “antibody” may be a polyclonal antibody, a monoclonal antibody, or a functional fragment of an antibody.
  • “Antibody” also includes all classes and subclasses of immunoglobulins.
  • a “functional fragment” of an antibody refers to a portion (partial fragment) of an antibody that specifically recognizes the polypeptide encoded by the target gene in the present invention. Specifically, Fab, Fab′, F(ab′) 2 , variable region fragment (Fv), disulfide-bonded Fv, single-chain Fv (scFv), sc(Fv) 2 , diabody, multispecific antibodies, short-chain antibodies, and polymers thereof.
  • the anti-polypeptide antibody according to the present invention is a polyclonal antibody
  • an immunized animal is immunized with an antigen (target polypeptide, its partial peptide, or cells expressing these, etc.), and the antiserum is obtained by conventional means ( Salting out, centrifugation, dialysis, column chromatography, etc.) can be appropriately purified and obtained.
  • Monoclonal antibodies can also be produced by the hybridoma method or recombinant DNA method.
  • the hybridoma method includes, for example, Kohler and Milstein's method (Kohler & Milstein, Nature, 256:495 (1975)), and the recombinant DNA method includes, for example, the DNA encoding the anti-polypeptide antibody. It is cloned from B cells or the like, incorporated into an appropriate vector, introduced into host cells (mammalian cell lines, E. coli, yeast cells, insect cells, plant cells, etc.), and the anti-polypeptide antibody is produced as a recombinant antibody. (For example, P. J. Delves, Antibody Production: Essential Techniques, 1997 WILEY; P. Shepherd and C. Dean Monoclonal Antibodies, 2000 OXFORD UNIVERSITY PRESS; Vandamme AM et al., Eur.J. Biochem. 192:767-775 (1990)).
  • the anti-polypeptide antibody and the aptamer are each labeled with a labeling substance, the signal corresponding to the labeling substance is detected, and the amount of the detected signal is calculated as the amount of the target polypeptide. (that is, the expression level of the target gene).
  • the labeling substance at this time is not particularly limited as long as it can bind to the anti-polypeptide antibody or aptamer and can be detected by a chemical or optical method.
  • fluorescent substances such as fluorescein isothiocyanate (FITC) and rhodamine isothiocyanate (RITC)
  • enzymes such as peroxidase, ⁇ -D-galactosidase, microperoxidase, horseradish peroxidase (HRP) and alkaline phosphatase, and radioactive substances.
  • the anti-polypeptide antibody and/or aptamer When the anti-polypeptide antibody and/or aptamer is used, a method of using a secondary antibody bound with the labeling substance, or a method of using a polymer binding the secondary antibody and the labeling substance. Indirect detection methods such as can also be used.
  • the "secondary antibody” is an antibody that exhibits specific binding to the anti-polypeptide antibody and/or aptamer.
  • an anti-rabbit IgG antibody can be used as the secondary antibody.
  • Labeled secondary antibodies that can be used for antibodies derived from various species such as rabbits, goats, and mice are commercially available. Antibodies can be selected and used.
  • protein G for example, derived from Streptococcus spp.
  • protein A for example, derived from Staphylococcus aureus
  • protein L derived from Peptostreptococcus magnus
  • Detection with labeled amino acids include, for example, a method of culturing T cells in a medium supplemented with a labeled amino acid and detecting a target polypeptide labeled with the labeled amino acid.
  • T cell labeling medium refers to a medium containing at least labeling amino acids for culturing and labeling T cells.
  • essential amino acids for T cell proliferation refers to amino acids essential for T cell proliferation, more specifically, valine, isoleucine, leucine, methionine, lysine, phenylalanine, tryptophan, threonine, histidine.
  • the T cell labeling medium contains a labeled amino acid in which the amino acid is labeled instead of at least one of the essential amino acids, that is, does not contain at least one of the essential amino acids and the essential amino acid that is not contained Contains labeled amino acids, where the amino acids are labeled amino acids.
  • "Containing no essential amino acids” means that the T cell labeling medium does not substantially contain the essential amino acids. More specifically, the content of the essential amino acids in the T cell labeling medium is is 1 ⁇ 10 ⁇ 3 mmol/L or less, preferably less than 1 ⁇ 10 ⁇ 4 mmol/L.
  • the essential amino acids excluded from the T cell labeling medium may be any one of the above or a combination of two or more of them. Among them, methionine is preferred from the viewpoint of efficient incorporation into the protein synthesis pathway.
  • the "labeled amino acid” refers to an essential amino acid partially modified while maintaining the structure of the essential amino acid, and detectable based on the modification.
  • the above-mentioned “detectable” includes those that can be directly confirmed by visual observation such as coloring (color development), quenching, reflected light, luminescence, and fluorescence, as well as those that can be confirmed by a specific measuring method or measuring device.
  • labeled amino acids examples include isotope-labeled amino acids in which some of the atoms of the amino acid are substituted with pulse stable isotopes (e.g., 2 H, 13 C, 15 N); Examples include structural analogs of amino acids modified by modifying groups such as groups, and corresponding to the essential amino acids, any one of them or a combination of two or more of them good.
  • the labeling amino acid contained in the T cell labeling medium instead of methionine may be, for example, one of the atoms constituting methionine.
  • HPG L-Homopropargylglycine
  • HOG L-homoallylglycine
  • the content of the labeled amino acids includes the T cell uptake efficiency, the culture efficiency, the T cell It can be determined as appropriate in consideration of the toxicity etc., and is not particularly limited. Preferably, it is 0.001 to 0.02 mmol/L. More specifically, for example, when the labeled amino acid is AHA, it is preferably 0.0005 to 0.03 mmol/L, more preferably 0.001 to 0.03 mmol/L. More preferably 0.002 to 0.03 mmol/L, even more preferably 0.005 to 0.02 mmol/L.
  • the cells tend to weaken or die, whereas when the content is less than the lower limit, the polypeptide labeled with the labeled amino acid is secreted or exposed outside the cell. As a result, the number of cells to be detected tends to decrease, and the detection accuracy of the polypeptide tends to decrease.
  • the T cell labeling medium further contains common ⁇ -chain family cytokines.
  • a "common ⁇ chain family cytokine” refers to a cytokine that acts via a receptor containing a common ⁇ chain ( ⁇ subunit). More specifically, interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 7 (IL-7), interleukin 9 (IL-9), interleukin 15 (IL-15) , and interleukin 21 (IL-21), any one of which may be used in combination.
  • interleukin 7, interleukin 15, interleukin 21, and combinations of two or more of these are more preferable from the viewpoint of better maintenance of cell survival and growth.
  • the content of the common ⁇ -chain family cytokine (when there are two or more common ⁇ -chain family cytokines, the total content thereof; the same shall apply hereinafter) is determined according to the properties of T cells, etc. Although it is not limited to this, it is preferably 1 to 1000 ng/mL, more preferably 5 to 100 ng/mL, and 5 to 20 ng/mL. It is even more preferable to have When the content of the common ⁇ -chain family cytokine exceeds the upper limit, the effect of containing the common ⁇ -chain family cytokine tends to decrease or cell proliferation tends to be inhibited. The number of cells that secrete or extracellularly expose a polypeptide labeled with a labeled amino acid tends to decrease, resulting in a decrease in the detection accuracy of the polypeptide.
  • the T cell labeling medium preferably does not contain serum or, if it does contain serum, is dialyzed serum from which the essential amino acids have been removed.
  • the T cell labeling medium may also contain cytoprotective additives such as serum albumin and ITS (insulin-transferrin-sodium selenite); scaffolding agents such as ECM (extracellular matrix) components.
  • T cell labeling media or their basal media are not particularly limited as long as they do not contain the desired essential amino acids, and are DMEM (Dulbecco's Modified Eagle Medium), RPMI (Roswell Park Memorial Institute Medium), ⁇ MEM ( ⁇ -modified basal medium such as Eagle minimum essential medium); AIM V TM medium (manufactured by Thermo Fisher Scientific) which is a serum-free medium exclusively for T cells; PRIME-XV T cell Expansion XSFM (manufactured by Fujifilm Wako Pure Chemical) or a commercially available medium for T cells can be used as appropriate.
  • DMEM Disbecco's Modified Eagle Medium
  • RPMI Roswell Park Memorial Institute Medium
  • ⁇ MEM ⁇ -modified basal medium such as Eagle minimum essential medium
  • AIM V TM medium manufactured by Thermo Fisher Scientific
  • PRIME-XV T cell Expansion XSFM manufactured by Fujifilm Wako Pure Chemical
  • a commercially available medium for T cells
  • T cells are cultured in the T cell labeling medium.
  • the labeled amino acid is taken up by T cells, and a polypeptide labeled with the labeled amino acid (T cell-derived polypeptide) is secreted from the T cells or exposed outside the cells.
  • the culture method in the labeling step is not particularly limited, and known T cell proliferation culture conditions can be appropriately adopted, and the characteristics and concentration of T cells can be taken into consideration and adjusted as appropriate.
  • the culture temperature is 35 to 37.5° C., preferably 36 to 37° C.
  • the culture time is 2 to 48 hours, preferably 8 to 24 hours.
  • the culture apparatus for the labeling step is not particularly limited, and examples thereof include plates, dishes, columns, flasks, culture bags, and the like. Ejection means (sensors, valves, pumps, tanks, etc.) may also be provided.
  • the T cell-derived polypeptide that is labeled with the labeling amino acid and is secreted or exposed outside the cell is detected.
  • the detection step A may be performed simultaneously (in real time) with the labeling step.
  • a detection method in the detection step A a detection method suitable for the type of the labeled amino acid can be appropriately employed.
  • Such a detection method is not particularly limited, and a known method or a method based thereon can be appropriately employed.
  • the labeled amino acid is an isotope-labeled amino acid or a structural analogue of an amino acid
  • the medium after the labeling step or during the labeling step that is, during the culture
  • gas isotope ratio mass spectrometry By performing mass spectrometry such as infrared spectroscopy, liquid chromatography mass spectrometry (LC-MS/MS), a signal corresponding to the labeled amino acid is detected, and the amount of the detected signal is the amount of the target polypeptide (i.e. , expression level of the target gene).
  • the labeled amino acid when it is a structural analogue of an amino acid, it can be detected by staining the modified group specifically.
  • the modifying group is an azide group (such as AHA)
  • the azide group can be stained with a fluorescent reagent such as Click-iT TM Alexa Fluor 488 sDIBO Alkyne (manufactured by Thermo Fisher Scientific).
  • a fluorescent reagent such as Click-iT TM Alexa Fluor 488 sDIBO Alkyne (manufactured by Thermo Fisher Scientific).
  • a signal color development, fluorescence, etc.
  • the amount of the detected signal is the amount of the target polypeptide (i.e., the amount of the target gene). expression level).
  • detection methods for detection at the translation level include, for example, a method of recognizing and detecting the sugar chain of the target polypeptide. Such methods include, for example, methods using commercially available reagents such as Click-iT TM (manufactured by Thermo Fisher Scientific). The detection method for detection at the translation level may be used in combination with other methods as appropriate.
  • detection of the expression of the gene is preferably detection at the translation level (detection of the target polypeptide) from the viewpoint of convenience. Methods that detect polypeptides are preferred.
  • the quality of T cells is evaluated using the presence or absence or the expression level of the target gene detected in the detection step as an index.
  • the quality of T cells to be evaluated in the present invention includes proliferative ability and cancer cytotoxic activity.
  • the method of the present invention is preferable as a method for evaluating the proliferative ability of T cells. More specifically, the term "proliferative ability" indicates the ability to activate T cells and increase cells having cytotoxic activity in vitro or in vivo by TCR stimulation or antigen stimulation.
  • T cells can be confirmed, for example, by stimulating the TCR of T cells with TCR-stimulating magnetic beads by the method shown in the Examples below and comparing the cell count before stimulation to the cell count 14 days after stimulation. can, but is not limited to.
  • genes with a positive principal component loading value have a higher proliferation ability as the expression level of the gene increases. That is, it can be evaluated that the quality is good.
  • a gene with a negative principal component loading value can be evaluated as having a lower proliferation ability, ie, a lower quality, as the expression level of the gene increases.
  • the evaluation criteria for the quality of the T cells may be appropriately set according to the purpose, and are not particularly limited.
  • the detection step if even a little expression of the target gene is detected according to the expression level of the target gene (transcription level or translation level of the target gene), that is, the target polynucleotide or the target polypeptide If even a small amount is detected, the proliferative potential of T cells may be evaluated as high (or low), and if the expression level of the target gene above a certain threshold is detected, the proliferative potential of T cells is high (or low). Further, the degree of T cell proliferation ability may be evaluated according to the expression level of the target gene.
  • the expression level of the target gene transcription level or translation level of the target gene
  • a certain value or more, or the expression level of the target gene relative to the expression level of the target gene detected in a standard sample such as T cells that have been specified in advance to have high (or low) proliferative ability If the signal amount is 4 SD or more, 3 SD or more, or 2 SD or more of the average value of , the proliferative ability is high (or low), that is, the quality may be evaluated as good (or bad).
  • the T cell evaluation reagent of the present invention is a reagent (composition) for use in the T cell evaluation method of the present invention, and is at least one gene selected from the group consisting of the following genes (I) to (III).
  • a reagent containing a probe molecule that specifically binds to an expression product of a gene of a species hereinafter sometimes simply referred to as "the reagent of the present invention”).
  • the genes (I) to (III) and the expression products of the genes are as described in the method of the present invention, including preferred embodiments thereof.
  • probe molecules related to the reagent of the present invention include oligonucleotide probes, oligonucleotide primers, anti-polypeptide antibodies, and aptamers mentioned in the method of the present invention described above. in the method of the present invention.
  • the probe molecule may be one of these or a combination of two or more of them. Among them, the anti-polypeptide antibody and/or the aptamer are preferable, and the anti-polypeptide antibody is more preferable.
  • the reagent of the present invention may be liquid or solid such as powder. Additional ingredients may be included.
  • the kit for evaluating T cells of the present invention is a kit for use in the method of evaluating T cells of the present invention, and contains at least one gene selected from the group consisting of genes (I) to (III). It is a kit comprising a probe molecule that specifically binds to an expression product (hereinafter sometimes simply referred to as "the kit of the present invention").
  • the probe molecule according to the kit of the present invention is preferably the reagent of the present invention.
  • the kit of the present invention also contains a buffer, a solution for dilution or suspension such as physiological saline; a reagent for extracting and/or purifying DNA or protein; a blocking agent; a chelating agent; The labeling substance; the fluorescent dye; reagents necessary for the detection step; reagents such as pH adjusters; standard samples; instructions for use;
  • the present invention provides high-quality T cells obtained by the above T-cell evaluation method and pharmaceutical compositions containing the high-quality T cells as active ingredients.
  • the pharmaceutical composition containing T cells as an active ingredient of the present invention can be used to treat cancer patients.
  • the pharmaceutical composition of the present invention can be produced by a method commonly used in the field of formulation technology, such as the method described in the Japanese Pharmacopoeia.
  • the pharmaceutical composition of the present invention may contain pharmaceutically acceptable additives. Examples of such additives include cell culture media, physiological saline, and suitable buffers (eg, phosphate buffers).
  • the pharmaceutical composition of the present invention can be produced by suspending T cells in physiological saline, an appropriate buffer (eg, phosphate buffer), or the like. It is preferable to contain, for example, 1 ⁇ 10 7 cells or more in a single dose so as to achieve the desired therapeutic effect. It is more preferably 1 ⁇ 10 8 or more, still more preferably 1 ⁇ 10 9 or more.
  • the content of cells can be appropriately adjusted in consideration of the sex, age, weight, condition of the affected area, condition of cells, etc. of the subject.
  • the pharmaceutical composition of the present invention may contain dimethylsulfoxide (DMSO), serum albumin, and the like for the purpose of protecting cells.
  • DMSO dimethylsulfoxide
  • Antibiotics and the like may be added for the purpose of preventing bacterial contamination, and vitamins, cytokines, and the like may be included for the purpose of promoting activation and differentiation of cells.
  • other pharmaceutically acceptable components e.g., carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological saline, etc. It may be included in the pharmaceutical composition of the present invention.
  • a pharmaceutical composition containing the T cells of the present invention as an active ingredient can be cryopreserved.
  • the temperature is not particularly limited as long as it is suitable for cell preservation. Examples include -20°C, -80°C and -120 to -196°C.
  • cells can be stored in suitable containers such as vials. Manipulations to minimize the risk of cell damage during freezing and thawing of T cells are well known to those of skill in the art.
  • T cells are collected from the culture medium, washed with buffer or culture medium, counted, concentrated by centrifugation, etc., and placed in a freezing medium (e.g., containing 10% DMSO). culture solution), and cryopreserved.
  • T cells can be made into a single lot by combining cells cultured in multiple culture vessels.
  • the pharmaceutical composition containing T cells of the present invention contains, for example, 5 ⁇ 10 4 to 9 ⁇ 10 10 T cells per container such as a vial. It can be changed according to the route or the like.
  • Examples of administration routes of the pharmaceutical composition containing the T cells of the present invention include infusion, intratumoral injection, arterial injection, portal vein injection, intraperitoneal administration, and the like.
  • the administration route is not limited to this as long as the T cells, which are the active ingredients in the pharmaceutical composition of the present invention, are delivered to the affected area.
  • Dosage schedules can be single doses or multiple doses. For the period of multiple administrations, for example, a method of repeating administration once every 2 to 4 weeks, a method of repeating administration once every six months to a year, or the like can be adopted.
  • the sex, age, body weight, disease state, etc. of the target patient can be taken into consideration.
  • cancer patients from whom T cells are collected or The non-cancer patient preferably has the same HLA type as the cancer patient to whom the pharmaceutical composition of the present invention is administered for cancer treatment. Moreover, it is more preferable that the cancer patient to whom the pharmaceutical composition of the present invention is administered and the cancer patient from whom T cells are collected are the same person. That is, cancer treatment by autologous transplantation is more preferable than allogeneic transplantation.
  • the pharmaceutical composition of the present invention is used for prevention or treatment of cancer.
  • Cancers include ovarian cancer, hepatoblastoma, hepatocellular carcinoma, gastric cancer, esophageal cancer, pancreatic cancer, renal cell carcinoma, breast cancer, malignant melanoma, non-small cell lung cancer, cervical cancer, and glioblastoma. including, but not limited to, blastoma, prostate cancer, neuroblastoma, chronic lymphocytic leukemia, papillary thyroid cancer, colorectal cancer, and B-cell non-Hodgkin's lymphoma.
  • the timing of administration of the pharmaceutical composition and cancer vaccine of the present invention is not limited,
  • the pharmaceutical composition of the present invention and cancer vaccine may be administered to the subject at the same time or at different times.
  • the pharmaceutical composition of the present invention and cancer vaccine may be formulated separately, or may be a combination drug in which both are mixed.
  • the dosage of the pharmaceutical composition and cancer vaccine of the present invention may conform to the dosage used clinically, and can be appropriately selected depending on the disease, administration subject, administration route, combination with drugs, and the like.
  • the present invention provides a method for treating or preventing cancer, comprising administering T cells to a subject who has cancer or is at risk of having cancer, wherein the T cells have the following (I A detection step of detecting expression of at least one gene selected from the group consisting of genes from ) to (III); an evaluation step of evaluating the quality of T cells using the expression of the gene as an index; A method ((I) a gene encoding a polypeptide comprising an amino acid sequence according to any one of SEQ ID NOS: 1-77, comprising , gene (III) encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted and/or added amino acid according to any one of SEQ ID NOs: 1 to 77 A gene encoding a polypeptide comprising an amino acid sequence having greater than 70% identity with the sequence.).
  • a subject in the present invention is a vertebrate.
  • the vertebrate is a mammal.
  • Such mammals include, but are not limited to, farm animals (eg, cows), sport animals, pet/companion animals (eg, cats, dogs, and horses), primates, mice, and rats.
  • the mammal is human.
  • the present invention provides a method for producing T cells, comprising a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in the T cells; an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
  • a method for producing T cells ((I) a gene encoding a polypeptide comprising an amino acid sequence according to any one of SEQ ID NOS: 1-77 (II) according to any one of SEQ ID NOS: 1-77
  • Any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence of A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence described in .).
  • T cells Twenty-four types of T cell lines shown in Table 11 below were thawed and placed in medium A (15% (w/v) fetal bovine serum (FBS, BioWest) in a 96-well plate. , 5 ng/mL interleukin 7 (IL-7), and 5 ng/mL interleukin 15 (IL-15) added to ⁇ MEM ( ⁇ modified Eagle minimal essential medium)) at 1 ⁇ 10 6 cells/well, respectively. sown.
  • medium A 15% (w/v) fetal bovine serum (FBS, BioWest) in a 96-well plate.
  • IL-7 interleukin 7
  • IL-15 interleukin 15
  • T cell receptor (TCR) stimulation Magnetic beads (Dynabeads Human T-Activator CD3/CD28 for T Cell Expansion and Activation (manufactured by Thermo Fisher Scientific) are used to stimulate the TCR of the seeded T cells. , and cultured for 14 days under conditions of 5% CO 2 and 37° C. while replacing the medium with medium A as appropriate.
  • T cells of the same strain as the T cells classified into high-quality T cells (12 types) and low-quality T cells (12 types) in 1 (3) above TCR unstimulated
  • TCR unstimulated For each, stimulate the TCR using TCR-stimulating magnetic beads in the same manner as in 1 (1) to (2) above, and replace the medium with medium A as appropriate under the conditions of 5% CO 2 and 37 ° C. cultured.
  • the genes (sequence data) decoded in (4) and (3) were mapped to the human genome sequence using TopHat2 (JOHNS HOPKINS University) and Bowtie2 (JOHNS HOPKINS University).
  • TopHat2 JOHNS HOPKINS University
  • Bowtie2 JOHNS HOPKINS University
  • BUILD GRCh38 published by NCBI (National Center for Biological Information) was used.
  • the expression level for each gene was determined in units of FPKM (fragments per kilobase of exon per million reads mapped) from the number of reads of each gene decoded by Cufflinks (University of Washington).
  • T cells TCR unstimulated
  • T cells classified into high-quality T cells (12 types) and low-quality T cells (12 types) in 1 (3) above Each TCR was stimulated using TCR-stimulating magnetic beads in the same manner as in 1 (1) to (2) above, and cultured under the conditions of 5% CO 2 and 37° C. while exchanging the medium with medium A as appropriate. .
  • ⁇ MEM (-Met) medium supplemented with (insulin-transferrin-sodium selenite) supplement (x1) 50 ⁇ g/mL ascorbic acid, 5 ng/mL IL-7, and 5 ng/mL IL-15, 37 °C and 5% CO2 for 24 hours.
  • PCA Principal component analysis From (4) of the transcriptome analysis in 2 above and (5) of the secretome analysis in 3 above, the following (a) to (d): (a) 12,829 genes mapped by transcriptome analysis in day0 samples (transcriptome day0) and their expression levels, (b) 12,270 genes mapped by transcriptome analysis in day6 samples (transcriptome day6) and their expression levels; (c) 12,013 genes mapped by transcriptome analysis in day14 samples (transcriptome day14) and their expression levels, (d) Data on 800 genes (secretome) encoding proteins identified by secretome analysis and their expression levels were obtained. However, the numbers of genes in (a) to (d) do not include low-expressed or non-expressed genes.
  • PCA principal component analysis
  • the scatter diagram obtained by the PCA is shown in Figure 1.
  • the first principal component (PC1) score was high in high-quality T cells (cells No. 1-12), and the PC1 score was low in low-quality T cells (cells No. 13-21, 23, 24).
  • Table 12 below and FIG. 2 show the relationship between the descending order (proliferation order) of the 23 types of proliferation rate, the PC1 score (PC1), and the descending order (PC1 order).
  • the correlation coefficient (R) between the T cell proliferation rank and the PC1 rank is 0.9615, indicating a high correlation between the cell quality (in this case, proliferative capacity) and the PC1 score. was accepted.
  • the variable with the high value of the principal component loading of the first principal component in this case, the variable is the expression level of each gene
  • a variable that contributes positively to the first principal component and has a low (negative value) value of the principal component loading of the first principal component contributes negatively to the first principal component.
  • genes with a high principal component loading value of the first principal component are expressed in cells with a high PC1 score, that is, high-quality T cells, and genes with a low principal component loading value of the first principal component are expressed in It was found to be expressed in cells with low PC1 score, ie low quality T cells.
  • the principal component loading of the first principal component was determined for each gene expression level obtained in (a) to (d) above.
  • the contribution rate to the first principal component is high (specifically, the absolute value of the principal component loading for the first principal component of the gene expression level is 0.9 or more) and genes encoding transmembrane proteins are the genes shown in Tables 1 and 2 above.
  • the contribution rate to the first principal component is high (specifically, the absolute value of the principal component loading amount with respect to the first principal component of the gene expression level is 0.9 or more), and other than transmembrane proteins
  • Genes encoding proteins are those shown in Tables 3-5 above. In Tables 1 to 5, for each gene, the categories (a) to (d) above are shown in the column of "origin data", respectively, and the principal component loadings for the first principal component obtained above are also included. indicated.
  • ITGB7, BMPR2, and LGALS8 can be said to be markers capable of evaluating the quality of T cells in the state after cell stimulation with TCR.
  • ALCAM, BMPR2, and TP53I3 have absolute values of correlation coefficients of 0.6 or more, and these three proteins can be said to be particularly preferred embodiments of markers capable of evaluating the quality of T cells.
  • the present invention it is possible to provide a T cell evaluation method that can evaluate the quality of T cells using a new index, and a T cell evaluation reagent used for the method.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Sustainable Development (AREA)
  • Plant Pathology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A T cell evaluation method that comprises a detection step for detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in T cells, and an evaluation step for evaluating the qualities of the T cells with the use of the expression of the gene as an indicator. (I) A gene encoding a polypeptide containing an amino acid sequence represented by any one of SEQ ID NOS: 1-77. (II) A gene encoding a polypeptide containing an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted and/or added in an amino acid sequence represented by any one of SEQ ID NOS: 1-77. (III) A gene encoding a polypeptide containing an amino acid sequence having 70% or more identity with an amino acid sequence represented by any one of SEQ ID NOS: 1-77.

Description

T細胞の評価方法及びT細胞評価用の組成物Method for evaluating T cells and composition for evaluating T cells
 本発明は、T細胞の評価方法、T細胞評価用の組成物、T細胞の評価方法により得られた高品質T細胞を含有する医薬組成物及び該医薬組成物を用いる、がんの予防または治療方法等に関する。 The present invention provides a method for evaluating T cells, a composition for evaluating T cells, a pharmaceutical composition containing high-quality T cells obtained by the method for evaluating T cells, and cancer prevention or treatment using the pharmaceutical composition. It relates to treatment methods and the like.
 悪性腫瘍等のがんや慢性難治性感染症などの疾患に対する新しい治療方法として、当該疾患に関連する抗原特異的な細胞傷害性T細胞(CTL:Cytotoxic T Lymphocyte)を用いた細胞免疫療法が知られている。近年では、がん治療に有効な手段として、患者からT細胞(未改変T細胞)を採取し、がん攻撃能力が高まるように遺伝子改変して遺伝子改変T細胞とした後、当該遺伝子改変T細胞を元の患者に戻してがんを治療するがん免疫療法が期待を集めている。 Cellular immunotherapy using antigen-specific cytotoxic T-lymphocytes (CTL) related to the disease is known as a new treatment method for diseases such as cancer such as malignant tumors and chronic intractable infectious diseases. It is In recent years, as an effective means for cancer therapy, T cells (unmodified T cells) are collected from patients, genetically modified to enhance cancer-attacking ability, and then genetically modified T cells are obtained. Expectations are high for cancer immunotherapy, which treats cancer by returning cells to the original patient.
 また、患者から採取したT細胞では、細胞疲弊が起こりやすかったり、増殖培養(又は拡大培養)が困難であるといった課題を有していることから、用いることができる細胞数に制限のないiPS細胞に由来するT細胞を利用した、がん免疫療法の開発も進められている。例えば、特許文献1には、CD4CD8T細胞を、インターロイキン7及びTCR(T細胞受容体)活性化剤を含む培地で培養し、CD8αβCTLを誘導する方法が記載されている。 In addition, T cells collected from patients have problems such as cell exhaustion and difficulty in proliferation culture (or expansion culture), so there is no limit to the number of iPS cells that can be used. Cancer immunotherapy is also being developed using T cells derived from For example, Patent Document 1 describes a method of inducing CD8α + β + CTL by culturing CD4 + CD8 + T cells in a medium containing interleukin 7 and a TCR (T cell receptor) activator. there is
 iPS細胞に由来するT細胞では、iPS細胞株や培養条件等によって品質が異なるため、得られたT細胞においては、その品質を評価して前記がん免疫療法に適したものを選択する必要がある。また、得られた遺伝子改変T細胞についても、その品質を評価して目的の遺伝子改変T細胞として適したものを選択する必要がある。このようなT細胞の品質評価項目としては、具体的に、例えば、T細胞の各段階で発現する分化マーカーの分析、TCR刺激後に産生されるサイトカインの分析、増殖能、細胞傷害活性、動物実験でのがん細胞抑制活性等が挙げられる。T細胞の品質は、これらの項目によって総合的に評価されるが、かかる評価は、煩雑であったり長期間を要する場合が多い。さらに、評価にかかるコストも高額なことから、より簡便かつ短期間でT細胞の品質を評価できる方法が求められている。 Since the quality of iPS cell-derived T cells varies depending on the iPS cell line, culture conditions, etc., it is necessary to evaluate the quality of the obtained T cells and select those suitable for the cancer immunotherapy. be. In addition, it is also necessary to evaluate the quality of the obtained genetically modified T cells and select those suitable as the desired genetically modified T cells. Specific examples of such T cell quality evaluation items include, for example, analysis of differentiation markers expressed at each stage of T cells, analysis of cytokines produced after TCR stimulation, proliferation ability, cytotoxic activity, animal experiments and cancer cell-suppressing activity. The quality of T cells is comprehensively evaluated by these items, but such evaluations are often complicated and require a long period of time. Furthermore, since the cost of evaluation is high, there is a demand for a method that can evaluate the quality of T cells more simply and in a short period of time.
 このようなT細胞の品質評価方法としては、例えば、PD-1やlag3等の発現量とT細胞の疲弊化との間に関係があることが見出されているため(非特許文献1)、これらのタンパク質をT細胞の疲弊化マーカーとして、その発現量によってT細胞の疲弊化を評価する方法が挙げられる。 As a method for evaluating the quality of such T cells, for example, it has been found that there is a relationship between the expression level of PD-1, lag3, etc. and the exhaustion of T cells (Non-Patent Document 1). , using these proteins as T cell exhaustion markers, a method of evaluating T cell exhaustion based on the expression level thereof.
国際公開第2018/135646号WO2018/135646
 in vitroにおいて、上記の疲弊化マーカーは、長期間培養によりかなりT細胞の疲弊化が進んでから発現する。そのため、iPS細胞から分化誘導した直後のT細胞等、比較的若いT細胞の品質を評価するためには不向きであることを本発明者らは見出した。従来、このように比較的若いT細胞の品質も評価できるマーカーは特定されていない。 In vitro, the above-mentioned exhaustion markers are expressed after the exhaustion of T cells has progressed considerably due to long-term culture. Therefore, the present inventors found that it is unsuitable for evaluating the quality of relatively young T cells such as T cells immediately after being induced to differentiate from iPS cells. So far, no markers have been identified that can also assess the quality of such relatively young T cells.
 
 本発明は、前記従来技術の有する課題に鑑みてなされたものであり、新たな指標によって比較的若いT細胞の品質も評価できるT細胞の評価方法、及びそれに用いるT細胞評価用試薬を提供することを目的とする。

The present invention has been made in view of the above-mentioned problems of the prior art, and provides a T cell evaluation method capable of evaluating the quality of relatively young T cells using a new index, and a T cell evaluation reagent used therein. for the purpose.
 本発明者らは前記課題を解決すべく鋭意検討を重ねた結果、増殖能が高いT細胞(高品質T細胞)及び増殖能が比較的低いT細胞(低品質T細胞)についてセクレトーム(secretome)解析及びトランスクリプトーム(transcriptome)解析を行なった。これらの解析で得られたデータについて、前記高品質T細胞及び前記低品質T細胞における主成分解析(PCA)を行なったところ、T細胞の品質(増殖能)と、T細胞のTCR刺激後の各経過日数における特定の遺伝子の発現量との間に、特定の関係があることを見出した。より具体的には、T細胞の増殖倍率の順位と第一主成分(PC1)スコアの順位との間には高い相関が認められ、高品質T細胞でPC1スコアが高く、低品質T細胞ではPC1スコアが低くなった。そのため、第一主成分の主成分負荷量の値が高い遺伝子は、PC1スコアが高い細胞、すなわち、高品質T細胞で発現し、第一主成分の主成分負荷量の値が低い遺伝子は、PC1スコアが低い細胞、すなわち、低品質T細胞で発現することを見出した。 The present inventors have made intensive studies to solve the above problems, and as a result, secretome (secretome) for T cells with high proliferative potential (high-quality T cells) and T cells with relatively low proliferative potential (low-quality T cells) Analysis and transcriptome analysis were performed. The data obtained from these analyzes were subjected to principal component analysis (PCA) of the high-quality T cells and the low-quality T cells. It was found that there is a specific relationship between the expression levels of specific genes at each elapsed day. More specifically, a high correlation was observed between the order of the proliferation fold of T cells and the order of the first principal component (PC1) score. Lowered PC1 score. Therefore, genes with a high principal component loading value of the first principal component are expressed in cells with a high PC1 score, that is, high-quality T cells, and genes with a low principal component loading value of the first principal component are expressed in It was found to be expressed in cells with low PC1 score, ie low quality T cells.
 これらの知見から、本発明者らは、第一主成分に対する主成分負荷量の絶対値が大きい遺伝子をT細胞の品質評価用のマーカーとし、これらの発現を指標とすることによって、T細胞の品質を評価できることを見出した。さらに、これら遺伝子は、T細胞のTCR刺激後の比較的初期に発現する遺伝子であることから、その発現を指標とすることによって、比較的若いT細胞の品質も評価できることを見出し、本発明を完成させるに至った。 Based on these findings, the present inventors used genes with a large absolute value of the principal component loading with respect to the first principal component as markers for quality evaluation of T cells, and used their expression as an index to determine the quality of T cells. We found that quality can be evaluated. Furthermore, since these genes are genes that are expressed relatively early after TCR stimulation of T cells, it was found that the quality of relatively young T cells can also be evaluated by using their expression as an index, and the present invention has been made. I came to complete it.
 すなわち、本発明の態様は以下のとおりである。 That is, the aspects of the present invention are as follows.
 [1] T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む、T細胞の評価方法。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[1] a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in T cells;
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
A method for evaluating T cells, comprising:
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 [2] 前記検出工程が、前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を用いて前記発現産物を検出する工程である、[1]に記載の方法。 [2] The detection step detects the expression product using a probe molecule that specifically binds to the expression product of at least one gene selected from the group consisting of the genes (I) to (III) The method according to [1], which is a step.
 [3] 前記発現産物が、前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドであり、かつ、前記プローブ分子が、前記ポリペプチドに特異的に結合する抗体及び/又はアプタマーである、[2]に記載の方法。 [3] The expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule specifically binds to the polypeptide The method of [2], which is an antibody and/or an aptamer.
 [4] 前記評価工程が、前記遺伝子の発現を指標としてT細胞の増殖能を評価する工程である、[1]~[3]のいずれか一項に記載の方法。 [4] The method according to any one of [1] to [3], wherein the evaluation step is a step of evaluating T cell proliferation ability using the expression of the gene as an index.
 [5] 前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、[1]~[4]のいずれか一項に記載の方法。
(I’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[5] At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The method according to any one of [1] to [4], which is a gene.
(I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 5, 7, 15, 27, and 44 (II') SEQ ID NOS: 5, 7, 15, 27, and 44 A gene (III') encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence according to any one of SEQ ID NO: A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence of any one of 5, 7, 15, 27 and 44.
 [6] 前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、[1]~[5]のいずれか一項に記載の方法。
(I’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[6] At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The method according to any one of [1] to [5], which is a gene.
(I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 15, and 27; (II') set forth in any one of SEQ ID NOs: 5, 15, and 27; Any of gene (III') SEQ ID NOS: 5, 15, and 27 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence or a gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in one.
 [7] [2]~[6]のいずれか一項に記載の方法に用いるための試薬であり、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を含有する、T細胞評価用試薬。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[7] A reagent for use in the method according to any one of [2] to [6], wherein at least one gene selected from the group consisting of the following genes (I) to (III) A T cell evaluation reagent containing a probe molecule that specifically binds to an expression product.
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 [8] 前記発現産物が、前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドであり、かつ、前記プローブ分子が、前記ポリペプチドに特異的に結合する抗体及び/又はアプタマーである、[6]に記載の試薬。 [8] The expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule specifically binds to the polypeptide The reagent of [6], which is an antibody and/or an aptamer.
 [9] 前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、[7]又は[8]に記載の試薬。
(I’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[9] At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The reagent of [7] or [8], which is a gene.
(I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 5, 7, 15, 27, and 44 (II') SEQ ID NOS: 5, 7, 15, 27, and 44 A gene (III') encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence according to any one of SEQ ID NO: A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence of any one of 5, 7, 15, 27 and 44.
 [10] 前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である[7]~[9]のいずれか一項に記載の試薬。
(I’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[10] At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') The reagent according to any one of [7] to [9], which is a gene.
(I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 15, and 27; (II') set forth in any one of SEQ ID NOs: 5, 15, and 27; Any of gene (III') SEQ ID NOS: 5, 15, and 27 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence or a gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in one.
[11][1]~[6]のいずれかに記載のT細胞の評価方法により得られた高品質T細胞。 [11] High-quality T cells obtained by the method for evaluating T cells according to any one of [1] to [6].
[12][11]に記載の高品質T細胞を含有する医薬組成物。 [12] A pharmaceutical composition containing the high-quality T cells of [11].
[13] がんの予防又は治療に使用するための[12]に記載の医薬組成物。 [13] The pharmaceutical composition of [12] for use in preventing or treating cancer.
[14][12]に記載の医薬組成物を用いる、がんの予防または治療方法。 [14] A method for preventing or treating cancer using the pharmaceutical composition of [12].
[15]がんを有するかまたはがんを有する危険性のある被験者に、[12]に記載の医薬組成物を投与することを含む、がんを治療または予防する方法。 [15] A method of treating or preventing cancer, comprising administering the pharmaceutical composition of [12] to a subject who has or is at risk of having cancer.
[16]がんを有するかまたはがんを有する危険性のある被験者に、T細胞を投与することを含む、がんを治療または予防する方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む、方法。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[16] A method for treating or preventing cancer, comprising administering T cells to a subject having cancer or at risk of having cancer, wherein A detection step of detecting the expression of at least one gene selected from the group consisting of the genes of ~ (III);
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
A method, including
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
[17]T細胞の製造方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む、T細胞の製造方法。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
 
[17] A method for producing T cells, comprising a detection step of detecting expression of at least one gene selected from the group consisting of the following genes (I) to (III) in the T cells;
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
A method of producing a T cell, comprising:
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 本発明によれば、新たな指標によってT細胞の品質を評価できるT細胞の評価方法、及びそれに用いるT細胞評価用試薬を提供することが可能となる。 According to the present invention, it is possible to provide a T cell evaluation method that can evaluate the quality of T cells using a new index, and a T cell evaluation reagent used for the method.
T細胞株23種類における、全37,912遺伝子のデータについて主成分分析(PCA)を行なって得られた散布図である。FIG. 3 is a scatter plot obtained by performing principal component analysis (PCA) on data for all 37,912 genes in 23 T cell lines. T細胞株23種類の増殖倍率の順位(増殖順位)とPC1スコアの順位(PC1順位)との関係を示す図である。FIG. 3 is a diagram showing the relationship between the rank of proliferation rate (proliferation rank) and the rank of PC1 score (PC1 rank) of 23 types of T cell lines.
 以下、本発明をその好適な実施形態に即して詳細に説明する。 The present invention will be described in detail below in accordance with its preferred embodiments.
 <T細胞の評価方法>
 本発明のT細胞の評価方法は、
 T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む方法(以下、場合により、単に「本発明の方法」という)である。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
<Method for evaluating T cells>
The method for evaluating T cells of the present invention comprises
a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in T cells;
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
(hereinafter sometimes simply referred to as "the method of the present invention").
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 [T細胞] [T cells]
  本発明におけるT細胞とは、表面にT細胞受容体(T  cell  receptor、TCR)と称される抗原受容体を発現している細胞を意味し、アルファベータ(αβ)T細胞およびガンマデルタ(γδ)T細胞を含む。 The T cell in the present invention means a cell expressing an antigen receptor called T cell receptor (TCR) on the surface, alpha beta (αβ) T cells and gamma delta (γδ ) containing T cells.
  本発明におけるT細胞は、例えば、ヘルパーT細胞、細胞傷害性T細胞、制御性T細胞、ナチュラルキラーT細胞、ナイーブT細胞、メモリーT細胞(例えば、ステムセルメモリーT細胞(TSCM)、セントラルメモリーT細胞(TCM))、エフェクターメモリーT細胞等)、抗原刺激されたメモリーT細胞、又はターミナルエフェクターT細胞、それらの組み合わせ、またはそれらの亜集団を含むが、これらに限定されることはない。 T cells in the present invention include, for example, helper T cells, cytotoxic T cells, regulatory T cells, natural killer T cells, naive T cells, memory T cells (e.g., stem cell memory T cells (TSCM), central memory T cells). cells (TCM), effector memory T cells, etc.), antigen-stimulated memory T cells, or terminal effector T cells, combinations thereof, or subpopulations thereof.
  ヘルパーT細胞はCD4陽性細胞であり、更に発現するサイトカインによりTh1細胞、Th2細胞及びTh17細胞等に分類される(例えば、J Allergy Clin. Immunol., 135(3): 626-635,2012)。 Helper T cells are CD4-positive cells, and are further classified into Th1 cells, Th2 cells, Th17 cells, etc. according to the cytokines they express (e.g., J Allergy Clin. Immunol., 135(3): 626-635, 2012).
  Th1細胞としては、例えば、IFN-γ、IL-2又はTNF-α等を発現する細胞等が挙げられる。Th2細胞としては、例えば、IL-4、IL-5、IL-6、IL-10又はIL-13等を発現する細胞等が挙げられる。Th17細胞としては、例えば、IL-17又はIL-6等を発現する細胞等が挙げられる。 Th1 cells include, for example, cells expressing IFN-γ, IL-2, TNF-α, and the like. Th2 cells include, for example, cells expressing IL-4, IL-5, IL-6, IL-10, IL-13, and the like. Th17 cells include, for example, cells expressing IL-17 or IL-6.
  細胞傷害性T細胞は、CD8陽性細胞であり、ヘルパーT細胞同様に、更に発現するサイトカインによりTc1細胞及びTc2細胞等に分類される。   Cytotoxic T cells are CD8-positive cells, and like helper T cells, are further classified into Tc1 cells, Tc2 cells, etc. according to the cytokines they express.
  Tc1細胞としては、例えば、IFN-γ、IL-2又はTNF-α等を発現する細胞等が挙げられる。Tc2細胞としては、例えば、IL-4、IL-5、IL-6、IL-10、IL-13等を発現する細胞等が挙げられる。 Examples of Tc1 cells include cells expressing IFN-γ, IL-2, TNF-α, and the like. Examples of Tc2 cells include cells that express IL-4, IL-5, IL-6, IL-10, IL-13, and the like.
  制御性T細胞としては、例えば、CD4(+)CD25(+)FoxP3(+)細胞が好ましく挙げられる。 Regulatory T cells preferably include, for example, CD4(+) CD25(+) FoxP3(+) cells.
  ナイーブT細胞としては、例えば、CD4(+)CD45RA(+)CD62L(+)CCR7(+)細胞、CD8(+)CD45RA(+)CD62L(+)CCR7(+)細胞、CD4(+)CCR7(+)CD45RA(+)CD95(-)CD45RO(-)細胞又はCD8(+)CCR7(+)CD45RA(+)CD95(-)CD45RO(-)細胞等が好ましく挙げられる。 Naive T cells include, for example, CD4 (+) CD45RA (+) CD62L (+) CCR7 (+) cells, CD8 (+) CD45RA (+) CD62L (+) CCR7 (+) cells, CD4 (+) CCR7 ( +)CD45RA(+)CD95(-)CD45RO(-) cells or CD8(+)CCR7(+)CD45RA(+)CD95(-)CD45RO(-) cells are preferred.
ステムセルメモリーT細胞としては、例えば、CD4(+)CD45RA(+)CD62L(+)CCR7(+)CD95(+)細胞、CD8(+)CD45RA(+)CD62L(+)CCR7(+)CD95(+)細胞、CD4(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+)細胞又はCD8(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+)細胞等が好ましく挙げられる。 Stem cell memory T cells include, for example, CD4 (+) CD45RA (+) CD62L (+) CCR7 (+) CD95 (+) cells, CD8 (+) CD45RA (+) CD62L (+) CCR7 (+) CD95 (+) ) cells, CD4(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+) cells or CD8(+)CCR7(+)CD45RA(+)CD95(+)CD45RO(+) cells are preferred. be done.
  セントラルメモリーT細胞としては、例えば、CD4(+)CD45RA(-)CD62L(+)CCR7(+)CD95(+)細胞、CD8(+)CD45RA(-)CD62L(+)CCR7(+)CD95(+)細胞、CD4(+)CCR7(+)CD45RA(-)CD45RO(+)細胞又はCD8(+)CCR7(+)CD45RA(-)CD45RO(+)細胞等が好ましく挙げられる。 Central memory T cells include, for example, CD4 (+) CD45RA (-) CD62L (+) CCR7 (+) CD95 (+) cells, CD8 (+) CD45RA (-) CD62L (+) CCR7 (+) CD95 (+) ) cells, CD4(+)CCR7(+)CD45RA(-)CD45RO(+) cells, CD8(+)CCR7(+)CD45RA(-)CD45RO(+) cells, and the like are preferable.
  エフェクターメモリーT細胞としては、例えば、CD4(+)CCR7(-)CD45RA(-)CD45RO(+)細胞又はCD8(+)CCR7(-)CD45RA(-)CD45RO(+)細胞等が好ましく挙げられる。 Examples of effector memory T cells include preferably CD4(+) CCR7(-) CD45RA(-) CD45RO(+) cells or CD8(+) CCR7(-) CD45RA(-) CD45RO(+) cells.
  ターミナルエフェクターT細胞としては、例えば、CD4(+)CD45RA(+)CD62L(-)細胞又はCD8(+)CD45RA(+)CD62L(-)細胞等が好ましく挙げられる。 Terminal effector T cells preferably include, for example, CD4(+)CD45RA(+)CD62L(-) cells or CD8(+)CD45RA(+)CD62L(-) cells.
 本発明において、T細胞の由来としては特に制限はなく、動物由来であっても、人由来であってもよく、健常者由来のT細胞であってもよいし、患者(例えば、免疫機能が低下している人又は動物や、悪性腫瘍、感染症又は自己免疫疾患を患っている人又は動物)由来のT細胞であってもよい。さらには、人工多能性幹細胞(iPS細胞)、胚性幹細胞(ES細胞)等の多能性幹細胞から分化誘導したT細胞;造血幹細胞等の体性幹細胞から分化誘導したT細胞;株化したT細胞であってもよい。 In the present invention, the origin of T cells is not particularly limited. depleted humans or animals, or those suffering from malignancies, infectious diseases or autoimmune diseases). Furthermore, T cells differentiated from pluripotent stem cells such as induced pluripotent stem cells (iPS cells) and embryonic stem cells (ES cells); T cells differentiated from somatic stem cells such as hematopoietic stem cells; It may be a T cell.
 人から採取されたT細胞の場合には、例えば、末梢血から分離された末梢血単核細胞(PBMC:Peripheral Blood Mononuclear Cells)より、従来公知の方法や市販のキット等を用いてT細胞を単離することができる。 In the case of T cells collected from humans, for example, T cells are isolated from peripheral blood mononuclear cells (PBMC: Peripheral Blood Monoclear Cells) isolated from peripheral blood using conventionally known methods or commercially available kits. It can be isolated.
 また、本発明において、「T細胞」には、人為的な遺伝子改変がなされていないT細胞(以下、場合により「未改変T細胞」という)の他、前記未改変T細胞に人為的な遺伝子改変がなされた「遺伝子改変T細胞」も含む。なお、本発明において、「人為的な遺伝子改変」には、T細胞への遺伝子導入又はT細胞の遺伝子編集によりT細胞の機能を改変することを含む。前記人為的な遺伝子改変としては、T細胞が有する遺伝子そのものの改変であっても、T細胞が有する遺伝子そのものの欠損であっても、外来の遺伝子が導入された改変であっても、これらの2種以上を組み合わせたものであってもよい。 In addition, in the present invention, "T cells" include T cells that have not been artificially genetically modified (hereinafter sometimes referred to as "unmodified T cells"), as well as artificial genes in the unmodified T cells. Also included are "genetically modified T cells" that have been modified. In the present invention, "artificial genetic modification" includes modification of T cell functions by gene transfer to T cells or gene editing of T cells. The artificial genetic modification may be modification of the gene itself possessed by T cells, deletion of the gene itself possessed by T cells, or modification resulting from the introduction of a foreign gene. A combination of two or more types may also be used.
 前記人為的な遺伝子改変の方法としては、従来公知の方法やそれに準じた方法が挙げられ、特に限定されない。T細胞への遺伝子導入としては、例えば、T細胞の機能を改変させる遺伝子そのもの(DNA、mRNA、miRNA、アンタゴmir、ODN(オリゴデオキシリボヌクレオチド)等)、又は当該遺伝子を挿入したベクター(レンチウイルスベクター、γ-又はα-レトロウイルスベクター、アデノウイルスベクター、アデノ随伴ウイルスベクター、ヘルペスウイルスベクター、ウマ脳炎ウイルスベクター等のウイルスベクター、トランスポゾンベクターであるpiggyBac(登録商標)の非ウイルスベクター)のT細胞への導入が挙げられ、T細胞の遺伝子編集としては、例えば、部位特異的ヌクレアーゼ(メガヌクレアーゼ、ジンクフィンガーヌクレアーゼ、TALEN、PPR(Pentatricopeptide repeat)、CRISPR-Cas等)を用いたT細胞の遺伝子の編集(ゲノム編集)が挙げられる。本発明において、前記人為的な遺伝子改変の方法としては、これらの1種単独であっても2種以上を組み合わせたものであってもよい。 The method of artificial genetic modification includes conventionally known methods and methods based thereon, and is not particularly limited. Gene introduction into T cells includes, for example, the gene itself (DNA, mRNA, miRNA, antagomir, ODN (oligodeoxyribonucleotide), etc.) that modifies the function of T cells, or a vector into which the gene is inserted (lentiviral vector , γ- or α-retroviral vectors, adenoviral vectors, adeno-associated viral vectors, herpes viral vectors, equine encephalitis viral vectors, non-viral vectors such as transposon vector piggyBac (registered trademark) to T cells Gene editing of T cells includes, for example, gene editing of T cells using site-specific nucleases (meganuclease, zinc finger nuclease, TALEN, PPR (Pentatricpeptide repeat), CRISPR-Cas, etc.) (genome editing). In the present invention, the artificial genetic modification method may be one of these alone or a combination of two or more.
 前記T細胞の機能を改変させる遺伝子としては、例えば、T細胞より分泌されるタンパク質;T細胞表面に発現するタンパク質及び少なくとも1つの細胞内シグナリングドメインを含む融合タンパク質;T細胞表面に発現するタンパク質、少なくとも1つの共刺激ドメイン、及び少なくとも1つの細胞内シグナリングドメインを含む融合タンパク質が挙げられ、より具体的には、細胞表面酵素、細胞接着因子、受容体(例えば、キメラ抗原受容体)、及びこれらを構成するサブユニットが挙げられる。 Examples of the gene that modifies the function of T cells include proteins secreted from T cells; fusion proteins containing proteins expressed on the surface of T cells and at least one intracellular signaling domain; proteins expressed on the surface of T cells; Fusion proteins comprising at least one co-stimulatory domain and at least one intracellular signaling domain include, more specifically, cell surface enzymes, cell adhesion factors, receptors (e.g., chimeric antigen receptors), and these and subunits that make up the
 [検出工程]
 (標的遺伝子)
 本発明の方法に係る検出工程においては、T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
[Detection process]
(target gene)
In the detection step according to the method of the present invention, the expression of at least one gene selected from the group consisting of genes (I) to (III) below is detected in T cells.
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 ここで、前記(I)~(III)の遺伝子からなる群において、(I)の遺伝子、(II)の遺伝子、及び(III)の遺伝子としては、それぞれ独立に、1種単独であっても2種以上であってもよい。 Here, in the group consisting of the genes (I) to (III), the gene (I), the gene (II), and the gene (III) are independently of each other. Two or more types may be used.
 前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドは、T細胞由来のセクレトーム(secretome)に含まれることが好ましい。本発明において、「セクレトーム」とは、細胞から分泌される又は細胞膜の中から細胞外に露出されるタンパク質を示し、これらタンパク質の全部又は一部が細胞に内在する態様も含む。 The polypeptide encoded by any one of the genes (I) to (III) is preferably contained in the T cell-derived secretome. In the present invention, the term "secretome" refers to proteins that are secreted from cells or that are exposed outside the cells through cell membranes, and include embodiments in which all or part of these proteins are endogenous to cells.
 本発明において、発現が検出される遺伝子(以下、場合により「標的遺伝子」という)は、ヒト由来のものであれば、典型的には、「(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子」である。下記の表1~5に、配列番号1~77に記載のアミノ酸配列について、各アミノ酸配列の配列番号及びGenBank Accession No.(GenBank No.);各アミノ酸配列からなるポリペプチドをコードする遺伝子の典型的なヌクレオチド配列の配列番号とそのオープンリーディングフレーム(ORF位置)、及びGenBank Accession No.(GenBank No.);各アミノ酸配列からなるポリペプチドが対応するタンパク質の略称(タンパク質略称)を示す。また、下記の表6~10に、表1~5に記載の各タンパク質略称について、正式名(フルネーム)及び存在する場合には別名を示す。 In the present invention, the gene whose expression is to be detected (hereinafter sometimes referred to as "target gene") is, if it is human-derived, typically "(I) any one of SEQ ID NOS: 1 to 77 A gene encoding a polypeptide comprising the amino acid sequence described in . Tables 1 to 5 below show the sequence numbers and GenBank Accession Nos. of each amino acid sequence for the amino acid sequences set forth in SEQ ID NOS: 1 to 77. (GenBank No.); SEQ ID NO of a typical nucleotide sequence of a gene encoding a polypeptide consisting of each amino acid sequence, its open reading frame (ORF position), and GenBank Accession No. (GenBank No.); indicates the abbreviation of the protein (protein abbreviation) corresponding to the polypeptide consisting of each amino acid sequence. Tables 6 to 10 below also show the official name (full name) and alternative names, if any, for each protein abbreviation listed in Tables 1 to 5.
 表1~5には、下記の実施例において各遺伝子が得られた由来、すなわち、次の(a)~(d):
(a)day0の試料においてトランスクリプトーム解析によりマップされた遺伝子(トランスクリプトームday0)
(b)day6の試料においてトランスクリプトーム解析によりマップされた遺伝子(トランスクリプトームday6)、
(c)day14の試料においてトランスクリプトーム解析によりマップされた遺伝子(トランスクリプトームday14)、
(d)セクレトーム解析により同定されたタンパク質をコードする遺伝子(セクレトーム)
を「由来データ」欄に合わせて示す。さらに、下記の実施例の主成分分析(PCA)により前記(a)~(d)で得られた各遺伝子の発現量の、第一主成分の主成分負荷量(principal component loading)も合わせて示す。
Tables 1 to 5 show the origins from which each gene was obtained in the examples below, namely the following (a) to (d):
(a) Genes mapped by transcriptome analysis in day0 samples (transcriptome day0)
(b) genes mapped by transcriptome analysis in day6 samples (transcriptome day6);
(c) genes mapped by transcriptome analysis in day14 samples (transcriptome day14);
(d) genes encoding proteins identified by secretome analysis (secretome)
are shown together with the "Origin data" column. Furthermore, the principal component loading of the first principal component of the expression level of each gene obtained in (a) to (d) above by the principal component analysis (PCA) of the following example is also included. show.
Figure JPOXMLDOC01-appb-T000001
 
Figure JPOXMLDOC01-appb-T000001
 
Figure JPOXMLDOC01-appb-T000002
 

 
Figure JPOXMLDOC01-appb-T000002
 

 
Figure JPOXMLDOC01-appb-T000003
 
Figure JPOXMLDOC01-appb-T000003
 
Figure JPOXMLDOC01-appb-T000004
 
Figure JPOXMLDOC01-appb-T000004
 
Figure JPOXMLDOC01-appb-T000005
 
Figure JPOXMLDOC01-appb-T000005
 
Figure JPOXMLDOC01-appb-T000006
 
Figure JPOXMLDOC01-appb-T000006
 
Figure JPOXMLDOC01-appb-T000007
 
Figure JPOXMLDOC01-appb-T000007
 
Figure JPOXMLDOC01-appb-T000008
 

 
Figure JPOXMLDOC01-appb-T000008
 

 
Figure JPOXMLDOC01-appb-T000009
 
Figure JPOXMLDOC01-appb-T000009
 
Figure JPOXMLDOC01-appb-T000010
 

 
Figure JPOXMLDOC01-appb-T000010
 

 
 本発明者らは、高品質T細胞及び低品質T細胞において、遺伝子の発現量についての主成分解析(PCA)によって得られるPC1スコアとT細胞の品質との間には高い相関があり、第一主成分の主成分負荷量の値が高い遺伝子は、PC1スコアが高い細胞、すなわち高品質T細胞で発現し、第一主成分の主成分負荷量の値が低い遺伝子は、PC1スコアが低い細胞、すなわち低品質T細胞で発現することを見出した。表1~2に記載の、配列番号1~25で示されるアミノ酸配列からなるポリペプチドをコードする遺伝子は、TCR刺激後のT細胞において発現する遺伝子のうち、前記第一主成分への寄与率が高く(具体的には、遺伝子の発現量の第一主成分に対する主成分負荷量の絶対値が0.9以上)、かつ、膜貫通型タンパク質をコードする遺伝子であり、表3~5に記載の、配列番号26~77で示されるアミノ酸配列からなるポリペプチドをコードする遺伝子は、第一主成分への寄与率が高く(具体的には、遺伝子の発現量の第一主成分に対する主成分負荷量の絶対値が0.9以上)、かつ、膜貫通型タンパク質以外のタンパク質をコードする遺伝子である。よって、これらの前記第一主成分への寄与率が高い(具体的には、遺伝子の発現量の第一主成分に対する主成分負荷量の絶対値が0.9以上)、配列番号1~77のいずれか1つで示されるアミノ酸配列を含むポリペプチドをコードする遺伝子の発現は、T細胞の品質評価の指標とすることができる。 The present inventors have found that in high-quality T cells and low-quality T cells, there is a high correlation between the PC1 score obtained by principal component analysis (PCA) for gene expression levels and the quality of T cells. Genes with high values of principal component loading of one principal component are expressed in cells with high PC1 score, i.e. high-quality T cells, and genes with low values of principal component loading of the first principal component have low PC1 scores. cells, namely low quality T cells. Genes encoding polypeptides consisting of amino acid sequences represented by SEQ ID NOS: 1 to 2, shown in Tables 1 and 2, among genes expressed in T cells after TCR stimulation, contribute to the first principal component is high (specifically, the absolute value of the principal component loading with respect to the first principal component of the expression level of the gene is 0.9 or more), and is a gene that encodes a transmembrane protein, and is shown in Tables 3 to 5 The described genes encoding polypeptides consisting of amino acid sequences represented by SEQ ID NOs: 26 to 77 have a high contribution rate to the first principal component (specifically, the main The absolute value of the component load is 0.9 or more) and encodes a protein other than a transmembrane protein. Therefore, these have a high contribution rate to the first principal component (specifically, the absolute value of the principal component loading with respect to the first principal component of the gene expression level is 0.9 or more), SEQ ID NOS: 1 to 77 Expression of a gene encoding a polypeptide containing an amino acid sequence represented by any one of can be used as an index for evaluating the quality of T cells.
 本発明においては、前記「(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子」の相同遺伝子(例えば、ヒト以外の生物におけるカウンターパート遺伝子)を前記標的遺伝子とすることもできる。また、遺伝子のヌクレオチド配列は、その変異などにより、自然界において(すなわち、非人工的に)変異し得ることから、本発明においては、このような天然の変異体も、前記標的遺伝子とすることができる。さらに、現在の技術水準においては、当業者であれば、例えば、前記(I)の遺伝子情報が得られた場合、そのヌクレオチド配列を改変し、そのコードするアミノ酸配列とは異なるが、機能改変したタンパク質を調製することもできるため、このような改変体も、前記標的遺伝子としてよい。 In the present invention, homologous genes (e.g., counterpart genes in organisms other than humans) of "(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77" It can also be the target gene. In addition, since the nucleotide sequence of a gene can be mutated in nature (that is, non-artificially) due to mutation thereof, etc., such natural mutants can also be used as the target gene in the present invention. can. Furthermore, in the current state of the art, for example, when the genetic information of (I) is obtained, a person skilled in the art can modify the nucleotide sequence and modify the function, although it is different from the amino acid sequence that codes for it. Since proteins can also be prepared, such variants may also be used as the target gene.
 したがって、本発明に係る標的遺伝子の態様には、「(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が、置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子」も含まれる。ここで、「アミノ酸残基が、置換、欠失、挿入、及び/又は付加されたアミノ酸配列」とは、アミノ酸残基が、置換、欠失、挿入、若しくは付加されたアミノ酸配列、又は、これらの2種以上の組み合わせがなされたアミノ酸配列であることを示す。また、「複数個」とは、例えば、20、19、18、17、16、15、14、13、12、11、10、9、8、7、6、5、4、3、又は2個の整数であることが好ましいが、これに制限されない。1若しくは複数個のアミノ酸残基とは、好ましくはアミノ酸残基が1個以上10個以下、より好ましくは1個以上5個以下、さらに好ましくは1個以上3個以下、特に好ましくは2個以下である。 Therefore, in the aspect of the target gene according to the present invention, "(II) one or more amino acid residues are substituted, deleted, or inserted in the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77. , and/or a gene encoding a polypeptide comprising an added amino acid sequence". Here, the term "amino acid sequence in which amino acid residues are substituted, deleted, inserted, and/or added" means an amino acid sequence in which amino acid residues are substituted, deleted, inserted, or added, or It shows that the amino acid sequence is a combination of two or more of Further, "plurality" means, for example, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2 is preferably an integer of, but not limited to. One or more amino acid residues is preferably 1 to 10 amino acid residues, more preferably 1 to 5, still more preferably 1 to 3, particularly preferably 2 or less. is.
 さらに、現在の技術水準においては、当業者であれば、例えば、前記(I)の遺伝子情報が得られた場合、そのヌクレオチド配列に基づいて、ハイブリダイゼーション技術(Southern,E.M.,J.Mol.Biol.,98:503-517,1975)や、ポリメラーゼ連鎖反応(PCR)技術(Saiki,R.K.,et al.Science,230:1350-1354,1985、Saiki,R.K.et al.Science,239:487-491,1988)等により、他の生物や細胞から相同遺伝子を取得することが可能である。 Furthermore, in the current state of the art, for example, when the genetic information of (I) above is obtained, a person skilled in the art can perform hybridization techniques (Southern, EM, J. Med. Mol. Biol., 98: 503-517, 1975) and polymerase chain reaction (PCR) technology (Saiki, RK, et al. Science, 230: 1350-1354, 1985, Saiki, RK et al. al.
 したがって、本発明に係る標的遺伝子の態様には、「(IV)配列番号1~77のいずれか1つに記載のアミノ酸配列をコードするヌクレオチド配列の相補鎖とストリンジェントな条件下でハイブリダイズするヌクレオチド配列」も含まれる。相同遺伝子を単離するためには、通常、ストリンジェントな条件下でハイブリダイゼーション反応を行う。「ストリンジェントな条件」とは、ハイブリダイゼーション後のメンブレンの洗浄操作を、高温下、低塩濃度溶液中で行うことを示し、かかる条件としては、例えば、30mMクエン酸三ナトリウム及び300mM塩化ナトリウム(2×SSC)を含有する、0.5%(w/v)SDS溶液中で60℃、20分間の洗浄条件が挙げられる。また、ハイブリダイゼーションは、例えば、公知であるECLダイレクトDNA/RNAラベリング・検出システム(GEヘルスケア バイオサイエンス株式会社製)に添付の使用説明書に記載の方法にしたがって実施することができる。ハイブリダイゼーションの条件が厳しくなるほど、高い相同性を有するDNAの単離を期待し得る。ただし、これらの条件は例示であり、DNAの濃度、DNAの長さ、ハイブリダイゼーションの反応時間等を適宜組み合わせることにより、必要な厳密性(ストリンジェンシー)を実現することが可能である。 Therefore, in embodiments of the target gene according to the present invention, "(IV) hybridizes under stringent conditions with the complementary strand of the nucleotide sequence encoding the amino acid sequence set forth in any one of SEQ ID NOS: 1 to 77 Also included are "nucleotide sequences". Hybridization reactions are typically performed under stringent conditions to isolate homologous genes. The term "stringent conditions" means that the washing operation of the membrane after hybridization is performed at a high temperature in a low-salt solution. 2×SSC) in 0.5% (w/v) SDS solution at 60° C. for 20 minutes. Also, hybridization can be carried out, for example, according to the method described in the instructions attached to the known ECL direct DNA/RNA labeling/detection system (manufactured by GE Healthcare Bioscience). The more stringent the hybridization conditions, the more highly homologous DNA can be expected to be isolated. However, these conditions are only examples, and the required stringency can be achieved by appropriately combining DNA concentration, DNA length, hybridization reaction time, and the like.
 さらに、例えばこのような方法にて取得された相同遺伝子がコードするタンパク質は、通常、配列番号1~77のいずれか1つに記載のアミノ酸配列と高い同一性を有する。したがって、本発明に係る標的遺伝子の態様には、「(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子」も含まれる。アミノ酸配列の同一性は、例えば、BLASTPのプログラム(Altschulら,J.Mol.Biol.,215,1990年、p.403-410)を用いて決定することができる。また、配列番号1~77のいずれか1つに記載のアミノ酸配列との同一性は、典型的には、70%以上であることが好ましく、より好ましくは、75%以上、80%以上、85%以上、90%以上、95%以上(例えば、96%以上、97%以上、98%以上、99%以上)である。 Furthermore, for example, proteins encoded by homologous genes obtained by such methods usually have a high degree of identity with the amino acid sequence set forth in any one of SEQ ID NOS: 1-77. Therefore, in the embodiment of the target gene according to the present invention, "(III) encodes a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77 Also includes "genes". Amino acid sequence identity can be determined, for example, using the BLASTP program (Altschul et al., J. Mol. Biol., 215, 1990, p. 403-410). In addition, the identity with the amino acid sequence set forth in any one of SEQ ID NOs: 1 to 77 is typically preferably 70% or more, more preferably 75% or more, 80% or more, 85% or more. % or more, 90% or more, 95% or more (eg, 96% or more, 97% or more, 98% or more, 99% or more).
 本発明においては、好ましくは、標的遺伝子は、表1~5に記載の遺伝子のうち、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である。これらの遺伝子から発現されるタンパク質は、T細胞の膜タンパク質として膜外に露出して発現されるため、これを検出後、当該膜タンパク質を利用して高品質(又は低品質)のT細胞の捕捉、分離、又は精製も可能となるためである。
(I’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子
ここで、前記(I’)~(III’)の遺伝子からなる群において、(I’)の遺伝子、(II’)の遺伝子、及び(III’)の遺伝子としては、それぞれ独立に、1種単独であっても2種以上であってもよい。
In the present invention, the target gene is preferably at least one gene selected from the group consisting of the following genes (I') to (III') among the genes listed in Tables 1 to 5. Since the proteins expressed from these genes are exposed outside the membrane as membrane proteins of T cells, after detection, the membrane proteins are used to produce high-quality (or low-quality) T cells. This is because capture, separation, or purification is also possible.
(I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27) (II') ) Substitution or deletion of one or more amino acid residues in the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27, and 44 (more preferably SEQ ID NOs: 5, 15, and 27) Gene (III') SEQ ID NOS: 5, 7, 15, 27, and 44 (more preferably SEQ ID NOS: 5, 15, and 27) encoding a polypeptide comprising deleted, inserted, and/or added amino acid sequences A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence described in any one of the above, wherein in the group consisting of the genes (I') to (III'), (I The gene of '), the gene of (II'), and the gene of (III') may be each independently of one type or two or more types.
 また、本発明に係る標的遺伝子としては、より若い(TCR刺激後の経過日数が少ない)T細胞の品質も評価できる観点から、上記の表1~5に示した遺伝子の中でも、「由来データ」欄が「トランスクリプトームday0」である遺伝子が好ましい。 In addition, as the target gene according to the present invention, from the viewpoint of being able to evaluate the quality of younger T cells (less days after TCR stimulation), among the genes shown in Tables 1 to 5 above, "derived data" Genes whose column is "transcriptome day 0" are preferred.
 (遺伝子の発現の検出)
 本発明において、「遺伝子の発現を検出する」とは、遺伝子の発現の有無の検出及び発現の程度の検出の双方を含む意であり、本発明においては、前記標的遺伝子の発現を検出する。遺伝子の発現量は、絶対量として又は相対量として把握することができる。相対量を把握する場合には、例えば、用意した標準試料の遺伝子の発現量と比較して判断することができる。「標準試料」とは、標的遺伝子を発現しているか否か、又は発現している場合にはその量が事前に特定されている試料を示す。例えば、品質が良い又は悪いことが事前に特定されているT細胞を前記標準試料とすることができる。
(Detection of gene expression)
In the present invention, "detecting gene expression" includes both detection of the presence or absence of gene expression and detection of the degree of expression. In the present invention, the expression of the target gene is detected. The gene expression level can be grasped as an absolute amount or as a relative amount. When grasping the relative amount, for example, it can be judged by comparing with the gene expression level of a prepared standard sample. A "standard sample" refers to a sample for which it has been specified in advance whether or not the target gene is expressed, and if so, the amount of expression. For example, T cells that have been previously identified as having good or bad quality can be used as the standard sample.
 本発明において、「遺伝子の発現」とは、遺伝子の転写及び翻訳の双方を含む意である。したがって、本発明における「遺伝子の発現の検出」には、転写レベル(mRNAレベル)での検出、及び翻訳レベル(タンパク質レベル)での検出(すなわち、前記遺伝子にコードされるポリペプチドの検出)の双方が含まれる。 In the present invention, "expression of a gene" means both transcription and translation of a gene. Therefore, "detection of gene expression" in the present invention includes detection at the transcription level (mRNA level) and detection at the translation level (protein level) (that is, detection of the polypeptide encoded by the gene). both are included.
 また、真核細胞では、遺伝子の転写過程で、mRNA前駆体中のイントロンを除去し、前後のエキソンを再結合する反応(スプライシング(splicing))が生じるが、エキソンの再結合に多様性が生じる場合があり、これにより様々な成熟mRNAが生産される。ひいては、それにより様々なタンパク質が翻訳される。このようなスプライシングの違いにより生じる多様なmRNAやタンパク質を「スプライシングバリアント(splicing variant)」という。したがって、本発明における遺伝子の発現の検出には、下記のプローブ分子に特異的に認識される限り、当該スプライシングバリアントの検出も含まれる。 In eukaryotic cells, in the process of gene transcription, a reaction (splicing) occurs in which introns in the pre-mRNA are removed and the exons before and after are recombined (splicing). In some cases, this produces different mature mRNAs. In turn, various proteins are translated thereby. Various mRNAs and proteins resulting from such splicing differences are called "splicing variants". Therefore, detection of gene expression in the present invention includes detection of the splicing variant as long as it is specifically recognized by the following probe molecule.
 本発明における遺伝子の発現の検出には、適宜公知の手法又はそれに準じた手法を用いることができる。中でも、本発明の方法に係る検出工程としては、前記(I)~(III)の遺伝子(好ましくは前記(I’)~(III’)の遺伝子)からなる群から選択される少なくとも1種の遺伝子(すなわち、前記標的遺伝子)の発現産物に特異的に結合するプローブ分子を用いて前記発現産物を検出することが好ましい。 For the detection of gene expression in the present invention, a known technique or a technique based thereon can be used as appropriate. Among them, as the detection step according to the method of the present invention, at least one selected from the group consisting of the genes (I) to (III) (preferably the genes (I') to (III')) Preferably, the expression product of a gene (ie, said target gene) is detected using a probe molecule that specifically binds to said expression product.
 〔転写レベルでの検出(標的ポリヌクレオチドの検出)〕
 転写レベルでの検出において、前記標的遺伝子の発現産物は、当該標的遺伝子から転写されたmRNA(以下、場合により「標的ポリヌクレオチド」という)である。前記標的ポリヌクレオチドには、前記mRNAを鋳型とするcDNAも含む。この場合の検出方法としては、例えば、前記プローブ分子として、前記標的ポリヌクレオチドの塩基配列中の適切な位置にハイブリダイズするように設計したオリゴヌクレオチドプローブを用い、ノーザンブロッティング、ドットブロット、RNaseプロテクションアッセイ、DNAマイクロアレイ解析法、in situハイブリダイゼーション法等によって、前記標的ポリヌクレオチドを検出する方法が挙げられる。
[Detection at transcription level (detection of target polynucleotide)]
In detection at the transcriptional level, the expression product of the target gene is mRNA transcribed from the target gene (hereinafter sometimes referred to as "target polynucleotide"). The target polynucleotide also includes cDNA using the mRNA as a template. Detection methods in this case include, for example, Northern blotting, dot blotting, and RNase protection assay using oligonucleotide probes designed to hybridize to appropriate positions in the base sequence of the target polynucleotide as the probe molecule. , DNA microarray analysis, in situ hybridization, and the like to detect the target polynucleotide.
 この場合には、例えば、前記オリゴヌクレオチドプローブとして、標識物質によって標識されたものを用い、当該標識物質に応じたシグナルを検出して、検出されたシグナル量を前記標的ポリヌクレオチドの量(すなわち、標的遺伝子の発現量)とすることができる。このときの標識物質としては、例えば、FITC(フルオレセインイソチオシアネート)、FAM(フルオレセインアミダイト)、DEAC(7-(ジエチルアミノ)クマリン)、R6G(ローダミン6G)、Texas Red、Cy5、BODIPY FL(商品名)等の蛍光物質;β-D-グルコシダ―ゼ、ルシフェラーゼ、HRP(ホースラディッシュペルオキシダーゼ)等の酵素;H、14C、32P、35S、123I等の放射性同位体;ビオチン、ストレプトアビジン等の親和性物質;ルミノール、ルシフェリン、ルシゲニン等の発光物質が挙げられる。 In this case, for example, as the oligonucleotide probe, one labeled with a labeling substance is used, a signal corresponding to the labeling substance is detected, and the detected signal amount is the amount of the target polynucleotide (i.e., expression level of the target gene). Examples of labeling substances at this time include FITC (fluorescein isothiocyanate), FAM (fluorescein amidite), DEAC (7-(diethylamino)coumarin), R6G (rhodamine 6G), Texas Red, Cy5, and BODIPY FL (trade name). enzymes such as β-D-glucosidase, luciferase, HRP (horseradish peroxidase); radioactive isotopes such as 3 H, 14 C, 32 P, 35 S, 123 I; biotin, streptavidin, etc. luminescence substances such as luminol, luciferin, and lucigenin.
 なお、本発明において、「シグナル」には、呈色(発色)、反射光、発光、消光、蛍光、放射性同位体による放射線等が含まれ、肉眼で確認できるものの他、シグナルの種類に応じた測定方法・装置によって確認できるものも含まれる。 In the present invention, the "signal" includes coloration (coloration), reflected light, luminescence, quenching, fluorescence, radiation from a radioactive isotope, and the like, and in addition to those that can be confirmed with the naked eye, depending on the type of signal It also includes those that can be confirmed by measuring methods and equipment.
 また、例えば、前記プローブ分子として、前記標的ポリヌクレオチド中の適切な位置を挟み込むように設計したオリゴヌクレオチドプライマーを用い、PCR法、RT-PCR法、TRC(Transcription Reverse transcription Concerted)法、NASBA(Nucleic Acid Sequence-Based Amplification)法、TMA(Transcription-Mediated Amplification)法等によって、前記標的ポリヌクレオチドを増幅して検出する方法も挙げられる。 Further, for example, as the probe molecule, oligonucleotide primers designed to sandwich appropriate positions in the target polynucleotide are used, PCR method, RT-PCR method, TRC (Transcription Reverse Transcription Concerned) method, NASBA (Nucleic Acid Sequence-Based Amplification) method, TMA (Transcription-Mediated Amplification) method, or the like may also be used to amplify and detect the target polynucleotide.
 この場合には、例えば、得られた増幅産物に蛍光色素(例えば、エチジウムブロマイド、SYBR Green(商品名)、SYTO63(商品名)等)をインターカレートして得られたシグナル(蛍光)を検出して、検出されたシグナル量(蛍光強度)を、前記標的ポリヌクレオチドの量(すなわち、標的遺伝子の発現量)とすることができる。また、前記オリゴヌクレオチドプローブと組み合わせて検出してもよい(ダブルダイプローブ法等)。さらに、前記標的ポリヌクレオチドを含む試料を直接シーケンサーに供して解析することにより、前記標的ポリヌクレオチドの量としてもよい。 In this case, for example, a signal (fluorescence) obtained by intercalating the obtained amplification product with a fluorescent dye (e.g., ethidium bromide, SYBR Green (trade name), SYTO63 (trade name), etc.) is detected. Then, the detected signal amount (fluorescence intensity) can be used as the amount of the target polynucleotide (that is, the expression level of the target gene). Moreover, detection may be performed in combination with the oligonucleotide probe (double dye probe method, etc.). Furthermore, the amount of the target polynucleotide may be determined by directly subjecting a sample containing the target polynucleotide to a sequencer for analysis.
 このようなオリゴヌクレオチドプローブ及びオリゴヌクレオチドプライマーは、当業者であれば、前記標的ポリヌクレオチドの配列に基づいて、公知の方法又はそれに準じた方法で設計することができる。また、前記オリゴヌクレオチドプローブ及びオリゴヌクレオチドプライマーの長さは、それぞれ独立に、少なくとも15塩基であることが好ましく、通常は、15~100塩基であり、好ましくは17~30塩基であり、より好ましくは20~25塩基である。前記オリゴヌクレオチドプローブ及びオリゴヌクレオチドプライマーは、例えば、市販のオリゴヌクレオチド合成機により合成することができる。また、天然のヌクレオチド(デオキシリボヌクレオチド及び/又はリボヌクレオチド)のみから構成されていなくともよく、例えば、PNA(polyamide nucleic acid)、LNA(登録商標、locked nucleic acid)、ENA(登録商標、2’-O,4’-C-Ethylene-bridged nucleic acids)等の非天然型のヌクレオチドにてその一部又は全部が構成されていてもよい。 Such oligonucleotide probes and oligonucleotide primers can be designed by a person skilled in the art based on the sequence of the target polynucleotide by a known method or a method based thereon. In addition, the length of the oligonucleotide probe and oligonucleotide primer are each independently preferably at least 15 bases, usually 15 to 100 bases, preferably 17 to 30 bases, more preferably 20-25 bases. The oligonucleotide probes and oligonucleotide primers can be synthesized by, for example, a commercially available oligonucleotide synthesizer. In addition, it may not consist only of natural nucleotides (deoxyribonucleotides and/or ribonucleotides), for example, PNA (polyamide nucleic acid), LNA (registered trademark, locked nucleic acid), ENA (registered trademark, 2'- O,4'-C-Ethylene-bridged nucleic acids) may be partially or wholly composed of non-natural nucleotides.
 〔翻訳レベルでの検出(標的ポリペプチドの検出)〕
 翻訳レベルでの検出において、前記標的遺伝子の発現産物は、当該標的遺伝子にコードされるポリペプチド(以下、場合により「標的ポリペプチド」という)である。より具体的には、前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドであり、すなわち、下記(i)~(iii)のポリペプチドからなる群から選択されるポリペプチドである。
(i)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチド
(ii)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチド
(iii)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチド。
[Detection at translation level (detection of target polypeptide)]
In translational level detection, the expression product of the target gene is a polypeptide encoded by the target gene (hereinafter sometimes referred to as "target polypeptide"). More specifically, it is a polypeptide encoded by any one of the genes (I) to (III), that is, selected from the group consisting of the following polypeptides (i) to (iii) is a polypeptide that is
(i) a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 1-77; (ii) one or more amino acid residues in the amino acid sequence set forth in any one of SEQ ID NOS: 1-77; is a substituted, deleted, inserted, and / or added amino acid sequence (iii) an amino acid sequence having 70% or more identity with the amino acid sequence set forth in any one of SEQ ID NOS: 1 to 77 A polypeptide comprising:
 前記(i)~(iii)のポリペプチドからなる群において、(i)のポリペプチド、(ii)のポリペプチド、及び(iii)のポリペプチドとしては、それぞれ独立に、1種単独であっても2種以上であってもよい。 In the group consisting of the polypeptides (i) to (iii), the polypeptide of (i), the polypeptide of (ii), and the polypeptide of (iii) are each independently one species alone, may be two or more.
 これらの中でも、前記(i)~(iii)のポリペプチドとしては、前記(I’)~(III’)の遺伝子のうちのいずれか1つにコードされるポリペプチド、すなわち、下記(i’)~(iii’)のポリペプチドからなる群から選択されるポリペプチドであることが好ましい。
(i’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列を含むポリペプチド
(ii’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチド
(iii’)配列番号5、7、15、27、及び44(より好ましくは配列番号5、15、及び27)のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチド。
Among these, the polypeptides (i) to (iii) include polypeptides encoded by any one of the genes (I') to (III'), that is, the following (i' ) to (iii') are preferably selected from the group consisting of polypeptides.
(i') a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27); (ii') SEQ ID NO: 5 , 7, 15, 27, and 44 (more preferably SEQ ID NOS: 5, 15, and 27), wherein one or more amino acid residues are substituted, deleted, inserted, and/or an amino acid according to any one of SEQ ID NOs: 5, 7, 15, 27 and 44 (more preferably SEQ ID NOs: 5, 15 and 27), comprising a polypeptide (iii') comprising an added amino acid sequence A polypeptide comprising an amino acid sequence having 70% or more identity with a sequence.
 前記(i’)~(iii’)のポリペプチドからなる群においても、(i’)のポリペプチド、(ii’)のポリペプチド、及び(iii’)のポリペプチドとしては、それぞれ独立に、1種単独であっても2種以上であってもよい。 In the group consisting of the polypeptides (i') to (iii'), the polypeptide (i'), the polypeptide (ii'), and the polypeptide (iii') each independently It may be one type alone or two or more types.
 また、本発明において、検出対象となるこれらの標的ポリペプチドとしては、上記のセクレトーム(T細胞由来セクレトーム)であることが好ましい。 In addition, in the present invention, these target polypeptides to be detected are preferably the above secretomes (T cell-derived secretomes).
 翻訳レベルでの検出の場合の検出方法の一例としては、抗ポリペプチド抗体及び/又はアプタマーによる検出や、標識アミノ酸による検出が挙げられる。 Examples of detection methods for detection at the translation level include detection using anti-polypeptide antibodies and/or aptamers, and detection using labeled amino acids.
 〈抗ポリペプチド抗体及び/又はアプタマーによる検出〉
 翻訳レベルでの検出の場合の検出方法としては、例えば、前記プローブ分子として、前記標的ポリペプチドに特異的に結合する抗体(以下、場合により「抗ポリペプチド抗体」という)及び/又はアプタマー(以下、場合により、単に「アプタマー」という)を用い、免疫細胞染色法、イメージングサイトメトリー、フローサイトメトリー、ELISA(Enzyme-Linked Immuno Sorbent Assay)法、ラジオイムノアッセイ、免疫沈降法、イムノブロッティング(ウェスタンブロット法等)、抗体アレイ、in vivo イメージング等の抗体を用いて検出する方法(免疫学的手法);前記抗体に代えてアプタマーを用いて検出する方法が挙げられる。また、前記免疫学的手法は、必要な試薬を調製した上で、AIA-900(東ソー社製)等のエンザイムイムノアッセイ装置やAIA-CL2400(東ソー社製)等の化学発光酵素免疫測定装置を用いて自動的に行なってもよい。
<Detection by anti-polypeptide antibody and/or aptamer>
Detection methods for detection at the translation level include, for example, antibodies that specifically bind to the target polypeptide (hereinafter sometimes referred to as "anti-polypeptide antibodies") and/or aptamers (hereinafter , In some cases, simply referred to as "aptamer"), immune cell staining method, imaging cytometry, flow cytometry, ELISA (Enzyme-Linked Immuno Sorbent Assay) method, radioimmunoassay, immunoprecipitation method, immunoblotting (Western blotting method etc.), methods of detection using antibodies such as antibody arrays and in vivo imaging (immunological techniques); methods of detection using aptamers instead of the antibodies. In addition, the immunological method uses an enzyme immunoassay device such as AIA-900 (manufactured by Tosoh Corporation) or a chemiluminescent enzyme immunoassay device such as AIA-CL2400 (manufactured by Tosoh Corporation) after preparing necessary reagents. can be done automatically.
 本発明において、「抗体」は、ポリクローナル抗体であっても、モノクローナル抗体であってもよく、また、抗体の機能的断片であってもよい。また、「抗体」には、免疫グロブリンの全てのクラス及びサブクラスが含まれる。抗体の「機能的断片」とは、抗体の一部分(部分断片)であって、本発明においては、前記標的遺伝子にコードされるポリペプチドを特異的に認識するものを示す。具体的には、Fab、Fab’、F(ab’)、可変領域断片(Fv)、ジスルフィド結合Fv、一本鎖Fv(scFv)、sc(Fv)、ダイアボディー(Diabody)、多特異性抗体、短鎖抗体、及びこれらの重合体等が挙げられる。 In the present invention, an "antibody" may be a polyclonal antibody, a monoclonal antibody, or a functional fragment of an antibody. "Antibody" also includes all classes and subclasses of immunoglobulins. A "functional fragment" of an antibody refers to a portion (partial fragment) of an antibody that specifically recognizes the polypeptide encoded by the target gene in the present invention. Specifically, Fab, Fab′, F(ab′) 2 , variable region fragment (Fv), disulfide-bonded Fv, single-chain Fv (scFv), sc(Fv) 2 , diabody, multispecific antibodies, short-chain antibodies, and polymers thereof.
 本発明に係る抗ポリペプチド抗体は、ポリクローナル抗体であれば、抗原(標的ポリペプチド、その部分ペプチド、又はこれらを発現する細胞等)で免疫動物を免疫し、その抗血清から、従来の手段(塩析、遠心分離、透析、カラムクロマトグラフィー等)によって、適宜精製して取得することができる。また、モノクローナル抗体は、ハイブリドーマ法や組換えDNA法によって作製することができる。ハイブリドーマ法としては、例えば、コーラー及びミルスタインの方法(Kohler&Milstein,Nature,256:495(1975))が挙げられ、組換えDNA法としては、例えば、前記抗ポリペプチド抗体をコードするDNAをハイブリドーマやB細胞等からクローニングし、適当なベクターに組み込んで、これを宿主細胞(哺乳類細胞株、大腸菌、酵母細胞、昆虫細胞、植物細胞等)に導入し、前記抗ポリペプチド抗体を組換え抗体として産生させる手法が挙げられる(例えば、P.J.Delves,Antibody Production:Essential Techniques,1997 WILEY;P.Shepherd and C.Dean Monoclonal Antibodies,2000 OXFORD UNIVERSITY PRESS;Vandamme A.M.et al.,Eur.J.Biochem.192:767-775(1990))。 If the anti-polypeptide antibody according to the present invention is a polyclonal antibody, an immunized animal is immunized with an antigen (target polypeptide, its partial peptide, or cells expressing these, etc.), and the antiserum is obtained by conventional means ( Salting out, centrifugation, dialysis, column chromatography, etc.) can be appropriately purified and obtained. Monoclonal antibodies can also be produced by the hybridoma method or recombinant DNA method. The hybridoma method includes, for example, Kohler and Milstein's method (Kohler & Milstein, Nature, 256:495 (1975)), and the recombinant DNA method includes, for example, the DNA encoding the anti-polypeptide antibody. It is cloned from B cells or the like, incorporated into an appropriate vector, introduced into host cells (mammalian cell lines, E. coli, yeast cells, insect cells, plant cells, etc.), and the anti-polypeptide antibody is produced as a recombinant antibody. (For example, P. J. Delves, Antibody Production: Essential Techniques, 1997 WILEY; P. Shepherd and C. Dean Monoclonal Antibodies, 2000 OXFORD UNIVERSITY PRESS; Vandamme AM et al., Eur.J. Biochem. 192:767-775 (1990)).
 この場合には、前記抗ポリペプチド抗体及びアプタマーとして、それぞれ、標識物質によって標識されたものを用い、当該標識物質に応じたシグナルを検出して、検出されたシグナル量を前記標的ポリペプチドの量(すなわち、標的遺伝子の発現量)とすることができる。このときの標識物質としては、前記抗ポリペプチド抗体又はアプタマーに結合することができ、化学的又は光学的方法で検出できるものであれば特に制限されることはなく、例えば、フィコエリスリン(PE)、フルオレセインイソチオシアネート(FITC)、ローダミンイソチオシアネート(RITC)等の蛍光物質や、ペルオキシダーゼ、β-D-ガラクトシダーゼ、マイクロペルオキシダーゼ、ホースラディッシュペルオキシダーゼ(HRP)、アルカリホスファターゼ等の酵素、放射性物質が挙げられる。 In this case, the anti-polypeptide antibody and the aptamer are each labeled with a labeling substance, the signal corresponding to the labeling substance is detected, and the amount of the detected signal is calculated as the amount of the target polypeptide. (that is, the expression level of the target gene). The labeling substance at this time is not particularly limited as long as it can bind to the anti-polypeptide antibody or aptamer and can be detected by a chemical or optical method. ), fluorescent substances such as fluorescein isothiocyanate (FITC) and rhodamine isothiocyanate (RITC), enzymes such as peroxidase, β-D-galactosidase, microperoxidase, horseradish peroxidase (HRP) and alkaline phosphatase, and radioactive substances. .
 また、前記抗ポリペプチド抗体及び/又はアプタマーを用いる場合には、前記標識物質を結合させた二次抗体を利用する方法や、二次抗体と前記標識物質とを結合させたポリマーを利用する方法などの間接的検出方法を利用することもできる。ここで、「二次抗体」とは、前記抗ポリペプチド抗体及び/又はアプタマーに特異的な結合性を示す抗体である。例えば、前記抗ポリペプチド抗体をウサギ抗体として調製した場合には、二次抗体として抗ウサギIgG抗体を使用することができる。ウサギ、ヤギ、マウスなどの様々な生物種に由来する抗体に対して、使用可能な標識二次抗体が市販されており、用いる抗ポリペプチド抗体の由来する生物種に応じて、適切な二次抗体を選択し、使用することができる。また、二次抗体に代えて、標識物質を結合させたプロテインG(例えば、Streptococcus属菌由来)、プロテインA(例えば、Staphylococcus aureus由来)、プロテインL(Peptostreptococcus magnus由来)等を用いることも可能である。 When the anti-polypeptide antibody and/or aptamer is used, a method of using a secondary antibody bound with the labeling substance, or a method of using a polymer binding the secondary antibody and the labeling substance. Indirect detection methods such as can also be used. Here, the "secondary antibody" is an antibody that exhibits specific binding to the anti-polypeptide antibody and/or aptamer. For example, when the anti-polypeptide antibody is prepared as a rabbit antibody, an anti-rabbit IgG antibody can be used as the secondary antibody. Labeled secondary antibodies that can be used for antibodies derived from various species such as rabbits, goats, and mice are commercially available. Antibodies can be selected and used. In place of the secondary antibody, protein G (for example, derived from Streptococcus spp.), protein A (for example, derived from Staphylococcus aureus), protein L (derived from Peptostreptococcus magnus), etc. to which a labeling substance is bound can also be used. be.
 〈標識アミノ酸による検出〉
 翻訳レベルでの検出の場合の検出方法としては、他にも例えば、標識アミノ酸を添加した培地でT細胞を培養し、前記標識アミノ酸で標識された標的ポリペプチドを検出する方法が挙げられる。このような方法としては、
 少なくとも1種のT細胞増殖用必須アミノ酸に代えてそのアミノ酸が標識された標識アミノ酸を含有し、かつ、共通γ鎖ファミリーサイトカインを含有するT細胞標識用培地でT細胞を培養する標識工程と、
 前記標識アミノ酸で標識されたT細胞由来ポリペプチドを検出する検出工程(以下、場合により「検出工程A」ともいう)と、
を含む方法が好ましい。
<Detection with labeled amino acids>
Other detection methods for detection at the translation level include, for example, a method of culturing T cells in a medium supplemented with a labeled amino acid and detecting a target polypeptide labeled with the labeled amino acid. As such a method,
a labeling step of culturing T cells in a T cell labeling medium containing a labeled amino acid in which the amino acid is labeled instead of at least one essential amino acid for T cell proliferation and containing a common γ chain family cytokine;
a detection step of detecting the T-cell-derived polypeptide labeled with the labeled amino acid (hereinafter sometimes referred to as "detection step A");
is preferred.
 ここで、「T細胞標識用培地」とは、T細胞を培養して標識するための標識アミノ酸を少なくとも含有する培地のことを示す。また、「T細胞増殖用必須アミノ酸(以下、場合により単に「必須アミノ酸」という)」とは、T細胞が増殖するために必須のアミノ酸を示し、より具体的には、バリン、イソロイシン、ロイシン、メチオニン、リジン、フェニルアラニン、トリプトファン、スレオニン、ヒスチジンである。 Here, "T cell labeling medium" refers to a medium containing at least labeling amino acids for culturing and labeling T cells. In addition, “essential amino acids for T cell proliferation (hereinafter, sometimes simply referred to as “essential amino acids”)” refers to amino acids essential for T cell proliferation, more specifically, valine, isoleucine, leucine, methionine, lysine, phenylalanine, tryptophan, threonine, histidine.
 前記T細胞標識用培地は、前記必須アミノ酸の少なくとも1種に代えてそのアミノ酸が標識された標識アミノ酸を含有する、すなわち、前記必須アミノ酸の少なくとも1種を含有せず、かつ、当該含有されない必須アミノ酸が標識されたアミノ酸である標識アミノ酸を含有する。「必須アミノ酸を含有しない」とは、前記T細胞標識用培地中に前記必須アミノ酸を実質的に含有しないことをいい、より具体的には、当該必須アミノ酸の含有量がT細胞標識用培地中に1×10-3mmol/L以下であることをいい、1×10-4mmol/L未満であることが好ましい。前記T細胞標識用培地から除かれる(すなわち、前記T細胞標識用培地に実質的に含有されない)必須アミノ酸としては、前記のいずれか1種であっても、2種以上の組み合わせであってもよいが、中でも、タンパク質合成経路への効率的な取り込みの観点から、メチオニンが好ましい。 The T cell labeling medium contains a labeled amino acid in which the amino acid is labeled instead of at least one of the essential amino acids, that is, does not contain at least one of the essential amino acids and the essential amino acid that is not contained Contains labeled amino acids, where the amino acids are labeled amino acids. "Containing no essential amino acids" means that the T cell labeling medium does not substantially contain the essential amino acids. More specifically, the content of the essential amino acids in the T cell labeling medium is is 1×10 −3 mmol/L or less, preferably less than 1×10 −4 mmol/L. The essential amino acids excluded from the T cell labeling medium (that is, not substantially contained in the T cell labeling medium) may be any one of the above or a combination of two or more of them. Among them, methionine is preferred from the viewpoint of efficient incorporation into the protein synthesis pathway.
 前記「標識アミノ酸」とは、前記必須アミノ酸の構造を維持したままでその一部が修飾され、その修飾に基づいて検出可能であるものを示す。前記「検出可能」には、呈色(発色)、消光、反射光、発光、蛍光等の目視で直接的に確認できるものの他、特定の測定方法や測定装置によって確認できることを含む。 The "labeled amino acid" refers to an essential amino acid partially modified while maintaining the structure of the essential amino acid, and detectable based on the modification. The above-mentioned "detectable" includes those that can be directly confirmed by visual observation such as coloring (color development), quenching, reflected light, luminescence, and fluorescence, as well as those that can be confirmed by a specific measuring method or measuring device.
 このような標識アミノ酸としては、例えば、アミノ酸の原子の一部をパルス安定同位体(例えば、H、13C、15N)に置換した同位体標識アミノ酸;アミノ酸の一部がアジド基、アルキン基等の修飾基によって修飾されたアミノ酸の構造的類似体(analog)が挙げられ、前記必須アミノ酸に対応して、これらのいずれか1種であっても、2種以上の組み合わせであってもよい。 Examples of such labeled amino acids include isotope-labeled amino acids in which some of the atoms of the amino acid are substituted with pulse stable isotopes (e.g., 2 H, 13 C, 15 N); Examples include structural analogs of amino acids modified by modifying groups such as groups, and corresponding to the essential amino acids, any one of them or a combination of two or more of them good.
 例えば、前記T細胞標識用培地が、前記必須アミノ酸のうちのメチオニンを含有しない場合、メチオニンに代えて前記T細胞標識用培地に含有される標識アミノ酸としては、例えば、メチオニンを構成する原子の一部を前記安定同位体に置換した同位体標識メチオニンであるH標識メチオニン、13C標識メチオニン、15N標識メチオニン;メチオニンの構造的類似体であるL-アジドホモアラニン(AHA:L-Azidohomoalanine)、L-ホモプロパルギルグリシン(HPG:L-Homopropargylglycine)、L-ホモアリルグリシン(HAG:L-Homoallylglycine)が挙げられ、これらのいずれか1種であっても、2種以上の組み合わせであってもよい。 For example, when the T cell labeling medium does not contain methionine among the essential amino acids, the labeling amino acid contained in the T cell labeling medium instead of methionine may be, for example, one of the atoms constituting methionine. 2 H-labeled methionine, 13 C-labeled methionine, and 15 N-labeled methionine, which are isotopically-labeled methionines in which the moiety is replaced with the stable isotope; L-azidohomoalanine (AHA), which is a structural analogue of methionine , L-homopropargylglycine (HPG: L-Homopropargylglycine), L-homoallylglycine (HAG: L-Homoallylglycine), any one of these, even a combination of two or more good.
 前記T細胞標識用培地において、前記標識アミノ酸の含有量(標識アミノ酸が2種以上である場合にはそれらの合計含有量、以下同じ)としては、T細胞の取り込み効率や培養効率、同T細胞への毒性等を考慮して適宜決定することができ、特に制限されないが、例えば、0.0001~0.1mmol/Lの範囲が挙げられ、0.001~0.05mmol/Lであることが好ましく、0.001~0.02mmol/Lであることがより好ましい。より具体的に、例えば、前記標識アミノ酸がAHAである場合には、0.0005~0.03mmol/Lであることが好ましく、0.001~0.03mmol/Lであることがより好ましく、0.002~0.03mmol/Lであることがさらに好ましく、0.005~0.02mmol/Lであることがさらにより好ましい。前記標識アミノ酸の含有量が前記上限を超えると、細胞が弱ったり死滅し易くなる傾向にあり、他方、前記下限未満であると、前記標識アミノ酸で標識されたポリペプチドを分泌又は細胞外に露出する細胞数が少なくなり、ポリペプチドの検出精度が低下する傾向にある。 In the T cell labeling medium, the content of the labeled amino acids (the total content of the labeled amino acids when there are two or more labeled amino acids; the same shall apply hereinafter) includes the T cell uptake efficiency, the culture efficiency, the T cell It can be determined as appropriate in consideration of the toxicity etc., and is not particularly limited. Preferably, it is 0.001 to 0.02 mmol/L. More specifically, for example, when the labeled amino acid is AHA, it is preferably 0.0005 to 0.03 mmol/L, more preferably 0.001 to 0.03 mmol/L. More preferably 0.002 to 0.03 mmol/L, even more preferably 0.005 to 0.02 mmol/L. When the content of the labeled amino acid exceeds the upper limit, the cells tend to weaken or die, whereas when the content is less than the lower limit, the polypeptide labeled with the labeled amino acid is secreted or exposed outside the cell. As a result, the number of cells to be detected tends to decrease, and the detection accuracy of the polypeptide tends to decrease.
 前記T細胞標識用培地は、共通γ鎖ファミリーサイトカインをさらに含有する。これを含有することにより、前記標識アミノ酸で標識されたポリペプチドを発現する細胞を高効率で得ることが可能となる。「共通γ鎖ファミリーサイトカイン」とは、共通のγ鎖(common γ chain:γサブユニット)を含む受容体を介して作用するサイトカインを示す。より具体的には、インターロイキン2(IL-2)、インターロイキン4(IL-4)、インターロイキン7(IL-7)、インターロイキン9(IL-9)、インターロイキン15(IL-15)、及びインターロイキン21(IL-21)が挙げられ、これらのいずれか1種であっても、2種以上の組み合わせであってもよい。中でも、細胞の生存及び増殖をより維持する観点からは、インターロイキン7、インターロイキン15、インターロイキン21、及びこれらの2種以上の組み合わせがより好ましい。 The T cell labeling medium further contains common γ-chain family cytokines. By containing this, it becomes possible to obtain cells expressing the polypeptide labeled with the labeled amino acid with high efficiency. A "common γ chain family cytokine" refers to a cytokine that acts via a receptor containing a common γ chain (γ subunit). More specifically, interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 7 (IL-7), interleukin 9 (IL-9), interleukin 15 (IL-15) , and interleukin 21 (IL-21), any one of which may be used in combination. Among them, interleukin 7, interleukin 15, interleukin 21, and combinations of two or more of these are more preferable from the viewpoint of better maintenance of cell survival and growth.
 前記T細胞標識用培地において、前記共通γ鎖ファミリーサイトカインの含有量(共通γ鎖ファミリーサイトカインが2種以上である場合にはそれらの合計含有量、以下同じ)としては、T細胞の性状等を考慮して適宜決定することができるため、これに限定されるものではないが、1~1000ng/mLであることが好ましく、5~100ng/mLであることがより好ましく、5~20ng/mLであることがさらに好ましい。前記共通γ鎖ファミリーサイトカインの含有量が前記上限を超えると、共通γ鎖ファミリーサイトカインを含有させることの効果が低下したり細胞増殖を阻害する傾向にあり、他方、前記下限未満であると、前記標識アミノ酸で標識されたポリペプチドを分泌又は細胞外に露出する細胞数が少なくなり、ポリペプチドの検出精度が低下する傾向にある。 In the T cell labeling medium, the content of the common γ-chain family cytokine (when there are two or more common γ-chain family cytokines, the total content thereof; the same shall apply hereinafter) is determined according to the properties of T cells, etc. Although it is not limited to this, it is preferably 1 to 1000 ng/mL, more preferably 5 to 100 ng/mL, and 5 to 20 ng/mL. It is even more preferable to have When the content of the common γ-chain family cytokine exceeds the upper limit, the effect of containing the common γ-chain family cytokine tends to decrease or cell proliferation tends to be inhibited. The number of cells that secrete or extracellularly expose a polypeptide labeled with a labeled amino acid tends to decrease, resulting in a decrease in the detection accuracy of the polypeptide.
 前記T細胞標識用培地としては、前記必須アミノ酸を除くため、血清を含有しないか、含有する場合には前記必須アミノ酸が除かれた透析血清であることが好ましい。また、前記T細胞標識用培地としては、他に、血清アルブミンやITS(インスリン-トランスフェリン-亜セレン酸ナトリウム)などの細胞保護添加剤;ECM(細胞外基質)成分等の足場剤等を含有していてもよい。これらのT細胞標識用培地又はその基礎培地としては、目的の必須アミノ酸を含有しない限り特に制限されず、DMEM(ダルベッコ改変イーグル培地)、RPMI(ロズウェルパーク記念研究所培地)、αMEM(α改変型イーグル最小必須培地)等の基礎培地;T細胞専用の無血清培地であるAIM VTM培地(Thermo Fisher Scientific社製);PRIME-XV T cell Expansion XSFM(富士フイルム和光純薬社製)等の公知や市販のT細胞用の培地を適宜採用することができる。 In order to remove the essential amino acids, the T cell labeling medium preferably does not contain serum or, if it does contain serum, is dialyzed serum from which the essential amino acids have been removed. In addition, the T cell labeling medium may also contain cytoprotective additives such as serum albumin and ITS (insulin-transferrin-sodium selenite); scaffolding agents such as ECM (extracellular matrix) components. may be These T cell labeling media or their basal media are not particularly limited as long as they do not contain the desired essential amino acids, and are DMEM (Dulbecco's Modified Eagle Medium), RPMI (Roswell Park Memorial Institute Medium), αMEM (α-modified basal medium such as Eagle minimum essential medium); AIM V TM medium (manufactured by Thermo Fisher Scientific) which is a serum-free medium exclusively for T cells; PRIME-XV T cell Expansion XSFM (manufactured by Fujifilm Wako Pure Chemical) or a commercially available medium for T cells can be used as appropriate.
 前記標識工程では、前記T細胞標識用培地でT細胞を培養する。これにより、前記標識アミノ酸をT細胞に取り込ませ、T細胞から前記標識アミノ酸で標識されたポリペプチド(T細胞由来ポリペプチド)を分泌又は細胞外に露出させる。前記標識工程における培養方法としては、特に制限されず、公知のT細胞の増殖培養条件を適宜採用することができ、また、T細胞の性状や濃度等を考慮して適宜調整することができるが、例えば、培養温度35~37.5℃、好ましくは36~37℃において、培養時間2~48時間、好ましくは8~24時間の条件が挙げられる。前記標識工程の培養装置としては、特に制限されず、例えば、プレート、ディッシュ、カラム、フラスコ、培養バッグ等が挙げられ、前記T細胞標識用培地等を供給するための供給手段や排出するための排出手段(センサ、バルブ、ポンプ、タンク等)をさらに備えていてもよい。 In the labeling step, T cells are cultured in the T cell labeling medium. As a result, the labeled amino acid is taken up by T cells, and a polypeptide labeled with the labeled amino acid (T cell-derived polypeptide) is secreted from the T cells or exposed outside the cells. The culture method in the labeling step is not particularly limited, and known T cell proliferation culture conditions can be appropriately adopted, and the characteristics and concentration of T cells can be taken into consideration and adjusted as appropriate. For example, the culture temperature is 35 to 37.5° C., preferably 36 to 37° C., and the culture time is 2 to 48 hours, preferably 8 to 24 hours. The culture apparatus for the labeling step is not particularly limited, and examples thereof include plates, dishes, columns, flasks, culture bags, and the like. Ejection means (sensors, valves, pumps, tanks, etc.) may also be provided.
 前記検出工程Aでは、前記標識アミノ酸で標識され、分泌又は細胞外に露出されたT細胞由来ポリペプチドを検出する。前記検出工程Aとしては、前記標識工程と同時(リアルタイム)で行なってもよい。前記検出工程Aにおける検出方法としては、前記標識アミノ酸の種類に応じた検出方法を適宜採用することができる。このような検出方法としては、特に制限されず、適宜公知の方法又はそれに準じた方法を適宜採用することができる。前記標識アミノ酸が同位体標識アミノ酸又はアミノ酸の構造的類似体である場合には、例えば、前記標識工程後又は標識工程中(すなわち培養途中)の培地について、ガス同位体比質量分析、同位体比赤外分光、液体クロマトグラフ質量分析(LC-MS/MS)等の質量分析を行うことによって、前記標識アミノ酸に応じたシグナルを検出し、検出されたシグナル量を前記標的ポリペプチドの量(すなわち、標的遺伝子の発現量)とすることができる。 In the detection step A, the T cell-derived polypeptide that is labeled with the labeling amino acid and is secreted or exposed outside the cell is detected. The detection step A may be performed simultaneously (in real time) with the labeling step. As a detection method in the detection step A, a detection method suitable for the type of the labeled amino acid can be appropriately employed. Such a detection method is not particularly limited, and a known method or a method based thereon can be appropriately employed. When the labeled amino acid is an isotope-labeled amino acid or a structural analogue of an amino acid, for example, the medium after the labeling step or during the labeling step (that is, during the culture) is subjected to gas isotope ratio mass spectrometry, By performing mass spectrometry such as infrared spectroscopy, liquid chromatography mass spectrometry (LC-MS/MS), a signal corresponding to the labeled amino acid is detected, and the amount of the detected signal is the amount of the target polypeptide (i.e. , expression level of the target gene).
 また、前記標識アミノ酸がアミノ酸の構造的類似体である場合には、前記修飾基に特異的な染色をすることによって検出することができる。例えば、前記修飾基がアジド基である場合(AHA等)には、当該アジド基をClick-iTTM Alexa Fluor 488 sDIBO Alkyne(Thermo Fisher Scientific社製)等の蛍光試薬によって染色することができるため、蛍光顕微鏡、フローサイトメトリー、イメージングサイトメトリー等を用いて、前記標識アミノ酸に応じたシグナル(発色、蛍光等)を検出し、検出されたシグナル量を前記標的ポリペプチドの量(すなわち、標的遺伝子の発現量)とすることができる。 Moreover, when the labeled amino acid is a structural analogue of an amino acid, it can be detected by staining the modified group specifically. For example, when the modifying group is an azide group (such as AHA), the azide group can be stained with a fluorescent reagent such as Click-iT Alexa Fluor 488 sDIBO Alkyne (manufactured by Thermo Fisher Scientific). Using fluorescence microscopy, flow cytometry, imaging cytometry, etc., a signal (color development, fluorescence, etc.) corresponding to the labeled amino acid is detected, and the amount of the detected signal is the amount of the target polypeptide (i.e., the amount of the target gene). expression level).
 また、翻訳レベルでの検出の場合の検出方法としては、他にも例えば、標的ポリペプチドの糖鎖を認識して検出する方法も挙げられる。このような方法としては、例えば、Click-iTTM(Thermo Fisher Scientific社製)等の市販の試薬を用いる方法が挙げられる。前記翻訳レベルでの検出の場合の検出方法は、適宜他の方法と組み合わせて用いてもよい。 Other detection methods for detection at the translation level include, for example, a method of recognizing and detecting the sugar chain of the target polypeptide. Such methods include, for example, methods using commercially available reagents such as Click-iT (manufactured by Thermo Fisher Scientific). The detection method for detection at the translation level may be used in combination with other methods as appropriate.
 本発明において、前記遺伝子の発現の検出としては、簡便性の観点から、翻訳レベルでの検出(前記標的ポリペプチドの検出)であることが好ましく、特に、前記抗ポリペプチド抗体を用いて前記標的ポリペプチドを検出する方法が好ましい。 In the present invention, detection of the expression of the gene is preferably detection at the translation level (detection of the target polypeptide) from the viewpoint of convenience. Methods that detect polypeptides are preferred.
 [評価工程]
 本発明の方法に係る評価工程においては、前記検出工程で検出された標的遺伝子の発現の有無又はその発現量を指標として、T細胞の品質を評価する。本発明において評価されるT細胞の品質としては、増殖能、がん細胞傷害活性が挙げられ、中でも、本発明の方法は、T細胞の増殖能を評価する方法として好ましい。「増殖能」とは、より具体的には、In vitro又はIn vivoにおいて、TCR刺激又は抗原刺激によってT細胞が活性化し、細胞傷害活性を有する細胞が増加する能力を示す。T細胞の増殖能は、例えば、下記の実施例に示す方法でT細胞のTCRをTCR刺激磁性ビーズで刺激し、刺激前の細胞数と刺激から14日後の細胞数との比で確認することができるが、これに制限されない。
[Evaluation process]
In the evaluation step according to the method of the present invention, the quality of T cells is evaluated using the presence or absence or the expression level of the target gene detected in the detection step as an index. The quality of T cells to be evaluated in the present invention includes proliferative ability and cancer cytotoxic activity. Among them, the method of the present invention is preferable as a method for evaluating the proliferative ability of T cells. More specifically, the term "proliferative ability" indicates the ability to activate T cells and increase cells having cytotoxic activity in vitro or in vivo by TCR stimulation or antigen stimulation. The proliferative ability of T cells can be confirmed, for example, by stimulating the TCR of T cells with TCR-stimulating magnetic beads by the method shown in the Examples below and comparing the cell count before stimulation to the cell count 14 days after stimulation. can, but is not limited to.
 本発明においては、前記検出工程で検出された、表1~5に示す標的遺伝子のうち、主成分負荷量の値が正の遺伝子は、当該遺伝子の発現量が多いほど、増殖能が高い、すなわち品質が良いと評価することができる。他方、主成分負荷量の値が負の遺伝子は、当該遺伝子の発現量が多いほど、増殖能が低い、すなわち品質が悪いと評価することができる。前記T細胞の品質の評価基準としては、目的に応じて適宜設定すればよく、特に限定されるものではない。例えば、前記検出工程で、前記標的遺伝子の発現レベル(標的遺伝子の転写レベルや翻訳レベル)に応じた標的遺伝子の発現が少しでも検出されれば、すなわち、前記標的ポリヌクレオチドや前記標的ポリペプチドが少しでも検出されれば、T細胞の増殖能が高い(又は低い)と評価してもよく、一定の閾値以上の標的遺伝子の発現量が検出されれば、T細胞の増殖能が高い(又は低い)と評価してもよい。また前記標的遺伝子の発現量に応じて、T細胞の増殖能の度合いを評価してもよい。例えば、増殖能が高い(又は低い)ことが事前に特定されているT細胞等の標準試料で検出される前記標的遺伝子の発現量に対して、一定の値以上、又は当該標的遺伝子の発現量の平均値の4SD以上、3SD以上、若しくは2SD以上のシグナル量であれば、増殖能が高い(又は低い)、すなわち品質が良い(又は悪い)と評価してもよい。 In the present invention, among the target genes shown in Tables 1 to 5 detected in the detection step, genes with a positive principal component loading value have a higher proliferation ability as the expression level of the gene increases. That is, it can be evaluated that the quality is good. On the other hand, a gene with a negative principal component loading value can be evaluated as having a lower proliferation ability, ie, a lower quality, as the expression level of the gene increases. The evaluation criteria for the quality of the T cells may be appropriately set according to the purpose, and are not particularly limited. For example, in the detection step, if even a little expression of the target gene is detected according to the expression level of the target gene (transcription level or translation level of the target gene), that is, the target polynucleotide or the target polypeptide If even a small amount is detected, the proliferative potential of T cells may be evaluated as high (or low), and if the expression level of the target gene above a certain threshold is detected, the proliferative potential of T cells is high (or low). Further, the degree of T cell proliferation ability may be evaluated according to the expression level of the target gene. For example, a certain value or more, or the expression level of the target gene relative to the expression level of the target gene detected in a standard sample such as T cells that have been specified in advance to have high (or low) proliferative ability If the signal amount is 4 SD or more, 3 SD or more, or 2 SD or more of the average value of , the proliferative ability is high (or low), that is, the quality may be evaluated as good (or bad).
 <T細胞評価用試薬、T細胞評価用キット>
 本発明のT細胞評価用試薬は、前記本発明のT細胞の評価方法に用いるための試薬(組成物)であり、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を含有する試薬(以下、場合により、単に「本発明の試薬」という)である。
(I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
<T cell evaluation reagent, T cell evaluation kit>
The T cell evaluation reagent of the present invention is a reagent (composition) for use in the T cell evaluation method of the present invention, and is at least one gene selected from the group consisting of the following genes (I) to (III). A reagent containing a probe molecule that specifically binds to an expression product of a gene of a species (hereinafter sometimes simply referred to as "the reagent of the present invention").
(I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
 本発明の試薬において、前記(I)~(III)の遺伝子、及び前記遺伝子の発現産物としては、その好ましい態様も含めて、上記の本発明の方法において述べたとおりである。本発明の試薬に係るプローブ分子としては、上記の本発明の方法において挙げた、オリゴヌクレオチドプローブ、オリゴヌクレオチドプライマー、抗ポリペプチド抗体、及びアプタマーが挙げられ、それぞれ、その好ましい態様も含めて、上記の本発明の方法において述べたとおりである。前記プローブ分子としては、これらのいずれか1種であっても2種以上の組み合わせであってもよい。中でも、前記抗ポリペプチド抗体及び/又は前記アプタマーが好ましく、前記抗ポリペプチド抗体がより好ましい。 In the reagent of the present invention, the genes (I) to (III) and the expression products of the genes are as described in the method of the present invention, including preferred embodiments thereof. Examples of probe molecules related to the reagent of the present invention include oligonucleotide probes, oligonucleotide primers, anti-polypeptide antibodies, and aptamers mentioned in the method of the present invention described above. in the method of the present invention. The probe molecule may be one of these or a combination of two or more of them. Among them, the anti-polypeptide antibody and/or the aptamer are preferable, and the anti-polypeptide antibody is more preferable.
 本発明の試薬は、液体であっても粉末状等の固体であってもよく、前記プローブ分子に加えて、緩衝液、生理食塩水、安定剤、保存剤、防腐剤、培地等の他の成分がさらに含有されていてもよい。 The reagent of the present invention may be liquid or solid such as powder. Additional ingredients may be included.
 本発明のT細胞評価用キットは、前記本発明のT細胞の評価方法に用いるためのキットであり、前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を備えるキット(以下、場合により、単に「本発明のキット」という)である。 The kit for evaluating T cells of the present invention is a kit for use in the method of evaluating T cells of the present invention, and contains at least one gene selected from the group consisting of genes (I) to (III). It is a kit comprising a probe molecule that specifically binds to an expression product (hereinafter sometimes simply referred to as "the kit of the present invention").
 本発明のキットに係るプローブ分子としては、前記本発明の試薬であることが好ましい。また、本発明のキットは、前記プローブ分子に加えて、緩衝液、生理食塩水等の希釈又は懸濁用液;DNA又はタンパク質を抽出及び/又は精製するための試薬;ブロッキング剤;キレート剤;前記標識物質;前記蛍光色素;前記検出工程に必要な試薬;pH調整剤等の試薬;標準試料;使用説明書;安全データシート(Safety Data Sheet)等をさらに備えていてもよい。 The probe molecule according to the kit of the present invention is preferably the reagent of the present invention. In addition to the probe molecule, the kit of the present invention also contains a buffer, a solution for dilution or suspension such as physiological saline; a reagent for extracting and/or purifying DNA or protein; a blocking agent; a chelating agent; The labeling substance; the fluorescent dye; reagents necessary for the detection step; reagents such as pH adjusters; standard samples; instructions for use;
 本発明は、前記のT細胞の評価方法により得られた高品質T細胞および該高品質T細胞を有効成分として含有する医薬組成物を提供する。 The present invention provides high-quality T cells obtained by the above T-cell evaluation method and pharmaceutical compositions containing the high-quality T cells as active ingredients.
 本発明の、T細胞を有効成分として含む医薬組成物は、がん治療対象を処置するために使用することができる。本発明の医薬組成物は、製剤技術分野において慣用の方法、例えば、日本薬局方に記載の方法等により製造することができる。本発明の医薬組成物は、薬学的に許容される添加剤を含んでいてもよい。該添加剤としては、例えば、細胞培養液、生理食塩水や適当な緩衝液(例えば、リン酸系緩衝液)等が挙げられる。 The pharmaceutical composition containing T cells as an active ingredient of the present invention can be used to treat cancer patients. The pharmaceutical composition of the present invention can be produced by a method commonly used in the field of formulation technology, such as the method described in the Japanese Pharmacopoeia. The pharmaceutical composition of the present invention may contain pharmaceutically acceptable additives. Examples of such additives include cell culture media, physiological saline, and suitable buffers (eg, phosphate buffers).
 本発明の医薬組成物は、T細胞を生理食塩水や適当な緩衝液(例えばリン酸系緩衝液)等に懸濁することによって製造することができる。所望の治療効果が発揮されるように、一回投与分の量として、例えば1×10個以上の細胞を含有させることが好ましい。より好ましくは1×108個以上、さらに好ましくは1×109個以上である。細胞の含有量は、適用対象の性別、年齢、体重、患部の状態、細胞の状態等を考慮して適宜調整することができる。本発明の医薬組成物には、T細胞の他、細胞を保護する目的でジメチルスルフォキシド(DMSO)および血清アルブミン等を含有させてもよい。また、細菌の混入を阻止する目的で抗生物質等ならびに細胞の活性化および分化を促す目的でビタミン類やサイトカイン等を含有させてもよい。さらに、製剤上許容される他の成分(例えば、担体、賦形剤、崩壊剤、緩衝剤、乳化剤、懸濁剤、無痛化剤、安定剤、保存剤、防腐剤、生理食塩水等)を本発明の医薬組成物に含有させてもよい。 The pharmaceutical composition of the present invention can be produced by suspending T cells in physiological saline, an appropriate buffer (eg, phosphate buffer), or the like. It is preferable to contain, for example, 1×10 7 cells or more in a single dose so as to achieve the desired therapeutic effect. It is more preferably 1×10 8 or more, still more preferably 1×10 9 or more. The content of cells can be appropriately adjusted in consideration of the sex, age, weight, condition of the affected area, condition of cells, etc. of the subject. In addition to T cells, the pharmaceutical composition of the present invention may contain dimethylsulfoxide (DMSO), serum albumin, and the like for the purpose of protecting cells. Antibiotics and the like may be added for the purpose of preventing bacterial contamination, and vitamins, cytokines, and the like may be included for the purpose of promoting activation and differentiation of cells. Furthermore, other pharmaceutically acceptable components (e.g., carriers, excipients, disintegrants, buffers, emulsifiers, suspending agents, soothing agents, stabilizers, preservatives, preservatives, physiological saline, etc.) It may be included in the pharmaceutical composition of the present invention.
 本発明のT細胞を有効成分として含む医薬組成物は、低温凍結保存することができる。低温凍結保存する場合、温度は、細胞の保存に適した温度であれば特に限定されない。例えば、-20℃、-80℃および-120~-196℃が挙げられる。低温凍結保存する場合、細胞は、バイアル等の適切な容器中で保存することができる。T細胞の凍結時および解凍時の細胞損傷リスクを最小限にするための操作は、当業者には周知である。 A pharmaceutical composition containing the T cells of the present invention as an active ingredient can be cryopreserved. In the case of low-temperature cryopreservation, the temperature is not particularly limited as long as it is suitable for cell preservation. Examples include -20°C, -80°C and -120 to -196°C. For cryopreservation, cells can be stored in suitable containers such as vials. Manipulations to minimize the risk of cell damage during freezing and thawing of T cells are well known to those of skill in the art.
 T細胞の冷凍保存においては、T細胞を培養液から回収し、緩衝液または培養液により洗浄し、細胞数を計数し、遠心分離等により濃縮して、凍結媒質(例えば、10% DMSOを含む培養液)中に懸濁した後、低温凍結保存する。T細胞は、複数の培養容器で培養した細胞を合わせ、単一ロットにすることができる。本発明のT細胞を含む医薬組成物は、バイアル等の容器当たり、例えば、5×104個~9×1010個のT細胞を含むが、対象とするがんの種類、投与対象、投与経路等に応じて変更することができる。 For cryopreservation of T cells, T cells are collected from the culture medium, washed with buffer or culture medium, counted, concentrated by centrifugation, etc., and placed in a freezing medium (e.g., containing 10% DMSO). culture solution), and cryopreserved. T cells can be made into a single lot by combining cells cultured in multiple culture vessels. The pharmaceutical composition containing T cells of the present invention contains, for example, 5×10 4 to 9×10 10 T cells per container such as a vial. It can be changed according to the route or the like.
 本発明のT細胞を含む医薬組成物の投与経路としては、例えば、輸注、腫瘍内注射、動注、門脈注、腹腔内投与等が挙げられる。但し、本発明の医薬組成物中の有効成分であるT細胞が患部に送達される限り、投与経路はこれに限られるものではない。投与スケジュールとしては、単回投与または複数回投与とすることができる。複数回投与の期間は例えば、2~4週間に1回投与を繰り返す方法または半年から1年に1回投与を繰り返す方法等を採用することができる。投与スケジュールの作成においては、対象患者の性別、年齢、体重、病態等を考慮することができる。 Examples of administration routes of the pharmaceutical composition containing the T cells of the present invention include infusion, intratumoral injection, arterial injection, portal vein injection, intraperitoneal administration, and the like. However, the administration route is not limited to this as long as the T cells, which are the active ingredients in the pharmaceutical composition of the present invention, are delivered to the affected area. Dosage schedules can be single doses or multiple doses. For the period of multiple administrations, for example, a method of repeating administration once every 2 to 4 weeks, a method of repeating administration once every six months to a year, or the like can be adopted. In preparing the administration schedule, the sex, age, body weight, disease state, etc. of the target patient can be taken into consideration.
 本発明のT細胞を含む医薬組成物を、がんの予防または治療に使用する場合において、他家移植である場合、拒絶反応を起こしにくいという観点から、T細胞を採取されるがん患者または非がん患者は、がん治療のために、本発明の医薬組成物を投与されるがん患者と、HLAの型が一致していることが好ましい。また、本発明の医薬組成物が投与されるがん患者と、T細胞を採取されるがん患者とは同一人であることがより好ましい。すなわち、他家移植より自家的な移植によるがん治療がより好ましい。 In the case of using the pharmaceutical composition containing the T cells of the present invention for the prevention or treatment of cancer, in the case of allotransplantation, from the viewpoint that rejection is unlikely to occur, cancer patients from whom T cells are collected or The non-cancer patient preferably has the same HLA type as the cancer patient to whom the pharmaceutical composition of the present invention is administered for cancer treatment. Moreover, it is more preferable that the cancer patient to whom the pharmaceutical composition of the present invention is administered and the cancer patient from whom T cells are collected are the same person. That is, cancer treatment by autologous transplantation is more preferable than allogeneic transplantation.
 本発明の医薬組成物は、がんの予防または治療のために用いられる。がんとしては、卵巣がん、肝芽腫、肝細胞がん、胃がん、食道がん、膵臓がん、腎細胞がん、乳がん、悪性黒色腫、非小細胞肺がん、子宮頸がん、膠芽腫、前立腺がん、神経芽腫瘍、慢性リンパ性白血病、甲状腺乳頭がん、大腸がん、およびB細胞非ホジキンリンパ腫が挙げられるが、これらに限定されない。 The pharmaceutical composition of the present invention is used for prevention or treatment of cancer. Cancers include ovarian cancer, hepatoblastoma, hepatocellular carcinoma, gastric cancer, esophageal cancer, pancreatic cancer, renal cell carcinoma, breast cancer, malignant melanoma, non-small cell lung cancer, cervical cancer, and glioblastoma. including, but not limited to, blastoma, prostate cancer, neuroblastoma, chronic lymphocytic leukemia, papillary thyroid cancer, colorectal cancer, and B-cell non-Hodgkin's lymphoma.
 がんの予防または治療のために、本発明のT細胞を含む医薬組成物とがんワクチンとを組み合わせて使用する場合、本発明の医薬組成物およびがんワクチンの投与時期は限定されず、本発明の医薬組成物およびがんワクチンの投与を、投与対象に対し、同時に投与してもよく、また時間差をおいて投与してもよい。本発明の医薬組成物とがんワクチンとは別々に製剤化されていてもよく、また両者が混合された合剤であってもよい。本発明の医薬組成物およびがんワクチンの投与量は、臨床上用いられている投与量に準ずればよく、疾患、投与対象、投与経路、薬物との組み合わせ等により適宜選択することができる。 When the pharmaceutical composition containing T cells of the present invention and a cancer vaccine are used in combination for the prevention or treatment of cancer, the timing of administration of the pharmaceutical composition and cancer vaccine of the present invention is not limited, The pharmaceutical composition of the present invention and cancer vaccine may be administered to the subject at the same time or at different times. The pharmaceutical composition of the present invention and cancer vaccine may be formulated separately, or may be a combination drug in which both are mixed. The dosage of the pharmaceutical composition and cancer vaccine of the present invention may conform to the dosage used clinically, and can be appropriately selected depending on the disease, administration subject, administration route, combination with drugs, and the like.
 本発明は、がんを有するかまたはがんを有する危険性のある被験者に、T細胞を投与することを含む、がんを治療または予防する方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む、方法
((I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。)を提供する。
The present invention provides a method for treating or preventing cancer, comprising administering T cells to a subject who has cancer or is at risk of having cancer, wherein the T cells have the following (I A detection step of detecting expression of at least one gene selected from the group consisting of genes from ) to (III);
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
A method ((I) a gene encoding a polypeptide comprising an amino acid sequence according to any one of SEQ ID NOS: 1-77, comprising , gene (III) encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted and/or added amino acid according to any one of SEQ ID NOs: 1 to 77 A gene encoding a polypeptide comprising an amino acid sequence having greater than 70% identity with the sequence.).
 本発明における被験者は、脊椎動物である。特定の実施形態では、該脊椎動物は、哺乳動物である。該哺乳動物には、家畜(例えば、ウシ)、スポーツ動物、ペット/コンパニオン動物(例えば、ネコ、イヌ、及びウマ)、霊長類、マウス、及びラットが含まれるが、これらに限定されない。特定の実施形態では、哺乳動物は、ヒトである。 A subject in the present invention is a vertebrate. In certain embodiments, the vertebrate is a mammal. Such mammals include, but are not limited to, farm animals (eg, cows), sport animals, pet/companion animals (eg, cats, dogs, and horses), primates, mice, and rats. In certain embodiments, the mammal is human.
 本発明は、T細胞の製造方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
 前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
を含む、T細胞の製造方法
((I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
(II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
(III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。)を提供する。
The present invention provides a method for producing T cells, comprising a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in the T cells;
an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
A method for producing T cells ((I) a gene encoding a polypeptide comprising an amino acid sequence according to any one of SEQ ID NOS: 1-77 (II) according to any one of SEQ ID NOS: 1-77 Any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence of A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence described in .).
 以下、実施例に基づいて本発明をより具体的に説明するが、本発明は以下の実施例に限定されるものではない。 The present invention will be described in more detail below based on examples, but the present invention is not limited to the following examples.
 1.T細胞の品質(増殖能)評価
 (1)下記の表11に示すT細胞株24種類を解凍し、96wellプレートの培地A(15%(w/v)ウシ胎児血清(FBS、BioWest社製)、5ng/mLインターロイキン7(IL-7)、及び5ng/mLインターロイキン15(IL-15)を添加したαMEM(α改変型イーグル最小必須培地))に、それぞれ1×10cells/wellで播種した。
1. Evaluation of quality (proliferation ability) of T cells (1) Twenty-four types of T cell lines shown in Table 11 below were thawed and placed in medium A (15% (w/v) fetal bovine serum (FBS, BioWest) in a 96-well plate. , 5 ng/mL interleukin 7 (IL-7), and 5 ng/mL interleukin 15 (IL-15) added to αMEM (α modified Eagle minimal essential medium)) at 1×10 6 cells/well, respectively. sown.
 (2)T細胞受容体(TCR)刺激磁性ビーズ(Dynabeads Human T-Activator CD3/CD28 for T Cell Expansion and Activation(Thermo Fisher Scientific社製))を用いて、播種したT細胞のTCRをそれぞれ刺激後、培地Aで適宜培地交換しながら、5%CO、37℃の条件下で14日間培養した。 (2) T cell receptor (TCR) stimulation Magnetic beads (Dynabeads Human T-Activator CD3/CD28 for T Cell Expansion and Activation (manufactured by Thermo Fisher Scientific)) are used to stimulate the TCR of the seeded T cells. , and cultured for 14 days under conditions of 5% CO 2 and 37° C. while replacing the medium with medium A as appropriate.
 (3)培養14日目の刺激後の各T細胞を血球算定盤を用いて計数し、次式:
  増殖倍率=TCR刺激後の14日間培養後のT細胞数/TCR刺激前のT細胞数
により、各T細胞における増殖倍率(Fold proliferation)を算出した。結果を下記の表11に合わせて示す。増殖倍率の値より、同増殖倍率が20以上となったT細胞(表11の細胞No.1~12のT細胞)を「高品質T細胞」に、同増殖倍率が20未満となったT細胞(表11の細胞No.13~24のT細胞)を「低品質T細胞」に、それぞれ分類した。
(3) Each T cell after stimulation on day 14 of culture was counted using a hemocytometer, and the following formula:
The fold proliferation of each T cell was calculated by multiplying fold=number of T cells after 14 days culture after TCR stimulation/number of T cells before TCR stimulation. The results are shown together in Table 11 below. From the value of the proliferation rate, T cells with the same proliferation rate of 20 or more (T cells of cell Nos. 1 to 12 in Table 11) are classified as "high-quality T cells", and T cells with the same proliferation rate of less than 20 are classified as "high-quality T cells". The cells (T cells of Cell Nos. 13-24 in Table 11) were classified as "low quality T cells" respectively.
Figure JPOXMLDOC01-appb-T000011
 
 
Figure JPOXMLDOC01-appb-T000011
 
 
 2.T細胞のトランスクリプトーム解析
 (1)上記1の(3)で高品質T細胞(12種類)及び低品質T細胞(12種類)に分類したT細胞と同じ株のT細胞(TCR未刺激)について、それぞれ、上記1の(1)~(2)と同様にしてTCR刺激磁性ビーズを用いてTCRを刺激し、培地Aで適宜培地交換しながら、5%CO、37℃の条件下で培養した。
2. Transcriptome analysis of T cells (1) T cells of the same strain as the T cells classified into high-quality T cells (12 types) and low-quality T cells (12 types) in 1 (3) above (TCR unstimulated) For each, stimulate the TCR using TCR-stimulating magnetic beads in the same manner as in 1 (1) to (2) above, and replace the medium with medium A as appropriate under the conditions of 5% CO 2 and 37 ° C. cultured.
 (2)TCR刺激後0日目(day0)、6日目(day6)及び14日目(day14)の細胞をそれぞれ回収し、先ず、RNeasy Micro Kit(QIAGEN社製)を用いて、Total RNAを抽出した。次いで、抽出したTotal RNA 10ngから、SMART-Seq v4 Ultra Low Input RNA Kit for Sequencing(Clontech社製)を用いて、cDNAを合成・増幅した。 (2) Cells were collected on day 0 (day0), day 6 (day6), and day 14 (day14) after TCR stimulation, and total RNA was first extracted using RNeasy Micro Kit (QIAGEN). Extracted. Next, cDNA was synthesized and amplified from 10 ng of the extracted total RNA using SMART-Seq v4 Ultra Low Input RNA Kit for Sequencing (manufactured by Clontech).
 (3)(2)で得られたcDNA 1ng分を使用して、Nextera XT DNA Library Preparation Kit(Illumina社製)及びNextera XT v2 Index Kit Set A(Illumina社製)を用いてライブラリー調製を行ない、Next-seq500(illumina社製)を用いて、リード長:75bp、single end readの条件でシークエンス解析を行うことにより、1試料あたり500万リード以上の遺伝子を解読した。 (3) Using 1 ng of cDNA obtained in (2), library preparation was performed using Nextera XT DNA Library Preparation Kit (manufactured by Illumina) and Nextera XT v2 Index Kit Set A (manufactured by Illumina). , Next-seq500 (manufactured by Illumina), read length: 75 bp, sequence analysis was performed under the conditions of single end read to decode more than 5 million reads of genes per sample.
 (4)(3)で解読した遺伝子(シークエンスデータ)をTopHat2(JOHNS HOPKINS大学)及びBowtie2(JOHNS HOPKINS大学)を用いて、ヒトゲノム配列にマップした。なお、ヒトゲノム配列情報及びヒト遺伝子情報には、NCBI(National Center for Biological Information)より公開されているBUILD GRCh38を使用した。マップした遺伝子について、Cufflinks(Washington大学)によって、解読した各遺伝子のリード数から、FPKM(Fragments Per Kilobase of exon per Million reads mapped)を単位とする遺伝子ごとの発現量を求めた。 The genes (sequence data) decoded in (4) and (3) were mapped to the human genome sequence using TopHat2 (JOHNS HOPKINS University) and Bowtie2 (JOHNS HOPKINS University). For human genome sequence information and human gene information, BUILD GRCh38 published by NCBI (National Center for Biological Information) was used. For the mapped genes, the expression level for each gene was determined in units of FPKM (fragments per kilobase of exon per million reads mapped) from the number of reads of each gene decoded by Cufflinks (University of Washington).
 3.T細胞のセクレトーム解析
 (1)上記1の(3)で高品質T細胞(12種類)及び低品質T細胞(12種類)に分類したT細胞と同じ株のT細胞(TCR未刺激)について、それぞれ、上記1の(1)~(2)と同様にしてTCR刺激磁性ビーズを用いてTCRを刺激し、培地Aで適宜培地交換しながら、5%CO、37℃の条件下で培養した。
3. Secretome analysis of T cells (1) For T cells (TCR unstimulated) of the same strain as the T cells classified into high-quality T cells (12 types) and low-quality T cells (12 types) in 1 (3) above, Each TCR was stimulated using TCR-stimulating magnetic beads in the same manner as in 1 (1) to (2) above, and cultured under the conditions of 5% CO 2 and 37° C. while exchanging the medium with medium A as appropriate. .
 (2)TCR刺激後6日目の細胞をそれぞれ1×10cells回収し、15%(w/v)透析FBS、4mmol/L L-グルタミン、ITS(インシュリン-トランスフェリン-亜セレン酸ナトリウム)サプリメント(×1)、50μg/mLアスコルビン酸、5ng/mL IL-7、及び5ng/mL IL-15を添加したαMEM(-Met)培地(α改変型イーグル最小必須培地、メチオニン不含有)で懸濁して、37℃、5%COの条件下で1時間培養後、0.00625mmol/L L-アジドホモアラニン(AHA)、15%(w/v)透析FBS、4mmol/L L-グルタミン、ITS(インシュリン-トランスフェリン-亜セレン酸ナトリウム)サプリメント(×1)、50μg/mLアスコルビン酸、5ng/mL IL-7、及び5ng/mL IL-15を添加したαMEM(-Met)培地を用いて、37℃、5%COの条件下で24時間培養した。 (2) 1×10 6 cells were collected from each of the cells 6 days after TCR stimulation, and 15% (w/v) dialyzed FBS, 4 mmol/L L-glutamine, ITS (insulin-transferrin-sodium selenite) supplement. (×1), suspended in αMEM (-Met) medium (α-modified Eagle minimal essential medium, methionine-free) supplemented with 50 μg/mL ascorbic acid, 5 ng/mL IL-7, and 5 ng/mL IL-15. 0.00625 mmol/L L- azidohomoalanine (AHA), 15% (w/v) dialyzed FBS, 4 mmol/L L-glutamine, ITS Using αMEM (-Met) medium supplemented with (insulin-transferrin-sodium selenite) supplement (x1), 50 μg/mL ascorbic acid, 5 ng/mL IL-7, and 5 ng/mL IL-15, 37 °C and 5% CO2 for 24 hours.
 (3)培養後、培養上清0.4mLを回収し、培養上清に分泌された、AHAで標識されたタンパク質(セクレトーム)を、Click-iTTM Protein Enrichment Kit,for click chemistry capture of azide-modified protein(Thermo Fisher Scientific社製)を用いて、前記AHAのアジド基をアルキンアガロース樹脂に捕捉させることで抽出した。 (3) After culturing, 0.4 mL of the culture supernatant was collected, and the AHA-labeled protein (secretome) secreted into the culture supernatant was collected using Click-iT Protein Enrichment Kit, for click chemistry capture of azide-. A modified protein (manufactured by Thermo Fisher Scientific) was used to capture the azide group of the AHA on alkyne agarose resin for extraction.
 (4)(3)で抽出したタンパク質をLC-MS/MS(Ultimate 3000 RSLCnano及びQ Exactive、いずれもThermo Fisher Scientific社製)で解析した。解析データから、Proteome Discovererソフトウェア(ver.2.2、Thermo Fisher Scientific社製)を用いて、抽出したタンパク質のアミノ酸配列を抽出した。 (4) The proteins extracted in (3) were analyzed by LC-MS/MS (Ultimate 3000 RSLCnano and Q Exactive, both manufactured by Thermo Fisher Scientific). From the analyzed data, the amino acid sequences of the extracted proteins were extracted using Proteome Discoverer software (ver.2.2, manufactured by Thermo Fisher Scientific).
 (5)Mascotソフトウェア(ver.2.5、Matrix Science 社製)を用い、下記(A)~(C)に記載のアミノ酸配列を組み合わせて構築したデータベースと照合することで、決定したアミノ酸配列からタンパク質を同定した。なお、有意なタンパク質の同定は、同定のスコア閾値を、False discovery rate(FDR)が1%になるように調整して行なった。同定したタンパク質は、Proteome Discoverer 2.2のLabel Free Quantification(LFQ)を用いて定量した。
(A)SwissProt(http://www.uniprot.org/)2018_11版に登録されているヒト(Homo sapiens)由来のタンパク質エントリー(計20413件);
(B)SwissProt(http://www.uniprot.org/)2018_11版に登録されているウシ(Bos taurus)由来のタンパク質エントリー(計5992件);
(C)The Global Proteome Machine Organization(http://www.thegpm.org/crap/index.html)から取得した、ヒト由来及びウシ由来以外のタンパク質汚染物のアミノ酸配列(計46件)。
(5) Using Mascot software (ver.2.5, manufactured by Matrix Science), by matching with a database constructed by combining the amino acid sequences described in (A) to (C) below, from the determined amino acid sequence protein was identified. The identification of significant proteins was performed by adjusting the identification score threshold so that the false discovery rate (FDR) was 1%. Identified proteins were quantified using Label Free Quantification (LFQ) in Proteome Discoverer 2.2.
(A) Homo sapiens-derived protein entries registered in SwissProt (http://www.uniprot.org/) version 2018_11 (20413 in total);
(B) protein entries from bovine (Bos taurus) registered in SwissProt (http://www.uniprot.org/) version 2018_11 (5992 in total);
(C) Amino acid sequences of human and non-bovine protein contaminants obtained from The Global Proteome Machine Organization (http://www.thegpm.org/crap/index.html) (46 in total).
 4.主成分分析(PCA)
 上記2のトランスクリプトーム解析の(4)及び上記3のセクレトーム解析の(5)より、以下の(a)~(d):
(a)day0の試料においてトランスクリプトーム解析によりマップされた12,829遺伝子(トランスクリプトームday0)及びそれらの発現量、
(b)day6の試料においてトランスクリプトーム解析によりマップされた12,270遺伝子(トランスクリプトームday6)及びそれらの発現量、
(c)day14の試料においてトランスクリプトーム解析によりマップされた12,013遺伝子(トランスクリプトームday14)及びそれらの発現量、
(d)セクレトーム解析により同定されたタンパク質をコードする800遺伝子(セクレトーム)及びそれらの発現量
のデータが得られた。ただし、(a)~(d)の各遺伝子数には、低発現又は無発現の遺伝子を含めていない。各T細胞(23種類:細胞No.1~21、23、24)における、これら(a)~(d)を結合した全37,912遺伝子のデータについて、主成分分析(PCA)を行なった。なお、細胞No.22のT細胞は、上記2のトランスクリプトーム解析におけるday14の細胞が得られなかったため、結果に含めていない。
4. Principal component analysis (PCA)
From (4) of the transcriptome analysis in 2 above and (5) of the secretome analysis in 3 above, the following (a) to (d):
(a) 12,829 genes mapped by transcriptome analysis in day0 samples (transcriptome day0) and their expression levels,
(b) 12,270 genes mapped by transcriptome analysis in day6 samples (transcriptome day6) and their expression levels;
(c) 12,013 genes mapped by transcriptome analysis in day14 samples (transcriptome day14) and their expression levels,
(d) Data on 800 genes (secretome) encoding proteins identified by secretome analysis and their expression levels were obtained. However, the numbers of genes in (a) to (d) do not include low-expressed or non-expressed genes. A principal component analysis (PCA) was performed on the data of all 37,912 genes combining these (a) to (d) in each T cell (23 types: cell Nos. 1 to 21, 23, 24). In addition, the cell no. 22 T cells were not included in the results because day 14 cells were not obtained in the transcriptome analysis of 2 above.
 前記PCAで得られた散布図を図1に示す。図1より、高品質T細胞(細胞No.1~12)では第一主成分(PC1)スコアが高く、低品質T細胞(細胞No.13~21、23、24)ではPC1スコアが低かった。23種類の増殖倍率の降順(増殖順位)とPC1スコア(PC1)及びその降順(PC1順位)との関係を下記の表12及び図2に示す。 The scatter diagram obtained by the PCA is shown in Figure 1. From FIG. 1, the first principal component (PC1) score was high in high-quality T cells (cells No. 1-12), and the PC1 score was low in low-quality T cells (cells No. 13-21, 23, 24). . Table 12 below and FIG. 2 show the relationship between the descending order (proliferation order) of the 23 types of proliferation rate, the PC1 score (PC1), and the descending order (PC1 order).
Figure JPOXMLDOC01-appb-T000012
 

 
Figure JPOXMLDOC01-appb-T000012
 

 
 表12及び図2より、T細胞の増殖順位とPC1順位との間の相関係数(R)は0.9615となり、細胞の品質(この場合、増殖能)とPC1スコアとの間に高い相関が認められた。PCAでは、主成分スコアの係数と主成分負荷量の値とはほぼ比例するため、第一主成分の主成分負荷量の値が高い変数(この場合、変数は各遺伝子の発現量)は、第一主成分の正方向に寄与し、第一主成分の主成分負荷量の値が低い(負値の)変数は、第一主成分の負方向に寄与する。そのため、第一主成分の主成分負荷量の値が高い遺伝子は、PC1スコアが高い細胞、すなわち、高品質T細胞で発現し、第一主成分の主成分負荷量の値が低い遺伝子は、PC1スコアが低い細胞、すなわち、低品質T細胞で発現することがわかった。 From Table 12 and FIG. 2, the correlation coefficient (R) between the T cell proliferation rank and the PC1 rank is 0.9615, indicating a high correlation between the cell quality (in this case, proliferative capacity) and the PC1 score. was accepted. In PCA, since the coefficient of the principal component score and the value of the principal component loading are almost proportional, the variable with the high value of the principal component loading of the first principal component (in this case, the variable is the expression level of each gene) A variable that contributes positively to the first principal component and has a low (negative value) value of the principal component loading of the first principal component contributes negatively to the first principal component. Therefore, genes with a high principal component loading value of the first principal component are expressed in cells with a high PC1 score, that is, high-quality T cells, and genes with a low principal component loading value of the first principal component are expressed in It was found to be expressed in cells with low PC1 score, ie low quality T cells.
 そこで、前記PCAより、上記(a)~(d)で得られた各遺伝子の発現量について、第一主成分の主成分負荷量(principal component loading)をそれぞれ求めた。上記(a)~(d)の全37,912遺伝子のうち、第一主成分への寄与率が高く(具体的には、遺伝子の発現量の第一主成分に対する主成分負荷量の絶対値が0.9以上)、かつ、膜貫通型タンパク質をコードする遺伝子は、上記の表1~2に示した遺伝子である。また、第一主成分への寄与率が高く(具体的には、遺伝子の発現量の第一主成分に対する主成分負荷量の絶対値が0.9以上)、かつ、膜貫通型タンパク質以外のタンパク質をコードする遺伝子は、上記の表3~5に示した遺伝子である。表1~5には、各遺伝子について、「由来データ」の欄に上記(a)~(d)の別をそれぞれ示し、また、上記で得られた第一主成分に対する主成分負荷量も合わせて示した。 Therefore, from the PCA, the principal component loading of the first principal component was determined for each gene expression level obtained in (a) to (d) above. Among all 37,912 genes in (a) to (d) above, the contribution rate to the first principal component is high (specifically, the absolute value of the principal component loading for the first principal component of the gene expression level is 0.9 or more) and genes encoding transmembrane proteins are the genes shown in Tables 1 and 2 above. In addition, the contribution rate to the first principal component is high (specifically, the absolute value of the principal component loading amount with respect to the first principal component of the gene expression level is 0.9 or more), and other than transmembrane proteins Genes encoding proteins are those shown in Tables 3-5 above. In Tables 1 to 5, for each gene, the categories (a) to (d) above are shown in the column of "origin data", respectively, and the principal component loadings for the first principal component obtained above are also included. indicated.
 
 5.T細胞の品質(増殖能)とマーカータンパク質の発現との相関性(その1)
 「4.主成分分析」の結果、第一主成分への寄与率が高い遺伝子の中から、ITGB7(ヌクレオチド配列番号84)、BMPR2(ヌクレオチド配列番号92)、TP53I3(ヌクレオチド配列番号104)、及びLGALS8(ヌクレオチド配列番号121)を選択し、これら遺伝子から翻訳されるタンパク質の発現量とT細胞の品質との相関性を確認した。

5. Correlation between T cell quality (proliferative ability) and marker protein expression (part 1)
As a result of “4. LGALS8 (nucleotide sequence number 121) was selected, and the correlation between the expression levels of proteins translated from these genes and the quality of T cells was confirmed.
 (1)TCR刺激前の細胞(day0)試料調製
(1-1)10種類のT細胞株(表11に示す細胞No.5、10、11、13、15、16、18、19、22、24)を解凍し、96wellプレートの培地A(15%(w/v)FBS(BioWest社製)、5ng/mL IL-7、及び5ng/mL IL-15を添加したαMEM)に、それぞれ1×10cells/wellで播種した。
(1-2)播種した細胞を、Phosphate Buffered saline(PBS)(-)1mLに懸濁し凍結融解を3回繰り返す操作、又はLysis buffer 3(Mybiosource社製)1mL添加する操作により、溶解し、これを「刺激前細胞試料(day0)」とした。
(1) Cell sample preparation before TCR stimulation (day 0) (1-1) 10 types of T cell lines (cell Nos. 5, 10, 11, 13, 15, 16, 18, 19, 22, 24) was thawed and added to 96-well plate medium A (αMEM supplemented with 15% (w/v) FBS (manufactured by BioWest), 5 ng/mL IL-7, and 5 ng/mL IL-15), respectively. Seeded at 10 6 cells/well.
(1-2) The seeded cells were suspended in 1 mL of Phosphate Buffered Saline (PBS) (-) and freeze-thawed three times, or dissolved by adding 1 mL of Lysis buffer 3 (manufactured by Mybiosource). was defined as "pre-stimulation cell sample (day 0)".
 (2)TCR刺激後の細胞(day2)試料調製
(2-1)11種類のiPS-T細胞株(表11に示す細胞No.5、10、11、13、16、18~20、22~24)を解凍し、96wellプレートの培地Aに、それぞれ1×10cells/wellで播種した。
(2-2)T細胞受容体(TCR)刺激磁性ビーズ(Dynabeads Human T-Activator CD3/CD28 for T Cell Expansion and Activation(Thermo Fisher Scientific社製))を用いて、播種したT細胞のTCRをそれぞれ刺激後、培地Aで適宜培地交換しながら、5%CO、37℃の条件下で2日間培養した。
(2-3)培養後のiPS-T細胞を回収した(回収量:0.7×10~1×10cells)。
(2-4)回収した細胞を、(1-2)に記載の方法で溶解し、これを「刺激後細胞試料(day2)」とした。
(2) Sample preparation of cells (day 2) after TCR stimulation (2-1) 11 types of iPS-T cell lines (cell Nos. 5, 10, 11, 13, 16, 18-20, 22- 24) was thawed and seeded onto medium A of a 96-well plate at 1×10 6 cells/well.
(2-2) Using T cell receptor (TCR) stimulation magnetic beads (Dynabeads Human T-Activator CD3/CD28 for T Cell Expansion and Activation (manufactured by Thermo Fisher Scientific)), the TCR of the seeded T cells is induced. After the stimulation, the cells were cultured under conditions of 5% CO 2 and 37° C. for 2 days while appropriately replacing the medium with medium A.
(2-3) The cultured iPS-T cells were recovered (collected amount: 0.7×10 7 to 1×10 7 cells).
(2-4) The collected cells were lysed by the method described in (1-2), and this was used as the "post-stimulation cell sample (day 2)".
 (3)タンパク質発現量測定
 (1)で調製した刺激前細胞試料(day0)、及び(2)で調製した刺激後細胞試料(day2)について、下記の市販のキットを用いて、ELISA法による各タンパク質発現量測定を行なった。
ITGB7(アミノ酸配列番号7):Enzyme-linked Immunosorbent Assay Kit For Integrin Beta 7(ITGb7)(Mybiosource社製)
BMPR2(アミノ酸配列番号15):Human Bone Morphogenetic Protein Receptor II(BMPR-II) ELISA Kit(Mybiosource社製)
TP53I3(アミノ酸配列番号27):TP53I3 elisa kit(Mybiosource社製)
LGALS8(アミノ酸配列番号44):Human Galectin 8 ELISA Kit(Mybiosource社製)。
(3) Protein expression level measurement For the pre-stimulation cell sample (day0) prepared in (1) and the post-stimulation cell sample (day2) prepared in (2), using the following commercially available kit, each by ELISA method Protein expression level measurement was performed.
ITGB7 (amino acid sequence number 7): Enzyme-linked Immunosorbent Assay Kit For Integrin Beta 7 (ITGb7) (manufactured by Mybiosource)
BMPR2 (amino acid SEQ ID NO: 15): Human Bone Morphogenetic Protein Receptor II (BMPR-II) ELISA Kit (manufactured by Mybiosource)
TP53I3 (amino acid sequence number 27): TP53I3 elisa kit (manufactured by Mybiosource)
LGALS8 (amino acid sequence number 44): Human Galectin 8 ELISA Kit (manufactured by Mybiosource).
 (4)相関係数算出
 前記4つのタンパク質(ITGB7、BMPR2、TP53I3、及びLGALS8)について、増殖能(対数値)と前記タンパク質発現量(対数値)との相関係数を算出した。結果を下記の表13に示す。
(4) Correlation Coefficient Calculation For the four proteins (ITGB7, BMPR2, TP53I3, and LGALS8), correlation coefficients between the proliferation potential (logarithmic value) and the protein expression level (logarithmic value) were calculated. The results are shown in Table 13 below.
Figure JPOXMLDOC01-appb-T000013
 
Figure JPOXMLDOC01-appb-T000013
 
 6.T細胞の品質(増殖能)とマーカータンパク質の発現との相関性(その2)
 「4.主成分分析」の結果、第一主成分への寄与率が高い遺伝子の中から、ALCAM(ヌクレオチド配列番号82)を選択し、当該遺伝子から翻訳されるタンパク質の発現量とT細胞の品質との相関性を確認した。
6. Correlation between T cell quality (proliferative ability) and marker protein expression (part 2)
As a result of “4. Correlation with quality was confirmed.
 (1)18種類のT細胞株(表11に示す細胞No.5、8~24)を0.5×10~1×10cells取得し、5.(1-2)に記載の方法で細胞を溶解して「刺激前細胞試料(day0)」とした。 (1) Obtaining 0.5×10 7 to 1×10 7 cells of 18 kinds of T cell lines (Cell Nos. 5 and 8 to 24 shown in Table 11); Cells were lysed by the method described in (1-2) to obtain a “pre-stimulation cell sample (day 0)”.
 (2)(1)で調製した刺激前細胞試料(day0)をタンパク質量が30μgとなるようにそろえた後、抗体アレイ(Signaling Explorer Antibody Array、Full Moon BioSystems社製)に添加し、付属のプロトコールにしたがってALCAMタンパク質発現量測定を行なった。 (2) After aligning the pre-stimulated cell sample (day 0) prepared in (1) so that the amount of protein is 30 μg, add it to the antibody array (Signaling Explorer Antibody Array, Full Moon Bio Systems) and follow the attached protocol. ALCAM protein expression level measurement was performed according to.
 (3)5.(4)と同様の方法で相関係数を算出した。結果を下記の表14に示す。 (3) 5. Correlation coefficients were calculated in the same manner as in (4). The results are shown in Table 14 below.
Figure JPOXMLDOC01-appb-T000014
 
Figure JPOXMLDOC01-appb-T000014
 
 表13及び表14では、相関係数の絶対値が0.5以上であれば「相関性あり」と判断した。表13及び表14に示したように、刺激前細胞試料(day0)では、ALCAM(アミノ酸配列番号5)及びTP53I3(アミノ酸配列番号27)で増殖能とタンパク質の発現量との相関を確認した。このことから、ALCAM及びTP53I3は、定常状態のときにT細胞の品質を評価できるマーカーといえる。一方、刺激後細胞試料(day2)では、ITGB7(アミノ酸配列番号7)、BMPR2(アミノ酸配列番号15)、及びLGALS8(アミノ酸配列番号44)で増殖能とタンパク質の発現量との相関を確認した。このことから、ITGB7、BMPR2、及びLGALS8は、TCRによる細胞刺激後の状態のときにT細胞の品質を評価できるマーカーといえる。また、これらの中でも、ALCAM、BMPR2、及びTP53I3では、相関係数の絶対値が0.6以上であり、これら3つのタンパク質は、T細胞の品質を評価できるマーカーの特に好ましい態様といえる。 In Tables 13 and 14, if the absolute value of the correlation coefficient was 0.5 or more, it was judged to be "correlated". As shown in Tables 13 and 14, in the pre-stimulation cell sample (day 0), ALCAM (amino acid SEQ ID NO: 5) and TP53I3 (amino acid SEQ ID NO: 27) confirmed the correlation between proliferation ability and protein expression level. This suggests that ALCAM and TP53I3 are markers that can assess the quality of T cells at steady state. On the other hand, in the post-stimulation cell sample (day 2), ITGB7 (amino acid sequence number 7), BMPR2 (amino acid sequence number 15), and LGALS8 (amino acid sequence number 44) were confirmed to have a correlation between proliferation ability and protein expression level. Therefore, ITGB7, BMPR2, and LGALS8 can be said to be markers capable of evaluating the quality of T cells in the state after cell stimulation with TCR. Among these, ALCAM, BMPR2, and TP53I3 have absolute values of correlation coefficients of 0.6 or more, and these three proteins can be said to be particularly preferred embodiments of markers capable of evaluating the quality of T cells.
 本発明によれば、新たな指標によってT細胞の品質を評価できるT細胞の評価方法、及びそれに用いるT細胞評価用試薬を提供することが可能となる。 According to the present invention, it is possible to provide a T cell evaluation method that can evaluate the quality of T cells using a new index, and a T cell evaluation reagent used for the method.

Claims (17)

  1.  T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
     前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
    を含む、T細胞の評価方法。
    (I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    a detection step of detecting the expression of at least one gene selected from the group consisting of the following genes (I) to (III) in T cells;
    an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
    A method for evaluating T cells, comprising:
    (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  2.  前記検出工程が、前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を用いて前記発現産物を検出する工程である、請求項1に記載の方法。 The detecting step is a step of detecting the expression product using a probe molecule that specifically binds to the expression product of at least one gene selected from the group consisting of the genes (I) to (III). A method according to claim 1.
  3.  前記発現産物が、前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドであり、かつ、前記プローブ分子が、前記ポリペプチドに特異的に結合する抗体及び/又はアプタマーである、請求項2に記載の方法。 The expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule is an antibody that specifically binds to the polypeptide and/or or an aptamer.
  4.  前記評価工程が、前記遺伝子の発現を指標としてT細胞の増殖能を評価する工程である、請求項1~3のいずれか一項に記載の方法。  The method according to any one of claims 1 to 3, wherein the evaluation step is a step of evaluating the proliferative ability of T cells using the expression of the gene as an index.
  5.  前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、請求項1~4のいずれか一項に記載の方法。
    (I’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') , the method according to any one of claims 1 to 4.
    (I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 5, 7, 15, 27, and 44 (II') SEQ ID NOS: 5, 7, 15, 27, and 44 A gene (III') encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence according to any one of SEQ ID NO: A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence of any one of 5, 7, 15, 27 and 44.
  6.  前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、請求項1~5のいずれか一項に記載の方法。
    (I’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') , the method according to any one of claims 1 to 5.
    (I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 15, and 27; (II') set forth in any one of SEQ ID NOs: 5, 15, and 27; Any of gene (III') SEQ ID NOS: 5, 15, and 27 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence or a gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in one.
  7.  請求項2~6のいずれか一項に記載の方法に用いるための試薬であり、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現産物に特異的に結合するプローブ分子を含有する、T細胞評価用試薬。
    (I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    A reagent for use in the method according to any one of claims 2 to 6, which is specific for the expression product of at least one gene selected from the group consisting of the following genes (I) to (III) T cell evaluation reagent containing a probe molecule that binds to
    (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  8.  前記発現産物が、前記(I)~(III)の遺伝子のうちのいずれか1つにコードされるポリペプチドであり、かつ、前記プローブ分子が、前記ポリペプチドに特異的に結合する抗体及び/又はアプタマーである、請求項7に記載の試薬。 The expression product is a polypeptide encoded by any one of the genes (I) to (III), and the probe molecule is an antibody that specifically binds to the polypeptide and/or or an aptamer, the reagent of claim 7.
  9.  前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、請求項7又は8に記載の試薬。
    (I’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III’)配列番号5、7、15、27、及び44のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') , a reagent according to claim 7 or 8.
    (I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 5, 7, 15, 27, and 44 (II') SEQ ID NOS: 5, 7, 15, 27, and 44 A gene (III') encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence according to any one of SEQ ID NO: A gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence of any one of 5, 7, 15, 27 and 44.
  10.  前記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子が、下記(I’)~(III’)の遺伝子からなる群から選択される少なくとも1種の遺伝子である、請求項7~9のいずれか一項に記載の試薬。
    (I’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III’)配列番号5、15、及び27のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    At least one gene selected from the group consisting of the genes (I) to (III) is at least one gene selected from the group consisting of the following genes (I') to (III') , the reagent according to any one of claims 7-9.
    (I') a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 5, 15, and 27; (II') set forth in any one of SEQ ID NOs: 5, 15, and 27; Any of gene (III') SEQ ID NOS: 5, 15, and 27 encoding a polypeptide comprising an amino acid sequence in which one or more amino acid residues are substituted, deleted, inserted, and/or added in the amino acid sequence or a gene encoding a polypeptide comprising an amino acid sequence having 70% or more identity with the amino acid sequence set forth in one.
  11.  請求項1~6のいずれか一項に記載のT細胞の評価方法により得られた高品質T細胞。 High-quality T cells obtained by the T cell evaluation method according to any one of claims 1 to 6.
  12.  請求項11に記載の高品質T細胞を含有する医薬組成物。 A pharmaceutical composition containing the high-quality T cells according to claim 11.
  13.  がんの予防又は治療に使用するための請求項12に記載の医薬組成物。 The pharmaceutical composition according to claim 12 for use in preventing or treating cancer.
  14.  請求項12に記載の医薬組成物を用いる、がんの予防または治療方法。 A method for preventing or treating cancer using the pharmaceutical composition according to claim 12.
  15.  がんを有するかまたはがんを有する危険性のある被験者に、請求項12に記載の医薬組成物を投与することを含む、がんを治療または予防する方法。 A method of treating or preventing cancer, comprising administering the pharmaceutical composition of claim 12 to a subject who has cancer or is at risk of having cancer.
  16.  がんを有するかまたはがんを有する危険性のある被験者に、T細胞を投与することを含む、がんを治療または予防する方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
     前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
    を含む、方法。
    (I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    A method of treating or preventing cancer, comprising administering T cells to a subject who has cancer or is at risk of having cancer, wherein the following (I) to (III) in the T cells ) detecting the expression of at least one gene selected from the group consisting of genes;
    an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
    A method, including
    (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
  17.  T細胞の製造方法であって、該T細胞における、下記(I)~(III)の遺伝子からなる群から選択される少なくとも1種の遺伝子の発現を検出する検出工程と、
     前記遺伝子の発現を指標としてT細胞の品質を評価する評価工程と、
    を含む、T細胞の製造方法。
    (I)配列番号1~77のいずれか1つに記載のアミノ酸配列を含むポリペプチドをコードする遺伝子
    (II)配列番号1~77のいずれか1つに記載のアミノ酸配列において、1若しくは複数個のアミノ酸残基が置換、欠失、挿入、及び/又は付加されたアミノ酸配列を含むポリペプチドをコードする遺伝子
    (III)配列番号1~77のいずれか1つに記載のアミノ酸配列と70%以上の同一性を有するアミノ酸配列を含むポリペプチドをコードする遺伝子。
    A method for producing T cells, comprising a detection step of detecting expression of at least one gene selected from the group consisting of the following genes (I) to (III) in the T cells;
    an evaluation step of evaluating the quality of T cells using the expression of the gene as an index;
    A method of producing a T cell, comprising:
    (I) a gene encoding a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOs: 1-77 (II) one or more of the amino acid sequences set forth in any one of SEQ ID NOs: 1-77 70% or more of the amino acid sequence described in any one of gene (III) SEQ ID NOS: 1 to 77 encoding a polypeptide comprising an amino acid sequence in which amino acid residues are substituted, deleted, inserted, and / or added A gene encoding a polypeptide comprising an amino acid sequence having the identity of
PCT/JP2022/039806 2021-10-27 2022-10-26 T cell evaluation method and composition for t cell evaluation WO2023074711A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2021-175126 2021-10-27
JP2021175126A JP2023064788A (en) 2021-10-27 2021-10-27 Evaluation method of t-cell, and composition for evaluating t-cell

Publications (1)

Publication Number Publication Date
WO2023074711A1 true WO2023074711A1 (en) 2023-05-04

Family

ID=86158030

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2022/039806 WO2023074711A1 (en) 2021-10-27 2022-10-26 T cell evaluation method and composition for t cell evaluation

Country Status (2)

Country Link
JP (1) JP2023064788A (en)
WO (1) WO2023074711A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170233808A1 (en) * 2014-10-17 2017-08-17 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of t-cell exhaustion using cd39 biomarkers and modulators
JP2019154320A (en) * 2018-03-13 2019-09-19 ロート製薬株式会社 Method for evaluating quality of cell, and quality determination kit of cell
JP2021501318A (en) * 2017-10-27 2021-01-14 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and Compositions for Treating Diseases Related to Exhausted T Cells

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170233808A1 (en) * 2014-10-17 2017-08-17 Dana-Farber Cancer Institute, Inc. Compositions and methods for identification, assessment, prevention, and treatment of t-cell exhaustion using cd39 biomarkers and modulators
JP2021501318A (en) * 2017-10-27 2021-01-14 ザ トラスティーズ オブ ザ ユニバーシティ オブ ペンシルバニア Methods and Compositions for Treating Diseases Related to Exhausted T Cells
JP2019154320A (en) * 2018-03-13 2019-09-19 ロート製薬株式会社 Method for evaluating quality of cell, and quality determination kit of cell

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GEORGE ELIAS; BENSON OGUNJIMI; VIGGO VAN TENDELOO: "Tracking Dye‐Independent Approach to Identify and Isolate In Vitro Expanded T Cells", CYTOMETRY A, WILEY-LISS, HOBOKEN, USA, vol. 95, no. 10, 29 July 2019 (2019-07-29), Hoboken, USA, pages 1096 - 1107, XP072332516, ISSN: 1552-4922, DOI: 10.1002/cyto.a.23867 *

Also Published As

Publication number Publication date
JP2023064788A (en) 2023-05-12

Similar Documents

Publication Publication Date Title
JP6259012B2 (en) How to treat alopecia
Maazi et al. Activated plasmacytoid dendritic cells regulate type 2 innate lymphoid cell–mediated airway hyperreactivity
Hughes et al. Interleukin-1β selectively expands and sustains interleukin-22+ immature human natural killer cells in secondary lymphoid tissue
US9125869B2 (en) Method for expanding monocytes
JP2021534783A (en) Method for producing chimeric antigen receptor-expressing cells
JP6416131B2 (en) Method for producing an enriched tumor-reactive T cell population from a tumor
Reutner et al. CD27 expression discriminates porcine T helper cells with functionally distinct properties
US20190353643A1 (en) Method For Sorting Highly Effective Stem Cells For Treating Immune Disorder
US20230330145A1 (en) Natural killer cells
CN114929752A (en) Chimeric cytokine receptors
Teteloshvili et al. Involvement of MicroRNAs in the aging-related decline of CD28 expression by human T cells
Talker et al. Monocyte biology conserved across species: Functional insights from cattle
Richert et al. Expansion of antigen-specific T cells from cerebrospinal fluid of patients with multiple sclerosis
Flinn et al. Thymopoiesis, alterations in dendritic cells and tregs, and reduced T cell activation in successful extracorporeal photopheresis treatment of GVHD
Davies et al. Robust production of merkel cell polyomavirus oncogene specific T cells from healthy donors for adoptive transfer
WO2023074711A1 (en) T cell evaluation method and composition for t cell evaluation
Crispim et al. Interferon-γ+ 874 polymorphism in the first intron of the human interferon-γ gene and kidney allograft outcome
WO2023074712A1 (en) Method for evaluating t cells and composition for evaluating t cells
WO2023074713A1 (en) T cell quality evaluation method, and reagent to be used therein
US9678062B2 (en) Methods and compositions for expanding cells and improving engraftment
CN113227357A (en) Accelerated differentiation of human hematopoietic stem cells into mature natural killer cells with enhanced antibody-dependent cytotoxic activity
Yu et al. Mechanism study of ubiquitination in T cell development and autoimmune disease
Mumprecht et al. Chronic myelogenous leukemia maintains specific CD8+ T cells through IL‐7 signaling
Dawson et al. Functional effects of chimeric antigen receptor co-receptor signaling domains in human Tregs
Kustermann et al. Adoptively transferred in vitro-generated myeloid-derived suppressor cells improve T-cell function and antigen-specific immunity after traumatic lung injury

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: 22887037

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE