[go: up one dir, main page]

WO2021100585A1 - Method for producing chimeric antigen receptor gene-modified lymphocytes - Google Patents

Method for producing chimeric antigen receptor gene-modified lymphocytes Download PDF

Info

Publication number
WO2021100585A1
WO2021100585A1 PCT/JP2020/042187 JP2020042187W WO2021100585A1 WO 2021100585 A1 WO2021100585 A1 WO 2021100585A1 JP 2020042187 W JP2020042187 W JP 2020042187W WO 2021100585 A1 WO2021100585 A1 WO 2021100585A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
antibody
preparation
culture
car
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/JP2020/042187
Other languages
French (fr)
Japanese (ja)
Inventor
信博 西尾
義行 ▲高▼橋
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tokai National Higher Education and Research System NUC
Original Assignee
Tokai National Higher Education and Research System NUC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tokai National Higher Education and Research System NUC filed Critical Tokai National Higher Education and Research System NUC
Priority to JP2021558324A priority Critical patent/JPWO2021100585A1/ja
Publication of WO2021100585A1 publication Critical patent/WO2021100585A1/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/40Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
    • A61K40/41Vertebrate antigens
    • A61K40/42Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/11T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the present invention relates to genetically modified lymphocytes expressing chimeric antigen receptors (CAR transgenic lymphocytes). Specifically, the present invention relates to a method for producing CAR gene-introduced lymphocytes, uses of the cells, and the like.
  • CAR transgenic lymphocytes chimeric antigen receptors
  • CAR-T therapy Genetically modified T cell therapy
  • CAR-NK therapy genetically modified NK cell therapy
  • CAR-NK therapy genetically modified NK cell therapy
  • CAR typically comprises a structure in which the single-chain variable region of an antibody is the extracellular domain, which is linked to the transmembrane domain, the CD3 ⁇ , and the intracellular domain of the molecule that transmits the co-stimulation signal.
  • CAR transgenic lymphocytes are activated by binding to the antigen according to the specificity of the antibody, damaging target cells (such as cancer cells).
  • CAR therapy has advantages such as relatively easy cell preparation, high cytotoxic activity, and long-lasting effect, and is particularly resistant to refractory and conventional treatments.
  • CAR for the CD19 antigen expressed on the cell surface is gene-introduced into peripheral blood T cells collected from the patients, cultured, and infused.
  • the test has been conducted in Europe and the United States, and good results with a remission rate of 80 to 90% have been reported (Non-Patent Documents 1 to 3).
  • CAR therapy is attracting attention as one of the most promising treatments for refractory cancer in the United States.
  • Non-Patent Document 4 Non-Patent Document 4
  • the transposon method which is one of the gene modification techniques using non-viral vectors
  • the transposon method enables permanent gene transfer like the viral vector method
  • the gene transfer efficiency is lower than that of the viral vector method, and it is suitable for gene transfer operations (electroporation and its improvement methods, etc.).
  • the cells are damaged, and there is a problem that the cell viability and the cell proliferation rate decrease.
  • the research group of the present inventors said, "Protecting lymphocytes after CAR gene transfer by co-culturing with activated T cells prepared separately, gene transfer efficiency and cells.
  • the main object of the present invention is to provide a novel preparation (culture) means for increasing the number of CAR cells, and to contribute to the progress of clinical application of CAR therapy and the improvement of therapeutic results.
  • the present inventors have two strategies, that is, a method of stimulating cells after CAR gene transfer (CAR cells) with an anti-iditope antibody (first strategy), and We have created a method (second strategy) for stimulating cells after CAR gene transfer (CAR cells) with cells expressing the target antigen.
  • the first strategy is to directly stimulate CAR cells with anti-iditope antibodies, and CAR cell-specific proliferation can be expected.
  • a significant increase in the number of CAR cells improved in gene transfer efficiency / cell proliferation rate
  • was observed as compared with the conventional method stimulation with anti-CD3 antibody / anti-CD28 antibody
  • the second strategy stimulation by the target antigen is added during the culture of CAR cells, and CAR cell-specific proliferation can be expected as in the first strategy.
  • the two new strategies make it possible or facilitate to secure the number of cells required for treatment, and their clinical significance is extremely large.
  • the inventions shown below are based on these two strategies.
  • a method for preparing a genetically modified lymphocyte expressing a chimeric antigen receptor which comprises the following steps (1) to (3): (1) A step of preparing a genetically modified lymphocyte into which a target antigen-specific chimeric antigen receptor gene has been introduced; (2) The genetically modified lymphocyte prepared in step (1) is used in the presence of an anti-iditope antibody against the antigen recognition region of the chimeric antigen receptor on which it is expressed, or the target antigen of the chimeric antigen receptor on which it is expressed. The step of culturing in the presence of expressing cells; and (3) the step of collecting the genetically modified lymphocytes after culturing.
  • step (2) is performed after 8 to 48 hours have passed from the operation of introducing the target antigen-specific chimeric antigen receptor gene in step (1).
  • step 5 The preparation method according to any one of [1] to [4], wherein the culture period of step (2) is 1 to 14 days.
  • [6] The preparation method according to any one of [1] to [5], wherein the culture in step (2) is not stimulated by the anti-CD3 antibody and the anti-CD28 antibody.
  • [7] The preparation method according to any one of [1] to [6], wherein the anti-iditope antibody is a solid phase antibody or a beaded antibody.
  • Any of [1] to [7], wherein the genetically modified lymphocyte of step (1) can be obtained by introducing a target antigen-specific chimeric antigen receptor gene into a cell population containing T cells or progenitor cells thereof.
  • [9] The preparation method according to [8], wherein the cell population is peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • the cells expressing the target antigen in step (2) are prepared by introducing the gene encoding the target antigen into peripheral blood mononuclear cells (PBMCs), according to [1] to [9].
  • the preparation method according to any one item. [10-1]
  • the transposon method is used for the introduction of the target antigen-specific chimeric antigen receptor gene in step (1). Any one of [1] to [10], wherein the cell expressing the target antigen in step (2) is prepared by introducing a gene encoding the target antigen into peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • step (a) The preparation method according to any one of [1] to [15], wherein the following step (a) is performed between steps (1) and (2): (A) Non-proliferative cells obtained by stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody and then performing a treatment for losing proliferative ability, and genetically modified lymphocytes prepared in step (1). A step of mixing lymphocytes and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody.
  • step (b) The preparation method according to any one of [1] to [15], wherein the following step (b) is performed between steps (1) and (2): (B) Non-proliferative PBMCs obtained by treating peripheral blood mononuclear cells (PBMCs) as they are or by stimulating them with anti-CD3 antibody and anti-CD28 antibody and then performing a treatment to lose proliferative ability, and step (1). The step of mixing and co-culturing the genetically modified lymphocytes prepared in.
  • PBMCs peripheral blood mononuclear cells
  • step (c) The preparation method according to any one of [1] to [15], wherein the following step (c) is performed between steps (1) and (2): (C) A cell population containing T cells was stimulated with an anti-CD3 antibody and an anti-CD28 antibody, and then cultured in the presence of the viral peptide antigen and treated to lose the proliferative ability to retain the viral peptide antigen. A step of mixing non-proliferative cells with the transgenic lymphocytes prepared in step (1) and co-culturing them. [19] The preparation method according to [16] or [18], wherein the cell population containing T cells is peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • [20] The preparation method according to any one of [16] to [19], wherein the treatment for losing the proliferative ability is irradiation.
  • [21] The preparation method according to any one of [16] to [20], wherein the non-proliferative cells and the genetically modified lymphocytes are derived from the same individual.
  • [22] A genetically modified lymphocyte expressing a chimeric antigen receptor obtained by the preparation method according to any one of [1] to [21].
  • [23] A cell preparation containing a therapeutically effective amount of the genetically modified lymphocyte according to [22].
  • a method for treating cancer which comprises a step of administering a therapeutically effective amount of the genetically modified lymphocyte according to [22] to a cancer patient.
  • Preparation of CAR-T cells by the conventional method (culture method 1).
  • Preparation of CAR-T cells by a novel method (culture method 2).
  • Preparation of CAR-T cells by a novel method (culture method 3).
  • Composition of pIRII-CAR.CD19.28z vector (SEQ ID NO: 1).
  • the CD19CAR gene has a structure sandwiched between a 5'inverted repeat sequence (5'IR) and a 3'inverted repeat sequence (3'IR).
  • CD19CAR has a leader sequence (SEQ ID NO: 2), light chain variable region (VL) (SEQ ID NO: 3), heavy chain variable region (VH) (SEQ ID NO: 4), Fc region (CH2, CH3) (SEQ ID NO: 5), Includes the transmembrane region of CD28 and the intracellular domain (SEQ ID NO: 6) and CD3 ⁇ (SEQ ID NO: 7).
  • Composition of pCMV-pigBac vector SEQ ID NO: 8).
  • the piggyBac transposase gene is located under the control of the CMV earliest promoter (CMV immediate ear1y promoter). Improvement of gene transfer efficiency by a new method (culture method 2).
  • the cells on the 14th day were evaluated by flow cytometry, first cultured for 7 days by the conventional method (culture method 1), then cultured for 7 days by the new method (culture method 2) (upper right), and first 7 After culturing by the conventional method (culture method 1) for 1 day, the gene transfer efficiency was further compared with that cultivated by the conventional method (culture method 1) for 7 days (lower right). Improvement of gene transfer efficiency by a new method (culture method 2). First cultivated by the conventional method (culture method 1) for 7 days, then cultivated by the new method (culture method 2) for 7 days (left), and first cultivated by the conventional method (culture method 1) for 7 days, and then.
  • the proliferation rates of CAR-T cells from the 7th day were compared with those cultured for 7 days by the conventional method (culture method 1) (right).
  • the Fc region CH2, CH3 has been deleted.
  • Improvement of gene transfer efficiency by a new method (culture method 2).
  • the cells on day 7 were evaluated by flow cytometry, and the gene transfer efficiency was compared between the novel method (culture method 2) (upper right) and the conventional method (culture method 1) (lower right). Improvement of gene transfer efficiency by a new method (culture method 2).
  • the number of CAR-T cells was compared between the new method (culture method 2) (left) and the conventional method (culture method 1) (right).
  • CAR cells genetically modified lymphocytes expressing chimeric antigen receptors
  • CAR cells CAR transgenic lymphocytes
  • the preparation method CAR cells (typically CAR-T cells) obtained by the preparation method of the present invention can be used for CAR therapy.
  • the following steps (1) to (3) are performed.
  • various cells for example, T cells in the present specification are human cells.
  • Step of preparing a genetically modified lymphocyte into which a target antigen-specific chimeric antigen receptor gene has been introduced (2) The gene-modified lymphocyte prepared in step (1) is expressed by the chimeric antigen receptor antigen. Steps of culturing in the presence of an anti-idiotype antibody against the recognition region or in the presence of cells expressing the target antigen of the chimeric antigen receptor on which it is expressed (3) Steps of collecting genetically modified lymphocytes after culturing
  • Step (1) is a step of preparing a cell genetically engineered to forcibly express the chimeric antigen receptor gene (CAR gene), and the target antigen-specific CAR gene is introduced into the target cell.
  • CAR gene chimeric antigen receptor gene
  • the CAR gene encodes a chimeric antigen receptor (CAR) that recognizes a specific target antigen.
  • CAR is a structure that contains a target-specific extracellular domain, a transmembrane domain, and an intracellular signal domain for the effector function of immune cells. Hereinafter, each domain will be described.
  • the extracellular domain contains an antigen recognition region and exhibits target-specific binding.
  • the extracellular domain contains a scFv fragment of an antitargeted monoclonal antibody.
  • the monoclonal antibody for example, a rodent (mouse, rat, rabbit, etc.) antibody, a human antibody, a humanized antibody, or the like is used.
  • a humanized monoclonal antibody is an antibody in which the structure of a monoclonal antibody of another animal species (for example, mouse or rat) is similar to that of a human antibody, and only the constant region of the antibody is replaced with that of a human antibody.
  • Human-type CDR-grafted antibody (PT Johons et al., Nature 321,522 (1986) in which the chimeric antibody and the portion other than the CDR (complementarity determining region) existing in the constant region and the variable region are replaced with those of the human antibody. ))including.
  • a method of selecting a human antibody framework (FR) that is highly homologous to a mouse antibody a method of producing a highly homologous humanized antibody, and a mouse CDR to a human antibody in order to enhance the antigen-binding activity of the human CDR transplanted antibody.
  • Further improvements have been made to the method of substituting amino acids in the FR region after transplantation (US Pat. No. 5585089, US Pat. No. 5697361, US Pat. No. 5693762, US Pat. No. 6180370, European Patent No. 451216 , European Patent No. 682040, Patent No. 2828340, etc.), which can also be used to produce humanized antibodies.
  • the scFv fragment is a structure in which the light chain variable region (VL) and heavy chain variable region (VH) of immunoglobulin are linked via a linker, and retains the ability to bind to an antigen.
  • a linker for example, a peptide linker can be used.
  • a peptide linker is a linker composed of peptides in which amino acids are linearly linked.
  • a typical example of a peptide linker is a linker composed of glycine and serine (GGS linker or GS linker).
  • the GGS linker and the amino acids that make up the GS linker, glycine and serine, are small in size and difficult to form higher-order structures in the linker.
  • the length of the linker is not particularly limited. For example, a linker having 5 to 25 amino acid residues can be used. The number of amino acid residues constituting the linker is preferably 8 to 25, more preferably 15 to 20.
  • the target is typically an antigen whose expression is specific to tumor cells.
  • the term "specific expression” as used herein means that significant or remarkable expression is observed as compared with cells other than tumor, and there is no intention of limiting the expression to those having no expression in cells other than tumor.
  • target antigens are CD19 antigen, CD20 antigen, GD2 antigen, CD22 antigen, CD30 antigen, CD33 antigen, CD44variant7 / 8 antigen, CD123 antigen, CEA antigen, Her2 / neu antigen, MUC1 antigen, MUC4 antigen, MUC6 antigen, IL. -13 Receptor-alpha2, immunoglobulin light chain, PSMA antigen, VEGF receptor2, mesothelin antigen, EGFRvIII, EphA2 antigen, IGFR and the like can be mentioned.
  • GM-CSF granulocyte-macricular colony stimulating factor
  • GM-CSF granulocyte-macricular colony stimulating factor
  • Leukemia stem cells, leukemia precursor cells, leukemia cells, etc. of myelogenous tumors are targeted by CAR cells, and myeloproliferative tumors, myelodysplastic / myeloproliferative tumors (CMML, JMML, CML, MDS / MPN-UC) Cells applicable to the prevention and treatment of myelodysplastic syndrome, acute myelogenous leukemia, etc. are prepared.
  • CMML myelodysplastic / myeloproliferative tumors
  • transmembrane domain intervenes between the extracellular domain and the intracellular signal domain.
  • a transmembrane domain such as CD28, CD3 ⁇ , CD8 ⁇ , CD3, CD4 or 4-1BB can be used.
  • a transmembrane domain consisting of an artificially constructed polypeptide may be used.
  • the intracellular signal domain transmits signals necessary for exerting the effector function of immune cells. That is, when the extracellular domain binds to the target antigen, an intracellular signal domain capable of transmitting a signal necessary for activation of immune cells is used.
  • the intracellular signal domain includes a domain for transmitting a signal via the TCR complex (referred to as "first domain” for convenience) and a domain for transmitting a co-stimulation signal (for convenience, "second domain”). Called) is included.
  • first domain an intracellular domain such as Fc ⁇ RI ⁇ can be used in addition to CD3 ⁇ .
  • CD3 ⁇ is used.
  • the intracellular domain of the co-stimulating molecule is used as the second domain. Examples of co-stimulatory molecules include CD28, 4-1BB (CD137), CD2, CD4, CD5, CD134, OX-40 or ICOS.
  • the intracellular domain of CD28 or 4-1BB is adopted.
  • the mode of connection between the first domain and the second domain is not particularly limited, but preferably, the transmembrane domain is preferably transmitted because it is known that the co-stimulation was strongly transmitted when the CD3 ⁇ was connected distally in the past cases. Place the second domain on the side.
  • a plurality of intracellular domains of the same or different species may be linked in a tandem manner to form a first domain. The same applies to the second domain.
  • the first domain and the second domain may be directly linked to each other, or a linker may be interposed between them.
  • a linker for example, a peptide linker can be used.
  • a peptide linker is a linker composed of peptides in which amino acids are linearly linked. The structure, characteristics, etc. of the peptide linker are as described above.
  • a linker composed of only glycine may be used.
  • the length of the linker is not particularly limited. For example, a linker having 2 to 15 amino acid residues can be used.
  • Leader sequences are used to facilitate the transport of CAR onto the cell membrane.
  • the leader sequence of the GM-CSF receptor can be used.
  • the spacer domain is used to promote the binding of CAR to the target antigen.
  • an Fc fragment of human IgG eg, human IgG1, human IgG4 can be used as a spacer domain.
  • a part of the extracellular domain of CD28, a part of the extracellular domain of CD8 ⁇ , and the like can also be used as the spacer domain.
  • a spacer domain can also be provided between the transmembrane domain and the intracellular signal domain.
  • the gene transfer method is roughly classified into a method using a viral vector and a method using a non-viral vector.
  • the former skillfully utilizes the phenomenon that the virus infects cells, and high gene transfer efficiency can be obtained.
  • viral vectors retroviral vectors, lentiviral vectors, adenoviral vectors, adeno-associated virus vectors, herpesvirus vectors, Sendai viral vectors and the like have been developed.
  • the target gene incorporated into the vector is integrated into the host chromosome, and stable and long-term expression can be expected.
  • Each viral vector can be prepared according to previously reported methods or using commercially available dedicated kits.
  • non-viral vectors include plasmid vectors, liposome vectors, and positively charged liposome vectors (Felgner, PL, Gadek, TR, Holm, M. et al., Proc. Natl. Acad. Sci., 84: 7413-7417. , 1987), YAC vector, BAC vector can be mentioned.
  • transposon method is one of the non-viral gene transfer methods.
  • Transposon is a general term for short gene sequences that cause gene translocations that have been conserved during evolution.
  • a pair of gene enzyme (transposase) and its specific recognition sequence causes gene translocation.
  • the transposon method for example, the piggyBac transposon method can be used.
  • the PiggyBac transposon method utilizes transposons isolated from insects (Fraser MJ et al., Insect Mol Biol. 1996 May; 5 (2): 141-51 .; Wilson MH et al., Mol Ther. .
  • the transposon method applicable to the present invention is not limited to the one using piggyBac, for example, Sleeping Beauty (Ivics Z, hackett PB, Plasterk RH, Izsvak Z (1997) Cell 91: 501-510.), Frog Prince (Miskey C, Izsvak Z, Plasterk RH, Ivics Z (2003) Nucleic Acids Res 31: 6873-6881.), Tol1 (Koga A, Inagaki H, Bessho Y, Hori H. 249 (4): 400-5 .; Koga A, Shimada A, Kuroki T, Hori H, Kusumi J, Kyono-Hamaguchi Y, Hamaguchi S. J Hum Genet.
  • the CAR gene transfer operation by the transposon method may be performed by a conventional method, and past literature (for example, for the piggyBac transposon method, Nakazawa Y, et al., J Immunother 32: 826-836, 2009, Nakazawa Y et al., J Immunother 6: 3-10, 2013, Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5; (69): e4235, Saito S, Nakazawa Y, Sueki A, et al. Anti-leukemic potential of piggyBac-mediated CD19-specific T cells against refractory Philadelphia chromasome-positive acute lymphoblastic leukemia. Cytotherapy. 2014; 16: 1257-69.) Is helpful.
  • the piggyBac transposon method is adopted.
  • a vector carrying a gene encoding the piggyBac transposase (transposase plasmid) and a desired nucleic acid construct (CAR expression cassette and / or siRNA expression cassette) are sandwiched between piggyBac reverse repeat sequences.
  • Vectors having a structure (transposon plasmid) are prepared, and these vectors are introduced (transfected) into target cells.
  • various methods such as electroporation, nucleofection, lipofection, and calcium phosphate method can be used.
  • a poly A addition signal sequence is placed downstream of the CAR gene. Transcription is terminated by the use of the poly A addition signal sequence.
  • a poly A addition signal sequence a poly A addition sequence of SV40, a poly A addition sequence of a bovine growth hormone gene, or the like can be used.
  • the transposon plasmid may include a detection gene (reporter gene, cell or tissue-specific gene, selectable marker gene, etc.), enhancer sequence, WRPE sequence, and the like.
  • the detection gene is used for determining the success or failure and efficiency of the introduction of the expression cassette, detecting the expression of the CAR gene or determining the expression efficiency, selecting and sorting the cells expressing the CAR gene, and the like.
  • the expression efficiency can be improved by using the enhancer sequence.
  • the genes for detection include the neo gene that imparts resistance to neomycin, the npt gene that imparts resistance to kanamycin, etc. (Herrera Estrella, EMBO J.
  • npt II gene (Messing & Vierra. Gene 1). 9: 259-268 (1982)), hph gene conferring resistance to neomycin (Blochinger & Digglmann, Mol Cell Bio 4: 2929-2931), dhfr gene conferring resistance to metatrexate (Bourouis et al. , EMBO J.2 (7)), etc. (marker gene), luciferase gene (Giacomin, P1. Sci. 116 (1996), 59-72; Scikantha, J. Bact.
  • GUS ⁇ -Glucronidase
  • genes for fluorescent proteins such as GFP (Gerdes, FEBS Lett. 389 (1996), 44-47) and their variants (EGFP, d2EGFP, etc.
  • GFP Gerdes, FEBS Lett. 389 (1996), 44-47) and their variants (EGFP, d2EGFP, etc.
  • intracellular domains Genes such as the missing epithelial growth factor receptor (EGFR) gene can be used.
  • the detection gene is linked to the CAR gene via, for example, a bicistronic control sequence (eg, ribosome internal recognition sequence (IRES)) or a sequence encoding a self-cleaving peptide.
  • a bicistronic control sequence eg, ribosome internal recognition sequence (IRES)
  • IRS ribosome internal recognition sequence
  • An example of a self-cleaving peptide is, but is not limited to, a 2A peptide (T2A) derived from Thosea signa virus.
  • E2A horse rhinitis A virus
  • P2A porcine teschovirus
  • Target cells include CD4-positive CD8-negative T cells, CD4-negative CD8-positive T cells, T cells prepared from iPS cells, ⁇ -T cells, ⁇ -T cells, NK cells, and NKT cells. Can be mentioned. Various cell populations can be used as long as they contain lymphocytes or progenitor cells as described above.
  • Peripheral blood mononuclear cells (PBMCs) collected from peripheral blood are one of the preferred target cells. That is, in a preferred embodiment, a gene transfer operation is performed on PBMCs, which are a cell population containing T cells or progenitor cells.
  • PBMCs may be prepared by a conventional method.
  • PBMCs For the method of preparing PBMCs, refer to, for example, Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5; (69): e4235. it can.
  • Step (2) The genetically modified lymphocytes (CAR cells) prepared in step (1) are subjected to culturing under specific conditions (step (2)).
  • the genetically modified lymphocyte prepared in step (1) is cultured in the presence of an anti-iditope antibody against the antigen recognition region of the chimeric antigen receptor on which it is expressed.
  • the genetically modified lymphocyte prepared in step (1) is cultured in the presence of cells expressing the target antigen of the chimeric antigen receptor on which it is expressed (target antigen expressing cells).
  • the target CAR cells (that is, those into which the target antigen-specific chimeric antigen receptor gene has been introduced) are specifically stimulated by culturing in the presence of anti-iditope antibody or target antigen-expressing cells. , To improve the proliferation rate.
  • the anti-iditope antibody used in the first aspect may be prepared by using an immunological method, a phage display method, a ribosome display method, or the like. Specifically, it can be produced according to a known method such as the method described in PLoS One. 2013; 8 (3): e57838. Further, an anti-iditope antibody may be prepared by using a contract service (for example, GenScript, Creative Biolabs, Immuno-Biological Laboratories, Inc., GeneFrontier Corp.). The anti-iditope antibody may be either a polyclonal antibody or a monoclonal antibody.
  • the anti-iditope antibody is an antibody that recognizes the antigen recognition region (variable domain) of an antibody molecule and binds to an epitope (idiotope) in a specific idiotype.
  • the anti-iditope antibody includes an anti-iditope antibody against CD19CAR, an anti-iditopes antibody against GD2CAR, an anti-iditopes antibody against CD22CAR, an anti-iditopes antibody against BCMACAR, and an anti-iditopes antibody against CD5CAR.
  • Examples thereof include an anti-iditope antibody against CD123CAR and an anti-iditopes antibody against HER2CAR.
  • Preferred examples thereof include an anti-iditope antibody against CD19CAR and an anti-iditopes antibody against GD2CAR.
  • the anti-iditope antibody may contain a constant region of immunoglobulin or may not contain a constant region of immunoglobulin.
  • the constant region of the heavy chain (CH1, CH2, and CH3) and the constant region of the light chain (CL) may be all included, and any one or more of these may be included. May include a combination of.
  • Specific examples of anti-iditope antibodies include immunoglobulin structure, Fab structure, F (ab') 2 structure, minibody structure, scFv-Fc structure, Fv structure, scFv structure, and diabody structure. , Triabody structure, tetrabody structure and the like.
  • anti-iditope antibodies also include low molecular weight antibodies such as Nanobodies and monobodies.
  • CAR cells and anti-iditope antibody By culturing in the presence of anti-iditope antibody, CAR cells and anti-iditope antibody can be brought into contact with each other.
  • an anti-iditopes antibody is immobilized on the culture surface of a culture vessel, the contact state can be formed during the culture.
  • the substrate used for solid phase formation include a substrate containing plastic such as polystyrene, glass, nitrocellulose and the like as a main component.
  • solid phase is meant that the anti-iditopes antibody is immobilized by binding directly or indirectly to the substrate. Fixation of the anti-iditopes antibody to the substrate can be performed according to or according to a conventional method.
  • fixation examples thereof include fixation via a covalent bond, fixation via a bond between avidin or streptavidin and biotin, and fixation by physical adsorption. Further, by adding an anti-iditope antibody (beaded antibody) bound to beads (for example, magnetic beads) to the culture medium, it is possible to form a state in which CAR cells and the anti-iditope antibody can be contacted.
  • the material of the beads is not particularly limited, and for example, metal particles such as gold, silver, copper, iron, aluminum, nickel, manganese, titanium, and oxides thereof; resin particles such as polystyrene and latex; silica particles and the like. Can be mentioned.
  • the shape of the bead is not particularly limited, and examples thereof include a sphere, a rectangular parallelepiped, a cube, a triangular pyramid, and the like, or a shape close to these.
  • the bead preferably has a substance on its surface to make the binding of the anti-iditope antibody easier and / or stronger. Examples of such substances include substances having a reactive group such as an epoxy group, an amino group, a carboxy group and an azide group; and substances having an affinity for other molecules such as avidin, protein A and protein B. Can be mentioned.
  • the target antigen-expressing cell used in the second aspect is not particularly limited as long as it is a cell in which the antigen (particularly the CAR recognition portion) is exposed on the cell surface.
  • it can be prepared by introducing a gene encoding a target antigen into peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • a specific cell population for example, T cells, B cells, NK cells, dendritic cells, or a combination of two or more of these
  • a gene encoding an antigen may be introduced.
  • a cell such as K562 can also be used as a cell into which a gene encoding a target antigen is introduced.
  • the expression of the target antigen may be transient or constant.
  • various vectors such as a plasmid may be used. More specifically, gene transfer can be carried out in the same manner as, for example, the above-mentioned CAR gene transfer.
  • the origin of the genetically modified lymphocyte (CAR cell) prepared in step (1) and the target antigen-expressing cell are the same.
  • CAR cells and target antigen-expressing cells are prepared using cells isolated from the same individual. In this way, it is possible to avoid the addition of unintended stimuli to CAR cells, the problem of immune rejection, infectious diseases, and the like.
  • the individual from which the CAR cells and the target antigen-expressing cells are derived is a patient (recipient) (autologous transplantation) or another person (allogeneic transplantation) who receives the CAR cells obtained by the preparation method of the present invention. is there.
  • the latter (ie, in the case of allogeneic transplantation) is applied to patients who have difficulty in preparing the cells necessary for the preparation of CAR cells, for example, when there are few lymphocytes in the blood or the activity of lymphocytes is low. It can be said that it is suitable for preparing.
  • Step (2) may be performed after 48 hours, more preferably 16 to 24 hours) have elapsed.
  • T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful.
  • a culture solution to which IL-7 is added in addition to IL-15 is used.
  • the amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml.
  • T cell growth factors such as IL-15 and IL-7 can be prepared according to a conventional method. In addition, a commercially available product can also be used. Although the use of T cell growth factors in non-human animal species is not excluded, T cell growth factors are usually derived from humans (may be recombinants). Growth factors such as human IL-15 and human IL-7 can be easily obtained (for example, provided by Miltenyi Biotec, R & D Systems, etc.).
  • a medium supplemented with serum (human serum, fetal bovine serum, etc.) may be used, but by adopting a serum-free medium, it is highly safe for clinical application and the culture efficiency due to the difference between serum lots is high. It is possible to prepare cells that have the advantage of being less likely to make a difference.
  • Specific examples of serum-free media for lymphocytes are TexMACS TM (Miltenyi Biotec) and AIM V® (Thermo Fisher Scientific).
  • a medium to which autologous serum (recipient's serum) is added may be used.
  • a medium suitable for lymphocyte culture may be used as the basal medium, and specific examples thereof are TexMACS TM and AIM V (registered trademark) described above.
  • Other culture conditions may be any one suitable for the survival and proliferation of lymphocytes, and general ones may be adopted.
  • the cells may be cultured in a CO 2 incubator (CO 2 concentration 5%) set at 37 ° C.
  • the culture period of step (2) is, for example, 1 to 14 days, preferably 1 to 7 days, and more preferably 2 to 7 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, the cell activity (life force) may decrease.
  • the transposon method is used to introduce the target antigen-specific chimeric antigen receptor gene in step (1), and the cells expressing the target antigen in step (2) encode the target antigen. It is prepared by introducing the gene into peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • the piggyBac transposon method is used to introduce the target antigen-specific chimeric antigen receptor gene in step (1), and the cells expressing the target antigen in step (2) use the target antigen. It is prepared by introducing the encoding gene into peripheral blood mononuclear cells (PBMCs).
  • Step (3) the genetically modified lymphocytes (CAR cells) after culturing are collected.
  • the collection operation may be performed by a conventional method. For example, it is collected by pipetting, centrifugation, or the like.
  • a step of culturing the cultured CAR cells in the presence of T cell growth factor is performed between steps (2) and (3). This step enables efficient expansion culture and also has the advantage of increasing cell viability.
  • IL-15, IL-7, etc. can be used as T cell growth factors.
  • the cells are cultured in a medium supplemented with IL-15 and IL-7 in the same manner as in step (2).
  • the culture period is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days. If the culture period is too short, a sufficient increase in the number of cells cannot be expected, and if the culture period is too long, there is a risk of a decrease in cell activity (life force), cell exhaustion / fatigue, and the like. It may be subcultured in the middle of culturing.
  • the medium is changed as necessary during the culture. For example, replace about 1/3 to 2/3 of the culture medium with a new medium once every 3 days.
  • step (1) and step (2) the following step (a) is performed.
  • Non-proliferative cells obtained by stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody and then performing a treatment for losing proliferative ability, and genetically modified lymphocytes prepared in step (1). Steps of mixing lymphocytes and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody
  • step (a) non-proliferative cells used for protection of CAR cells (CAR cells used in step (2)) after the transfer operation of the target antigen-specific chimeric antigen receptor gene are prepared, and in step (1). It is used for co-culture with the prepared CAR cells.
  • PBMCs peripheral blood mononuclear cells collected from peripheral blood are preferably used. It is also possible to use PBMCs purified to increase the T cell content, mononuclear cells collected from peripheral blood by feresis, or the like as the "cell population containing T cells" here.
  • T cells in the cell population can be stimulated with anti-CD3 antibody and anti-CD28 antibody.
  • Anti-CD3 antibody for example, the trade name CD3pure antibody provided by Miltenyi Biotec
  • anti-CD28 antibody for example, the trade name CD28pure antibody provided by Miltenyi Biotec can be used
  • step (a) it is also possible to perform the stimulation in step (a) using magnetic beads coated with anti-CD3 antibody and anti-CD28 antibody (for example, Dynabeads T-Activator CD3 / CD28 provided by VERITAS). It is preferable to use an "OKT3" clone as the anti-CD3 antibody.
  • anti-CD3 antibody and anti-CD28 antibody for example, Dynabeads T-Activator CD3 / CD28 provided by VERITAS. It is preferable to use an "OKT3" clone as the anti-CD3 antibody.
  • Cells stimulated with anti-CD3 antibody and anti-CD28 antibody are subjected to a treatment to lose proliferative ability, but before that, it is advisable to culture in the presence of T cell growth factor.
  • This culture enhances the activity of the cells after the stimulation treatment.
  • the culture period here is, for example, 1 to 10 days, preferably 2 to 7 days, and more preferably 3 to 4 days. If the culture period is too short, sufficient activation cannot be expected, and if the culture period is too long, co-stimulatory molecules may be attenuated.
  • the cultured cells may be cryopreserved once. In this case, the cells may be thawed at the time of use, stimulated again with anti-CD3 antibody and CD28 antibody (conditions conform to the above), and then subjected to "treatment to lose proliferative ability".
  • Activated T cells that have lost their proliferative ability can be obtained by undergoing the "treatment that causes them to lose their proliferative ability".
  • the treatment that causes the loss of proliferative capacity is typically irradiation, but agents may also be used.
  • An example of irradiation conditions is a treatment using gamma rays at an intensity of 25 Gy to 50 Gy for 15 to 30 minutes.
  • the non-proliferative cells prepared as described above and the CAR cells prepared in step (1) are mixed and co-cultured while stimulating with anti-CD3 antibody and anti-CD28 antibody.
  • stimulation by co-stimulatory molecules by non-proliferative cells and stimulation by anti-CD3 antibody and anti-CD28 antibody are added, CAR cells are activated, and their survival and proliferation are promoted.
  • This co-culture is preferably carried out immediately after the CAR gene transfer operation when preparing CAR cells. For example, co-culture is started immediately after the CAR gene transfer operation or within 1 day.
  • the co-culture (that is, step (a)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (a) (for example). , Immediately after the CAR gene transfer operation or within 1 day).
  • the ratio of the number of non-proliferative cells to the number of genetically modified lymphocytes (CAR cells) used for co-culture is not particularly limited, but is, for example, 0.025 to 0.5. And.
  • T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful.
  • IL-15 a culture solution to which IL-7 is added in addition to IL-15 is used.
  • the amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml.
  • the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml.
  • a medium supplemented with serum (human serum, fetal bovine serum, etc.) may be used, but by adopting a serum-free medium, it is highly safe for clinical application and the culture efficiency due to the difference between serum lots is high. It is possible to prepare cells that have the advantage of being less likely to make a difference.
  • Specific examples of serum-free media for lymphocytes are TexMACS TM (Miltenyi Biotec) and AIM V® (Thermo Fisher Scientific).
  • a medium to which autologous serum (recipient's serum) is added may be used.
  • a medium suitable for lymphocyte culture may be used as the basal medium, and specific examples thereof are TexMACS TM and AIM V (registered trademark) described above.
  • Other culture conditions may be any one suitable for the survival and proliferation of lymphocytes, and general ones may be adopted.
  • the cells may be cultured in a CO 2 incubator (CO 2 concentration 5%) set at 37 ° C.
  • the co-culture period is, for example, 1 to 10 days, preferably 1 to 7 days, and more preferably 2 to 4 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, the cell activity (life force) may decrease.
  • step (b) Protection of cells after CAR gene transfer operation by co-culture with non-proliferative PBMCs
  • step (b) is performed between steps (1) and (2).
  • step (B) Non-proliferative PBMCs obtained by stimulating PBMCs as they are or with anti-CD3 antibody and anti-CD28 antibody and then performing a treatment to lose proliferative ability, and genetically modified lymphocytes prepared in step (1). Steps to mix and co-culture
  • step (b) non-proliferative PBMCs used for protection of CAR cells (CAR cells used in step (2)) after the transfer operation of the target antigen-specific chimeric antigen receptor gene are prepared, and in step (1). It is used for co-culture with the prepared CAR cells.
  • the method for stimulating PBMCs with anti-CD3 antibody and anti-CD28 antibody is as described in 1-2 above. Is similar to.
  • the treatment for losing the proliferative ability, and prior to that, culturing in the presence of T cell growth factor and the method thereof are also described in 1-2. Is similar to.
  • "as is PBMCs" means that PBMCs are not stimulated by anti-CD3 antibody and anti-CD28 antibody.
  • the non-proliferative PBMCs prepared as described above are mixed with the genetically modified lymphocytes (CAR cells) prepared in step (1) and co-cultured.
  • Co-culture may be performed while stimulating with an anti-CD3 antibody and an anti-CD28 antibody.
  • This co-culture is preferably carried out immediately after the CAR gene transfer operation when preparing CAR cells. For example, co-culture is started immediately after the CAR gene transfer operation or within 1 day.
  • the co-culture (that is, step (b)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (b) (for example). , Immediately after the CAR gene transfer operation or within 1 day).
  • the ratio of the number of non-proliferative PBMCs used for co-culture to the number of genetically modified lymphocytes (CAR cells) is not particularly limited, but is, for example, 0.025 to 0.5. , Preferably 0.05 to 1.0.
  • T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful.
  • a culture solution to which IL-7 is added in addition to IL-15 is used.
  • the amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml.
  • the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml.
  • Conditions not mentioned are described in 1-2. Is the same as in the case of the above aspect.
  • Non-proliferative PBMCs may be added in the middle of co-culture (for example, 3 to 11 days after the start of co-culture, preferably 5 to 9 days, and more preferably 7 days).
  • the cells after co-culture may be collected, mixed with other non-proliferative PBMCs, and then cultured again. These operations may be repeated twice or more. In this way, if stimulation or activation using non-proliferative PBMCs is performed a plurality of times, it is expected that the induction rate of CAR cells will be improved and the number of CAR cells will be increased.
  • those prepared again, or those prepared by culturing a part of the cells prepared at the beginning or those stored in a cryopreservation can be used as the non-proliferative PBMCs here.
  • non-proliferative PBMCs are prepared using a part of PBMCs isolated from peripheral blood obtained by one blood sampling and CAR cells are prepared from the other part, the present invention can be carried out.
  • the number of blood samplings involved can be reduced, which is an extremely great advantage in clinical application.
  • non-proliferative PBMCs If it is used as additional non-proliferative PBMCs, then the three required cells, namely CAR cells, non-proliferative PBMCs used for co-culture with the cells, in the middle of co-culture Since non-proliferative PBMCs for addition can be prepared by a single blood collection, the burden on the patient in the treatment using the CAR cells obtained in the present invention is greatly reduced.
  • virus-specific chimeric antigen receptor gene-modified lymphocytes by co-culture with T cells carrying a virus peptide
  • virus-specific chimeric antigen receptor gene-modified lymphocytes (virus-specific). CAR cells) are prepared.
  • Virus-specific CAR cells can be expected to improve internal persistence by stimulation from viral T cell receptors when used for autologous transplantation, and further reduce allogeneic immune response (GVHD) when used for allogeneic transplantation. This has important advantages in clinical application, such as the possibility of producing CAR cells from a transplant donor and the possibility of formulating CAR cells from a third-party donor.
  • virus-specific CAR cells have been reported to persist in the body for longer periods of time (Pule MA, et al. Nat Med. 2008 Nov; 14 (11): 1264-70.).
  • CTL cytotoxic T cells
  • step (c) is performed to prepare virus-specific CAR cells. Matters not mentioned (for example, method for preparing a cell population containing T cells, basic operation for stimulation with anti-CD3 antibody and anti-CD28 antibody, method for processing to lose proliferative ability, basic operation for co-culture, etc. ) Is described in 1-2. Since it is the same as the aspect of the above, duplicate explanations are omitted and the corresponding explanations are used.
  • C A cell population containing T cells was stimulated with an anti-CD3 antibody and an anti-CD28 antibody, and then cultured in the presence of the viral peptide antigen and treated to lose the proliferative ability to retain the viral peptide antigen. The step of mixing non-proliferative cells with the genetically modified lymphocytes prepared in step (1) and co-culturing them.
  • step (c) first, a cell population containing T cells is stimulated with an anti-CD3 antibody and an anti-CD28 antibody to obtain activated T cells. Then, the cells are cultured in the presence of the viral peptide antigen and treated to lose their proliferative ability. As a result, non-proliferative "activated T cells holding a viral peptide antigen on the cell surface" (hereinafter referred to as "viral peptide-retaining non-proliferative cells”) are obtained.
  • the order of culturing in the presence of the viral peptide antigen and the treatment for losing the proliferative ability is not particularly limited.
  • the growth ability may be lost after culturing in the presence of the virus peptide antigen, or the culture may be performed in the presence of the virus peptide antigen after the loss of the growth ability.
  • the former order is adopted because of the expectation that the uptake of the viral peptide antigen will be better before the loss of proliferative capacity.
  • a medium to which the viral peptide antigen has been added may be used.
  • the viral peptide antigen may be added to the medium during culturing.
  • the concentration of the viral peptide antigen added is, for example, 0.5 ⁇ g / ml to 1 ⁇ g / ml.
  • the culture period is, for example, 10 minutes to 5 hours, preferably 20 minutes to 3 hours.
  • viral peptide antigen refers to an epitope peptide or a long peptide containing an epitope capable of inducing cytotoxic T cells (CTL) specific to a specific virus.
  • the viral peptide antigen is not limited to these, but is, for example, an adenovirus (AdV) antigen peptide (see, for example, WO2007015540 A1) and a cytomegalovirus (CMV) antigen peptide (for example, JP-A-2002).
  • AdV adenovirus
  • CMV cytomegalovirus
  • Epstein-Barr virus (EBV) antigen peptide for example, WO 2007049737 A1, Japanese Patent Application No. 2011-177487, Japanese Patent Application Laid-Open No. (See 2006-188513), etc.
  • the viral peptide antigen can be prepared by a conventional method (for example, liquid phase synthesis method, solid phase synthesis method) based on the sequence information.
  • some viral peptide antigens are commercially available (for example, provided by Medical & Biological Laboratories, Ltd., Takara Bio, Miltenyi Biotec, etc.).
  • antigen peptide mixture usually two or more types of antigen peptides (antigen peptide mixture) are used.
  • antigen peptide mixture usually two or more types of antigen peptides (antigen peptide mixture) are used.
  • AdV antigen peptide mixture CMV antigen peptide mixture or EBV antigen peptide mixture, or a combination of two or more of these antigen peptide mixture (for example, AdV antigen peptide mixture, CMV antigen peptide mixture and EBV antigen peptide mixture).
  • AdV antigen peptide mixture for example, AdV antigen peptide mixture, CMV antigen peptide mixture and EBV antigen peptide mixture.
  • a plurality of activated T cells having different targets can be obtained, and the virus-specific CAR cells obtained by the preparation method of the present invention are effective therapeutic targets ( We can expect an increase in the number of patients (improvement of coverage).
  • the use of the virus-specific CAR cells obtained by the preparation method of the present invention specifically, the disease to be treated and the pathological condition of the patient should be considered. Good.
  • the antigen-peptide mixture of EBV virus may be used alone or in combination with an antigen-peptide mixture of other viruses.
  • AdV antigen-peptide mixture, CMV antigen-peptide mixture, and EBV antigen-peptide mixture are also commercially available (for example, PepTivator (registered trademark) AdV5 Hexon, PepTivator (registered trademark) CMV pp65, PepTivator (for example, provided by Milteny Biotech).
  • EBV EBNA-1 Registered trademarks
  • PepTivator registered trademark
  • EBV BZLF1 PepMix TM Collection HCMV provided by JPT Peptide Technologies
  • PepMix TM EBV (EBNA1), etc.) can be easily obtained.
  • the viral peptide-retaining non-proliferative cells prepared as described above and the genetically modified lymphocytes (CAR cells) prepared in step (1) are mixed and co-cultured.
  • stimulation via a co-stimulating molecule and a virus antigen peptide by a virus peptide-carrying non-proliferative cell is applied, and virus antigen-specific genetically modified lymphocytes are activated and their survival and proliferation are promoted.
  • this co-culture is used for CAR genes in preparing CAR cells. It is preferable to carry out immediately after the introduction operation.
  • co-culture is started immediately after the CAR gene transfer operation or within 1 day.
  • the co-culture (that is, step (c)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (c) (for example).
  • the start time of step (2) is the same as the start time of step (c) (for example).
  • the ratio of the number of viral peptide-carrying non-proliferative cells used for co-culture to the number of genetically modified lymphocytes (CAR cells) is not particularly limited. For example, 0.025 to 0.5, preferably 0.05 to 1.0.
  • this step is to stimulate with anti-CD3 antibody and anti-CD28 antibody for reasons such as selectively proliferating virus-specific CAR cells, avoiding strong stimulation and preventing T cell exhaustion / fatigue. Do not add.
  • a culture medium to which a T cell growth factor has been added during co-culture examples include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful.
  • IL-15 a culture solution to which IL-7 is added in addition to IL-15 is used.
  • the amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml.
  • the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml.
  • Conditions not mentioned are described in 1-2. Is the same as in the case of the above aspect.
  • Viral peptide-carrying non-proliferative cells may be added during co-culture. Alternatively, the cells after co-culture may be collected, mixed with another virus peptide-carrying non-proliferative cell, and then co-cultured again. These operations may be repeated twice or more. As described above, if the activation without stimulation using the virus peptide-retaining non-proliferative cells is performed a plurality of times, the induction rate of the virus-specific CAR cells can be improved and the number of virus-specific CAR cells can be expected to increase. In addition, a cell prepared again or a cell in which a part of the cell prepared at the beginning is preserved can be used as "another viral peptide-carrying non-proliferative cell" here.
  • the co-culture period is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, there is a risk of a decrease in cell activity (life force), cell exhaustion / fatigue, and the like.
  • the genetically modified lymphocytes (CAR cells) prepared in step (1) Prior to co-culturing with viral peptide-carrying non-proliferative cells, the genetically modified lymphocytes (CAR cells) prepared in step (1) are co-cultured with viral peptide-carrying non-proliferative peripheral blood mononuclear cells (PBMCs). It may be.
  • the period of co-culture here is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days.
  • the cells obtained by co-culturing the genetically modified lymphocytes (CAR cells) prepared in step (1) and the viral peptide-carrying non-proliferative PBMCs (co-culture in the first step) are obtained by the above method.
  • the viral peptide-retaining non-proliferative PBMCs here can be prepared by subjecting the PBMCs to a culture in the presence of a viral peptide antigen and a treatment for losing the ability to proliferate. Specifically, for example, PBMCs isolated from peripheral blood are radiation-treated and then cultured in the presence of a viral peptide antigen to obtain viral peptide-retaining non-proliferative PBMCs.
  • the present invention if it is decided to prepare viral peptide-retaining non-proliferative PBMCs using a part of PBMCs isolated from peripheral blood obtained by one blood sampling and to prepare CAR cells from the other part, the present invention. It is possible to reduce the number of blood samplings associated with the implementation of the above, which is an extremely great advantage in clinical application.
  • the remaining PBMCs are used to prepare viral peptide-retaining non-proliferative cells (cells used for the second stage co-culture), or the viral peptide-retaining non-proliferative PBMCs prepared using a part of PBMCs.
  • the necessary three types Cells that is, CAR cells, viral peptide-retaining non-proliferative PBMCs used for co-culture with the cells (first-stage co-culture), and viral peptide-retaining non-proliferative cells used for second-stage co-culture. Since it can be prepared by collecting blood once, the burden on the patient in the treatment using the CAR cells obtained in the present invention is greatly reduced.
  • CAR cells Gene-modified lymphocytes expressing chimeric antigen receptor and their uses Further aspects of the present invention are the genetically modified lymphocytes expressing chimeric antigen receptor obtained by the preparation method of the present invention (hereinafter, “the present invention”. "CAR cells") and their uses.
  • the CAR cells of the present invention can be used for the treatment, prevention or amelioration of various diseases (hereinafter referred to as "target diseases") for which CAR therapy is considered to be effective.
  • target diseases various diseases
  • the representative of the target disease is cancer, but it is not limited to this.
  • target diseases include various B-cell lymphomas (follicle malignant lymphoma, diffuse malignant lymphoma, mantle cell lymphoma, MALT lymphoma, intravascular B-cell lymphoma, CD20-positive hodgkin lymphoma, etc.), myeloproliferative neoplasm, bone marrow.
  • B-cell lymphomas follicle malignant lymphoma, diffuse malignant lymphoma, mantle cell lymphoma, MALT lymphoma, intravascular B-cell lymphoma, CD20-positive hodgkin lymphoma, etc.
  • myeloproliferative neoplasm myeloproliferative neoplasm
  • bone marrow myeloproliferative neoplasm
  • CMML Hypoplastic / myeloproliferative neoplasms
  • JMML Hypoplastic / myeloproliferative neoplasms
  • CML Hypoplastic / myeloproliferative neoplasms
  • MDS Hypoplastic / myeloproliferative neoplasms
  • myelodysplastic syndrome acute myeloproliferative leukemia, neuroblastoma, brain tumor, Ewing sarcoma, osteosarcoma, retinoblastoma, small lung Celloma, melanoma, ovarian cancer, horizontal print myeloma, kidney cancer, pancreatic cancer, malignant mesoderma, prostate cancer, etc.
  • Treatment includes alleviating (mitigating) the symptoms characteristic of the target disease or associated symptoms, preventing or delaying the exacerbation of the symptoms, and the like.
  • Prevention means preventing or delaying the onset / delay of a disease (disorder) or its symptoms, or reducing the risk of onset / onset.
  • improvement means that the disease (disorder) or its symptoms are alleviated (mild), improved, ameliorated, or cured (including partial cure).
  • the CAR cells of the present invention can also be provided in the form of a cell preparation.
  • the cell preparation of the present invention contains a therapeutically effective amount of the CAR cells of the present invention. For example, it contains 10 4 to 10 10 cells for a single dose.
  • Various components such as dimethylsulfoxide (DMSO) and serum albumin for the purpose of protecting cells, antibiotics for the purpose of preventing bacterial contamination, and various components (vitamins) for the purpose of activating, proliferating or inducing differentiation of cells.
  • DMSO dimethylsulfoxide
  • serum albumin for the purpose of protecting cells
  • antibiotics for the purpose of preventing bacterial contamination
  • various components for the purpose of activating, proliferating or inducing differentiation of cells.
  • Cytokines, growth factors, steroids, etc. may be contained in the cell preparation.
  • the administration route of the CAR cell or cell preparation of the present invention is not particularly limited. For example, it is administered by intravenous injection, intraarterial injection, intraportal injection, intradermal injection, subcutaneous injection, intramuscular injection, or intraperitoneal injection. Topical administration may be used instead of systemic administration.
  • Topical administration may be used instead of systemic administration.
  • local administration direct injection into a target tissue / organ / organ can be exemplified.
  • the administration schedule may be prepared in consideration of the gender, age, body weight, pathological condition, etc. of the subject (patient). In addition to a single dose, multiple doses may be administered continuously or periodically.
  • PBMCs mononuclear cells
  • PBMCs mononuclear cells
  • 5 ⁇ g of a plasmid vector expressing the CAR target antigen is introduced by electroporation (nucleofection), and culture is started in an incubator.
  • Day 1 After irradiation, it is used as a feeder cell.
  • Day 1 Stimulate with anti-CD3 antibody / anti-CD28 antibody coated plate.
  • Day 4 Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 7 Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 10 Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 14 The culture is finished.
  • Day 1 Stimulate with an anti-iditope antibody coated plate.
  • Day 4 Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 7 Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 10 Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 14 The culture is finished.
  • PBMCs target antigen-expressing cells
  • Day 0 Mononuclear cells were isolated from peripheral blood, and 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) were added to 1x10 7 mononuclear cells. Then, the gene is introduced by electroporation (nucleofection). Then, the cells are floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and culture is started in a 37 ° C.
  • Day 1 Stimulate with target antigen gene-introduced PBMCs (feeder cells) (eg, culture in a culture dish inoculated with feeder cells).
  • Day 4 Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 7 Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 10 Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 14 The culture is finished.
  • the gene transfer efficiency was 8% for culture method 1 (A-1) and 18.1% for culture method 2 (A-2) (Fig. 6A), and culture method 2 was significantly higher than that of conventional culture method 1. Improvement of gene transfer efficiency was observed. That is, it was demonstrated that the novel culture method is effective in improving the efficiency of gene transfer.
  • the proliferation rate of CAR-T cells from day 7 was 20-fold with anti-iditopes antibody stimulation (A-2) and 13-fold with conventional OKT3 / anti-CD28 antibody stimulation (A-1). (Fig. 6B).
  • the new culture method overcomes the weaknesses of the transposon method (gene transfer efficiency and cell viability / cell proliferation rate are lower than those of the viral vector method), and will expand the clinical application of CAR therapy using the transposon method. I can say.
  • the novel culture method is highly versatile and can be applied to the preparation of CAR cells by the viral vector method or the like, and its utility value is great.
  • Day 1 Stimulate with anti-iditope antibody coated plate (B-2, culture method 2) or OKT3 / anti-CD28 antibody coated plate (B-1, culture method 1 (conventional method)).
  • Day 4 Transfer cells to a 24-well plate and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 7 Culture finished
  • the gene transfer efficiency was 20.5% for anti-iditope antibody stimulation (B-2, culture method 2) and 3.58% for OKT3 / anti-CD28 antibody stimulation (B-1, culture method 1 (conventional method)) (Fig. 8A). ), Compared with the conventional method of OKT3 / anti-CD28 antibody stimulation, anti-iditopes antibody stimulation showed a significant improvement in gene transfer efficiency.
  • the number of CAR-T cells was 550,000 for anti-iditopes antibody stimulation (B-2) and 180,000 for OKT3 / anti-CD28 antibody stimulation (B-1) (Fig. 8B). / Compared with anti-CD28 antibody stimulation, anti-iditope antibody stimulation showed a significant improvement in CAR-T cell number.
  • Method (1) Preparation of activated T cells (1-1) Preparation of anti-CD3 antibody / anti-CD28 antibody coated (sensitized) plate
  • Anti-CD3 antibody and anti-CD28 antibody were diluted with PBS to 1 mg / ml and diluted with PBS. Add to a 24-well uncoated plate to 0.5 ml / well. The plate is allowed to stand in an incubator at 37 ° C for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.
  • PBMCs Mononuclear cells
  • PBMCs Mononuclear cells
  • 5 ⁇ g of a plasmid vector expressing the CAR target antigen is introduced by electroporation (nucleofection), and culture is started in an incubator.
  • Day 1 After irradiation, it is used as a feeder cell.
  • Day 1 Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
  • Day 4 Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 7 Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
  • Day 10 Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 14 The culture is finished.
  • the number of CAR-T cells is increased by cell stimulation (protective action of transgenic cells damaged during electroporation) by expression of co-stimulating molecules, cytokine stimulation by culture microenvironment, and the like. It can be expected that the increase and the efficiency of gene transfer will increase further.
  • the transgenic cells and irradiated viral peptide-added activated T cells are mixed, floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and cultured in a 24-well plate in a 37 ° C incubator.
  • Day 1 Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
  • Day 2-5 If necessary, replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 7 Cells are collected and counted.
  • the number of CAR-T cells is further increased by cell stimulation by the expression of co-stimulating molecules, cytokine stimulation by the culture microenvironment, and relatively gentle cell stimulation from virus-specific T cell receptors. And further increase in gene transfer efficiency can be expected.
  • a decrease in allogeneic reactivity effect that can be expected for virus-specific CTL
  • the possibility of using CAR-T cells derived from a third party due to this and the stimulation of virus antigen receptors in the body by a virus in the body. Persistency may increase, and further improvement in safety and increase in therapeutic effect are expected.
  • ⁇ Culture method 6 (improvement of culture method 5)> 1.
  • Method Day 0 Isolate mononuclear cells (PBMCs) from peripheral blood. After irradiating a part ( 6 PBMC 1x10), add viral peptides (PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50 ng each) and incubate at 37 ° C for 30 minutes.
  • viral peptides PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50 ng each
  • Activated T cells are prepared from the remaining PBMCs according to the method (1).
  • Day 1 Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
  • Day 2-5 If necessary, replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 7 Cells are collected and counted.
  • virus peptides PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50 ng each
  • Day 10 Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
  • Day 14 The culture is finished.
  • the above culture method 6 makes it possible to obtain CAT-T cells by collecting blood once, and has an advantage that the burden on the patient is reduced. Further, the culture method 6 can be expected to further improve the gene transfer efficiency.
  • the gene transfer efficiency or cell proliferation rate when preparing CAR cells is improved. That is, the present invention makes it possible to efficiently prepare a larger number of CAR cells, and can contribute to improving the therapeutic results of CAR therapy, expanding the scope of application of CAR therapy, and the like.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Provided is a novel production (culture) means that expands a CAR cell population. Specifically provided is a method for producing genetically modified lymphocytes that express a chimeric antigen receptor, by (1) a step for preparing gene-modified lymphocytes into which a gene of a target antigen-specific chimeric antigen receptor has been introduced; (2) a step for culturing the gene-modified lymphocytes prepared in step (1) in the presence of anti-idiotype antibody against the antigen recognition region of the chimeric antigen receptor expressed by the gene-modified lymphocytes, or in the presence of cells that express the target antigen for the chimeric antigen receptor expressed by the genetically modified lymphocytes; and (3) a step for recovering cultured gene-modified lymphocytes.

Description

キメラ抗原受容体遺伝子改変リンパ球の調製方法Method for preparing chimeric antigen receptor genetically modified lymphocytes

 本発明はキメラ抗原受容体を発現する遺伝子改変リンパ球(CAR遺伝子導入リンパ球)に関する。詳細には、CAR遺伝子導入リンパ球の作製方法、当該細胞の用途等に関する。 The present invention relates to genetically modified lymphocytes expressing chimeric antigen receptors (CAR transgenic lymphocytes). Specifically, the present invention relates to a method for producing CAR gene-introduced lymphocytes, uses of the cells, and the like.

 キメラ抗原受容体(Chimeric Antigen Receptor。以下、「CAR」とも呼ぶ)を用いた遺伝子改変T細胞療法(CAR-T療法)や遺伝子改変NK細胞療法(CAR-NK療法)が臨床応用されてきている。CARは、典型的には、抗体の単鎖可変領域を細胞外ドメインとし、それに膜貫通領域、CD3ζ及び共刺激シグナルを伝える分子の細胞内ドメインをつないだ構造を備える。抗体の特異性に従って抗原に結合することによりCAR遺伝子導入リンパ球は活性化し、標的細胞(がん細胞など)を傷害する。CAR療法は、細胞の調製が比較的容易であること、高い細胞傷害活性を示すこと、持続的な効果を期待できることなどの利点を有し、特に、難治性や従来の治療法に抵抗性の症例に対する新たな治療手段として期待されている。実際、化学療法抵抗性急性リンパ性白血病(acute lymphoblastic leukemia)患者に対して、細胞表面に発現するCD19抗原に対するCARを、患者から採取した末梢血T細胞に遺伝子導入し、培養して輸注する臨床試験が欧米で行われ、寛解率80~90%の良好な成績が報告されている(非特許文献1~3)。CAR療法は、米国では難治性がんに対して最も将来有望な治療法の1つとして注目されている。 Genetically modified T cell therapy (CAR-T therapy) and genetically modified NK cell therapy (CAR-NK therapy) using a chimeric antigen receptor (hereinafter also referred to as "CAR") have been clinically applied. .. CAR typically comprises a structure in which the single-chain variable region of an antibody is the extracellular domain, which is linked to the transmembrane domain, the CD3ζ, and the intracellular domain of the molecule that transmits the co-stimulation signal. CAR transgenic lymphocytes are activated by binding to the antigen according to the specificity of the antibody, damaging target cells (such as cancer cells). CAR therapy has advantages such as relatively easy cell preparation, high cytotoxic activity, and long-lasting effect, and is particularly resistant to refractory and conventional treatments. It is expected as a new therapeutic means for cases. In fact, in patients with chemotherapy-resistant acute lymphoblastic leukemia, CAR for the CD19 antigen expressed on the cell surface is gene-introduced into peripheral blood T cells collected from the patients, cultured, and infused. The test has been conducted in Europe and the United States, and good results with a remission rate of 80 to 90% have been reported (Non-Patent Documents 1 to 3). CAR therapy is attracting attention as one of the most promising treatments for refractory cancer in the United States.

 従来、CAR療法に用いる細胞(代表例はCAR-T細胞)はウイルスベクターを用いて調製されてきた。しかしながら、一般に使用されるレトロウイルスは、がん原遺伝子への挿入変異の頻度が高く(造血幹細胞を用いた遺伝子治療では白血病が多発している)、安全性が問題となる。また、ウイルスベクターを使用する際には専用細胞培養設備が必要となるため、治療費が非常に高額となるという、経済性の問題もある(非特許文献4)。 Conventionally, cells used for CAR therapy (typically CAR-T cells) have been prepared using a viral vector. However, commonly used retroviruses have a high frequency of insertion mutations into protooncogenes (leukemia occurs frequently in gene therapy using hematopoietic stem cells), and safety becomes a problem. In addition, since a dedicated cell culture facility is required when using a viral vector, there is also an economical problem that the treatment cost is very high (Non-Patent Document 4).

 ウイルスベクターを利用した従来のCAR療法における問題点を解決するために、非ウイルスベクターを用いた遺伝子改変技術の一つであるトランスポゾン法の利用が検討されている。トランスポゾン法は、ウイルスベクター法と同様に永続的な遺伝子導入を可能にするものの、ウイルスベクター法と比較して遺伝子導入効率が低く、また、遺伝子導入操作(エレクトロポレーション及びその改良法など)に伴い細胞が傷害を受け、細胞生存率や細胞増殖率が低下するといった問題を抱える。この問題に対する解決策の一つとして、本発明者らの研究グループは、「CAR遺伝子導入後のリンパ球を、別途用意した活性化T細胞と共培養することによって保護し、遺伝子導入効率及び細胞生存率/細胞増殖率の向上を図るという戦略」、及び「CAR遺伝子導入後のリンパ球を、ウイルスペプチドを保持させた活性化T細胞あるいは単核球と共培養することによって、ウイルス特異的CAR-T細胞を効率的に調製する戦略」を提案した(特許文献1)。 In order to solve the problems in conventional CAR therapy using viral vectors, the use of the transposon method, which is one of the gene modification techniques using non-viral vectors, is being studied. Although the transposon method enables permanent gene transfer like the viral vector method, the gene transfer efficiency is lower than that of the viral vector method, and it is suitable for gene transfer operations (electroporation and its improvement methods, etc.). As a result, the cells are damaged, and there is a problem that the cell viability and the cell proliferation rate decrease. As one of the solutions to this problem, the research group of the present inventors said, "Protecting lymphocytes after CAR gene transfer by co-culturing with activated T cells prepared separately, gene transfer efficiency and cells. Strategy to improve survival / cell proliferation rate ”and“ Virus-specific CAR by co-culturing lymphocytes after CAR gene transfer with activated T cells or mononuclear cells carrying viral peptides -A strategy for efficiently preparing T cells "was proposed (Patent Document 1).

国際公開第2017/061615号パンフレットInternational Publication No. 2017/061615 Pamphlet

Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, Milone MC, Levine BL, June CH. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med, 368(16):1509-18. 2013Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, Milone MC, Levine BL, June CH. Chimeric antigen receptor-modified . N Engl J Med, 368 (16): 1509-18. 2013 Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, Teachey DT, Levine BL, June CH, Porter DL, Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med, 371(16):1507-17. 2014Maude SL, Frey N, Shaw PA, Applenc R, Barrett DM, Bunin NJ, Chew A, Gonzalez VE, Zheng Z, Lacey SF, Mahnke YD, Melenhorst JJ, Rheingold SR, Shen A, T , Porter DL, Grupp SA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med, 371 (16): 1507-17. 2014 Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, Steinberg SM, Stroncek D, Tschernia N, Yuan C, Zhang H, Zhang L, Rosenberg SA, Wayne AS, Mackall CL. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2014Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, Fry TJ, Orentas R, Sabatino M, Shah NN, Steinberg SM, Strong Zhang Rosenberg SA, Wayne AS, Mackall CL. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Morgan RA. Faster, cheaper, safer, T-cell engineering. J Immunother, 36(1):1-2. 2013Morgan RA. Faster, cheaper, safer, T-cell engineering. J Immunother, 36 (1): 1-2. 2013

 トランスポゾン法を利用したCAR療法は有望であるものの、遺伝子導入効率や細胞生存率/細胞増殖率の問題から、治療に必要な細胞数を用意できない場合が想定される。ウイルスベクター法でCAR細胞を調製する場合においても、患者(ドナー)によっては血液中の標的細胞(CAR遺伝子が導入される細胞)が少ないことや細胞の活性/生存率が低いがために十分な数のCAR細胞が得られないことがある。即ち、上記の問題はトランスポゾン法に限ったものではない。また、CD19等、白血球の細胞表面抗原が標的であれば、CAR遺伝子導入後の細胞を培養する際、共存する細胞からの刺激によってCAR細胞の生存率/増殖率の向上を望めるが、抗原によってはこのような効果を期待できない。
 そこで本発明の主たる課題は、CAR細胞数の増大をもたらす新規な調製(培養)手段を提供し、CAR療法の臨床応用の進展及び治療成績の向上に資することにある。
Although CAR therapy using the transposon method is promising, it is assumed that the number of cells required for treatment cannot be prepared due to problems of gene transfer efficiency and cell viability / cell proliferation rate. Even when CAR cells are prepared by the viral vector method, it is sufficient for some patients (donors) because there are few target cells (cells into which the CAR gene is introduced) in the blood and the cell activity / survival rate is low. The number of CAR cells may not be available. That is, the above problem is not limited to the transposon method. In addition, if the cell surface antigen of leukocytes such as CD19 is the target, it is expected that the survival rate / proliferation rate of CAR cells will be improved by stimulation from coexisting cells when culturing cells after CAR gene transfer. Cannot expect such an effect.
Therefore, the main object of the present invention is to provide a novel preparation (culture) means for increasing the number of CAR cells, and to contribute to the progress of clinical application of CAR therapy and the improvement of therapeutic results.

 上記課題を解決すべく検討を重ねた結果、本発明者らは二つの戦略、即ち、CAR遺伝子導入後の細胞(CAR細胞)を抗イディオタイプ抗体で刺激する方法(第1の戦略)、及びCAR遺伝子導入後の細胞(CAR細胞)を、標的抗原を発現する細胞で刺激する方法(第2の戦略)を創出した。第1の戦略は抗イディオタイプ抗体によって直接CAR細胞を刺激するものであり、CAR細胞特異的な増殖が期待できる。実際、従来法(抗CD3抗体/抗CD28抗体による刺激)に比べ、大幅なCAR細胞数の増加(遺伝子導入効率/細胞増殖率の改善)が認められた(後述の実施例を参照)。他方、第2の戦略によれば、CAR細胞の培養中に標的抗原による刺激が加わり、第1の戦略と同様、CAR細胞特異的な増殖を期待できる。このように、新規な二つの戦略は、治療に必要な細胞数の確保を可能或いは容易にするものであり、その臨床上の意義は極めて大きい。以下に示す発明は当該二つの戦略に基づく。 [1]以下のステップ(1)~(3)を含む、キメラ抗原受容体を発現する遺伝子改変リンパ球の調製方法:
 (1)標的抗原特異的キメラ抗原受容体遺伝子が導入された遺伝子改変リンパ球を用意するステップ;
 (2)ステップ(1)で用意した遺伝子改変リンパ球を、それが発現するキメラ抗原受容体の抗原認識領域に対する抗イディオタイプ抗体の存在下、又はそれが発現するキメラ抗原受容体の標的抗原を発現する細胞の存在下で培養するステップ;及び
 (3)培養後の遺伝子改変リンパ球を回収するステップ。
 [2]ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入にトランスポゾン法が用いられる、[1]に記載の調製方法。
 [3]トランスポゾン法がpiggyBacトランスポゾン法である、[2]に記載の調製方法。
 [4]ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入操作から8時間~48時間経過した後にステップ(2)を行う、[1]~[3]のいずれか一項に記載の調製方法。
 [5]ステップ(2)の培養期間が1日~14日である、[1]~[4]のいずれか一項に記載の調製方法。
 [6]ステップ(2)の培養の際、抗CD3抗体及び抗CD28抗体による刺激を加えない、[1]~[5]のいずれか一項に記載の調製方法。
 [7]抗イディオタイプ抗体が固相化抗体又はビーズ化抗体である、[1]~[6]のいずれか一項に記載の調製方法。
 [8]T細胞又はその前駆細胞を含む細胞集団に対する、標的抗原特異的キメラ抗原受容体遺伝子の導入によって、ステップ(1)の遺伝子改変リンパ球が得られる、[1]~[7]のいずれか一項に記載の調製方法。
 [9]細胞集団が末梢血単核細胞(PBMCs)である、[8]に記載の調製方法。
 [10]ステップ(2)における、標的抗原を発現する細胞が、該標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製される、[1]~[9]のいずれか一項に記載の調製方法。
 [10-1]ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入にトランスポゾン法が用いられ、
 ステップ(2)における、標的抗原を発現する細胞が、該標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製される、[1]~[10]のいずれか一項に記載の調製方法。
 [11]ステップ(2)における、標的抗原を発現する細胞と、遺伝子改変リンパ球が同一の個体に由来する、[1]~[10]のいずれか一項に記載の調製方法。
 [12]個体が、ステップ(3)で回収した遺伝子改変リンパ球の移植を受ける個体と別の個体である、[11]に記載の調製方法。
 [13]ステップ(2)とステップ(3)の間に、培養後の細胞をT細胞増殖因子の存在下で培養するステップを行う、[1]~[12]のいずれか一項に記載の調製方法。
 [14]T細胞増殖因子がIL-15である、[13]に記載の調製方法。
 [15]T細胞増殖因子としてIL-15とIL-7を併用する、[13]に記載の調製方法。
 [16]ステップ(1)とステップ(2)の間に、以下のステップ(a)を行う、[1]~[15]のいずれか一項に記載の調製方法:
 (a)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、抗CD3抗体及び抗CD28抗体で刺激しつつ共培養するステップ。
 [17]ステップ(1)とステップ(2)の間に、以下のステップ(b)を行う、[1]~[15]のいずれか一項に記載の調製方法:
 (b)末梢血単核細胞(PBMCs)をそのまま、又は抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる、非増殖性PBMCsと、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ。
 [18]ステップ(1)とステップ(2)の間に、以下のステップ(c)を行う、[1]~[15]のいずれか一項に記載の調製方法:
 (c)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、ウイルスペプチド抗原存在下での培養及び増殖能を喪失させる処理を行うことによって得られる、ウイルスペプチド抗原を保持した非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ。
 [19]T細胞を含む細胞集団が末梢血単核細胞(PBMCs)である、[16]又は[18]に記載の調製方法。
 [20]増殖能を喪失させる処理が放射線照射である、[16]~[19]のいずれか一項に記載の調製方法。
 [21]非増殖性細胞と、遺伝子改変リンパ球が同一の個体に由来する、[16]~[20]のいずれか一項に記載の調製方法。
 [22][1]~[21]のいずれか一項に記載の調製方法で得られた、キメラ抗原受容体を発現する遺伝子改変リンパ球。
 [23][22]に記載の遺伝子改変リンパ球を治療上有効量含む、細胞製剤。
 [24][22]に記載の遺伝子改変リンパ球を、治療上有効量、がん患者に投与するステップを含む、がんの治療法。
As a result of repeated studies to solve the above problems, the present inventors have two strategies, that is, a method of stimulating cells after CAR gene transfer (CAR cells) with an anti-iditope antibody (first strategy), and We have created a method (second strategy) for stimulating cells after CAR gene transfer (CAR cells) with cells expressing the target antigen. The first strategy is to directly stimulate CAR cells with anti-iditope antibodies, and CAR cell-specific proliferation can be expected. In fact, a significant increase in the number of CAR cells (improvement in gene transfer efficiency / cell proliferation rate) was observed as compared with the conventional method (stimulation with anti-CD3 antibody / anti-CD28 antibody) (see Examples described later). On the other hand, according to the second strategy, stimulation by the target antigen is added during the culture of CAR cells, and CAR cell-specific proliferation can be expected as in the first strategy. As described above, the two new strategies make it possible or facilitate to secure the number of cells required for treatment, and their clinical significance is extremely large. The inventions shown below are based on these two strategies. [1] A method for preparing a genetically modified lymphocyte expressing a chimeric antigen receptor, which comprises the following steps (1) to (3):
(1) A step of preparing a genetically modified lymphocyte into which a target antigen-specific chimeric antigen receptor gene has been introduced;
(2) The genetically modified lymphocyte prepared in step (1) is used in the presence of an anti-iditope antibody against the antigen recognition region of the chimeric antigen receptor on which it is expressed, or the target antigen of the chimeric antigen receptor on which it is expressed. The step of culturing in the presence of expressing cells; and (3) the step of collecting the genetically modified lymphocytes after culturing.
[2] The preparation method according to [1], wherein the transposon method is used for the introduction of the target antigen-specific chimeric antigen receptor gene in step (1).
[3] The preparation method according to [2], wherein the transposon method is the piggyBac transposon method.
[4] In any one of [1] to [3], step (2) is performed after 8 to 48 hours have passed from the operation of introducing the target antigen-specific chimeric antigen receptor gene in step (1). The preparation method described.
[5] The preparation method according to any one of [1] to [4], wherein the culture period of step (2) is 1 to 14 days.
[6] The preparation method according to any one of [1] to [5], wherein the culture in step (2) is not stimulated by the anti-CD3 antibody and the anti-CD28 antibody.
[7] The preparation method according to any one of [1] to [6], wherein the anti-iditope antibody is a solid phase antibody or a beaded antibody.
[8] Any of [1] to [7], wherein the genetically modified lymphocyte of step (1) can be obtained by introducing a target antigen-specific chimeric antigen receptor gene into a cell population containing T cells or progenitor cells thereof. The preparation method according to item 1.
[9] The preparation method according to [8], wherein the cell population is peripheral blood mononuclear cells (PBMCs).
[10] The cells expressing the target antigen in step (2) are prepared by introducing the gene encoding the target antigen into peripheral blood mononuclear cells (PBMCs), according to [1] to [9]. The preparation method according to any one item.
[10-1] The transposon method is used for the introduction of the target antigen-specific chimeric antigen receptor gene in step (1).
Any one of [1] to [10], wherein the cell expressing the target antigen in step (2) is prepared by introducing a gene encoding the target antigen into peripheral blood mononuclear cells (PBMCs). The preparation method described in the section.
[11] The preparation method according to any one of [1] to [10], wherein the cell expressing the target antigen and the genetically modified lymphocyte are derived from the same individual in step (2).
[12] The preparation method according to [11], wherein the individual is an individual different from the individual to be transplanted with the genetically modified lymphocyte recovered in step (3).
[13] The item according to any one of [1] to [12], wherein the step of culturing the cultured cells in the presence of T cell growth factor is performed between steps (2) and (3). Preparation method.
[14] The preparation method according to [13], wherein the T cell growth factor is IL-15.
[15] The preparation method according to [13], wherein IL-15 and IL-7 are used in combination as T cell growth factors.
[16] The preparation method according to any one of [1] to [15], wherein the following step (a) is performed between steps (1) and (2):
(A) Non-proliferative cells obtained by stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody and then performing a treatment for losing proliferative ability, and genetically modified lymphocytes prepared in step (1). A step of mixing lymphocytes and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody.
[17] The preparation method according to any one of [1] to [15], wherein the following step (b) is performed between steps (1) and (2):
(B) Non-proliferative PBMCs obtained by treating peripheral blood mononuclear cells (PBMCs) as they are or by stimulating them with anti-CD3 antibody and anti-CD28 antibody and then performing a treatment to lose proliferative ability, and step (1). The step of mixing and co-culturing the genetically modified lymphocytes prepared in.
[18] The preparation method according to any one of [1] to [15], wherein the following step (c) is performed between steps (1) and (2):
(C) A cell population containing T cells was stimulated with an anti-CD3 antibody and an anti-CD28 antibody, and then cultured in the presence of the viral peptide antigen and treated to lose the proliferative ability to retain the viral peptide antigen. A step of mixing non-proliferative cells with the transgenic lymphocytes prepared in step (1) and co-culturing them.
[19] The preparation method according to [16] or [18], wherein the cell population containing T cells is peripheral blood mononuclear cells (PBMCs).
[20] The preparation method according to any one of [16] to [19], wherein the treatment for losing the proliferative ability is irradiation.
[21] The preparation method according to any one of [16] to [20], wherein the non-proliferative cells and the genetically modified lymphocytes are derived from the same individual.
[22] A genetically modified lymphocyte expressing a chimeric antigen receptor obtained by the preparation method according to any one of [1] to [21].
[23] A cell preparation containing a therapeutically effective amount of the genetically modified lymphocyte according to [22].
[24] A method for treating cancer, which comprises a step of administering a therapeutically effective amount of the genetically modified lymphocyte according to [22] to a cancer patient.

従来法(培養法1)によるCAR-T細胞の調製。Preparation of CAR-T cells by the conventional method (culture method 1). 新規方法(培養法2)によるCAR-T細胞の調製。Preparation of CAR-T cells by a novel method (culture method 2). 新規方法(培養法3)によるCAR-T細胞の調製。Preparation of CAR-T cells by a novel method (culture method 3). pIRII-CAR.CD19.28zベクター(配列番号1)の構成。CD19CAR遺伝子が5'逆向き反復配列(5'IR)と3'逆向き反復配列(3'IR)に挟まれた構造を備える。CD19CARは、リーダー配列(配列番号2)、軽鎖可変領域(VL)(配列番号3)、重鎖可変領域(VH)(配列番号4)、Fc領域(CH2、CH3)(配列番号5)、CD28の膜貫通領域及び細胞内ドメイン(配列番号6)及びCD3ζ(配列番号7)を含む。Composition of pIRII-CAR.CD19.28z vector (SEQ ID NO: 1). The CD19CAR gene has a structure sandwiched between a 5'inverted repeat sequence (5'IR) and a 3'inverted repeat sequence (3'IR). CD19CAR has a leader sequence (SEQ ID NO: 2), light chain variable region (VL) (SEQ ID NO: 3), heavy chain variable region (VH) (SEQ ID NO: 4), Fc region (CH2, CH3) (SEQ ID NO: 5), Includes the transmembrane region of CD28 and the intracellular domain (SEQ ID NO: 6) and CD3ζ (SEQ ID NO: 7). pCMV-pigBacベクター(配列番号8)の構成。CMV最初期プロモーター(CMV immediate ear1y promoter)の制御下にpiggyBacトランスポザーゼ遺伝子が配置されている。Composition of pCMV-pigBac vector (SEQ ID NO: 8). The piggyBac transposase gene is located under the control of the CMV earliest promoter (CMV immediate ear1y promoter). 新規方法(培養法2)による遺伝子導入効率の改善。14日目の細胞をフローサイトメトリーで評価し、最初に7日間従来法(培養法1)で培養した後に、新規方法(培養法2)で7日間培養したもの(右上)と、最初に7日間従来法(培養法1)で培養した後に、さらに従来法(培養法1)で7日間培養したもの(右下)との間で遺伝子導入効率を比較した。Improvement of gene transfer efficiency by a new method (culture method 2). The cells on the 14th day were evaluated by flow cytometry, first cultured for 7 days by the conventional method (culture method 1), then cultured for 7 days by the new method (culture method 2) (upper right), and first 7 After culturing by the conventional method (culture method 1) for 1 day, the gene transfer efficiency was further compared with that cultivated by the conventional method (culture method 1) for 7 days (lower right). 新規方法(培養法2)による遺伝子導入効率の改善。最初に7日間従来法(培養法1)で培養した後に、新規方法(培養法2)で7日間培養したもの(左)と、最初に7日間従来法(培養法1)で培養した後に、さらに従来法(培養法1)で7日間培養したもの(右)との間で、CAR-T細胞の7日目からの増殖率を比較した。Improvement of gene transfer efficiency by a new method (culture method 2). First cultivated by the conventional method (culture method 1) for 7 days, then cultivated by the new method (culture method 2) for 7 days (left), and first cultivated by the conventional method (culture method 1) for 7 days, and then. Furthermore, the proliferation rates of CAR-T cells from the 7th day were compared with those cultured for 7 days by the conventional method (culture method 1) (right). 最適化したCAR遺伝子導入用ベクターであるpIRII-CAR.CD19_optimizedベクター(配列番号9)の構成。pIRII-CAR.CD19.28zベクターの構造と比較して、Fc領域(CH2、CH3)が削除されている。Composition of pIRII-CAR.CD19_optimized vector (SEQ ID NO: 9), which is an optimized CAR gene transfer vector. Compared with the structure of the pIRII-CAR.CD19.28z vector, the Fc region (CH2, CH3) has been deleted. 新規方法(培養法2)による遺伝子導入効率の改善。7日目の細胞をフローサイトメトリーで評価し、新規方法(培養法2)(右上)と従来法(培養法1)(右下)の間で遺伝子導入効率を比較した。Improvement of gene transfer efficiency by a new method (culture method 2). The cells on day 7 were evaluated by flow cytometry, and the gene transfer efficiency was compared between the novel method (culture method 2) (upper right) and the conventional method (culture method 1) (lower right). 新規方法(培養法2)による遺伝子導入効率の改善。新規方法(培養法2)(左)と従来法(培養法1)(右)の間で、CAR-T細胞数を比較した。Improvement of gene transfer efficiency by a new method (culture method 2). The number of CAR-T cells was compared between the new method (culture method 2) (left) and the conventional method (culture method 1) (right).

1.キメラ抗原受容体を発現する遺伝子改変リンパ球の調製方法
1-1.抗イディオタイプ抗体/標的抗原を発現する細胞による特異的刺激
 本発明はキメラ抗原受容体を発現する遺伝子改変リンパ球(即ち、CAR遺伝子導入リンパ球。以下、説明の便宜上「CAR細胞」と呼ぶことがある)の調製方法に関する。本発明の調製方法で得られるCAR細胞(典型的にはCAR-T細胞)はCAR療法に利用することができる。本発明の調製方法では、以下のステップ(1)~(3)を行う。尚、特に言及しない限り、本明細書における各種細胞(例えばT細胞)はヒト細胞である。
 (1)標的抗原特異的キメラ抗原受容体遺伝子が導入された遺伝子改変リンパ球を用意するステップ
 (2)ステップ(1)で用意した遺伝子改変リンパ球を、それが発現するキメラ抗原受容体の抗原認識領域に対する抗イディオタイプ抗体の存在下、又はそれが発現するキメラ抗原受容体の標的抗原を発現する細胞の存在下で培養するステップ
 (3)培養後の遺伝子改変リンパ球を回収するステップ
1. 1. Method for preparing genetically modified lymphocytes expressing chimeric antigen receptor 1-1. Specific Stimulation by Cells Expressing Anti-Idiotopal Antibodies / Target Antigens The present invention refers to genetically modified lymphocytes expressing chimeric antigen receptors (ie, CAR transgenic lymphocytes; hereinafter, referred to as "CAR cells" for convenience of explanation. There is) regarding the preparation method. CAR cells (typically CAR-T cells) obtained by the preparation method of the present invention can be used for CAR therapy. In the preparation method of the present invention, the following steps (1) to (3) are performed. Unless otherwise specified, various cells (for example, T cells) in the present specification are human cells.
(1) Step of preparing a genetically modified lymphocyte into which a target antigen-specific chimeric antigen receptor gene has been introduced (2) The gene-modified lymphocyte prepared in step (1) is expressed by the chimeric antigen receptor antigen. Steps of culturing in the presence of an anti-idiotype antibody against the recognition region or in the presence of cells expressing the target antigen of the chimeric antigen receptor on which it is expressed (3) Steps of collecting genetically modified lymphocytes after culturing

ステップ(1)
 ステップ(1)は、キメラ抗原受容体遺伝子(CAR遺伝子)を強制発現するように遺伝子操作された細胞を用意するステップであり、標的抗原特異的CAR遺伝子が標的細胞に導入される。
Step (1)
The step (1) is a step of preparing a cell genetically engineered to forcibly express the chimeric antigen receptor gene (CAR gene), and the target antigen-specific CAR gene is introduced into the target cell.

 CAR遺伝子は、特定の標的抗原を認識するキメラ抗原受容体(CAR)をコードする。CARは、標的に特異的な細胞外ドメインと、膜貫通ドメイン、及び免疫細胞のエフェクター機能のための細胞内シグナルドメインを含む構造体である。以下、各ドメインについて説明する。 The CAR gene encodes a chimeric antigen receptor (CAR) that recognizes a specific target antigen. CAR is a structure that contains a target-specific extracellular domain, a transmembrane domain, and an intracellular signal domain for the effector function of immune cells. Hereinafter, each domain will be described.

(a)細胞外ドメイン
 細胞外ドメインは抗原認識領域を含み、標的に特異的な結合性を示す。例えば、細胞外ドメインは、抗標的モノクローナル抗体のscFv断片を含む。ここでのモノクローナル抗体として、例えば、齧歯類(マウス、ラット、ウサギなど)の抗体、ヒト抗体、ヒト化抗体等が用いられる。ヒト化モノクローナル抗体は、他の動物種(例えばマウスやラット)のモノクローナル抗体の構造をヒトの抗体の構造に類似させた抗体であり、抗体の定常領域のみをヒト抗体のものに置換したヒト型キメラ抗体、及び定常領域及び可変領域に存在するCDR(相補性決定領域)以外の部分をヒト抗体のものに置換したヒト型CDR移植(CDR-grafted)抗体(P.T.Johons et al., Nature 321,522(1986))を含む。ヒト型CDR移植抗体の抗原結合活性を高めるため、マウス抗体と相同性の高いヒト抗体フレームワーク(FR)を選択する方法、相同性の高いヒト型化抗体を作製する方法、ヒト抗体にマウスCDRを移植した後さらにFR領域のアミノ酸を置換する方法の改良技術もすでに開発され(米国特許第5585089号、米国特許第5693761号、米国特許第5693762号、米国特許第6180370号、欧州特許第451216号、欧州特許第682040号、特許第2828340号などを参照)、ヒト化抗体の作製に利用することもできる。
(A) Extracellular domain The extracellular domain contains an antigen recognition region and exhibits target-specific binding. For example, the extracellular domain contains a scFv fragment of an antitargeted monoclonal antibody. As the monoclonal antibody here, for example, a rodent (mouse, rat, rabbit, etc.) antibody, a human antibody, a humanized antibody, or the like is used. A humanized monoclonal antibody is an antibody in which the structure of a monoclonal antibody of another animal species (for example, mouse or rat) is similar to that of a human antibody, and only the constant region of the antibody is replaced with that of a human antibody. Human-type CDR-grafted antibody (PT Johons et al., Nature 321,522 (1986) in which the chimeric antibody and the portion other than the CDR (complementarity determining region) existing in the constant region and the variable region are replaced with those of the human antibody. ))including. A method of selecting a human antibody framework (FR) that is highly homologous to a mouse antibody, a method of producing a highly homologous humanized antibody, and a mouse CDR to a human antibody in order to enhance the antigen-binding activity of the human CDR transplanted antibody. Further improvements have been made to the method of substituting amino acids in the FR region after transplantation (US Pat. No. 5585089, US Pat. No. 5697361, US Pat. No. 5693762, US Pat. No. 6180370, European Patent No. 451216 , European Patent No. 682040, Patent No. 2828340, etc.), which can also be used to produce humanized antibodies.

 scFv断片とは、免疫グロブリンの軽鎖可変領域(VL)と重鎖可変領域(VH)がリンカーを介して連結された構造体であり、抗原との結合能を保持している。リンカーとしては、例えばペプチドリンカーを用いることができる。ペプチドリンカーとは、直鎖状にアミノ酸が連結したペプチドからなるリンカーである。ペプチドリンカーの代表例は、グリシンとセリンから構成されるリンカー(GGSリンカーやGSリンカー)である。GGSリンカー及びGSリンカーを構成するアミノ酸であるグリシンとセリンは、それ自体のサイズが小さく、リンカー内で高次構造が形成されにくい。リンカーの長さは特に限定されない。例えば、アミノ酸残基数が5~25個のリンカーを用いることができる。リンカーを構成するアミノ酸残基数は好ましくは8~25個、更に好ましくは15~20個である。 The scFv fragment is a structure in which the light chain variable region (VL) and heavy chain variable region (VH) of immunoglobulin are linked via a linker, and retains the ability to bind to an antigen. As the linker, for example, a peptide linker can be used. A peptide linker is a linker composed of peptides in which amino acids are linearly linked. A typical example of a peptide linker is a linker composed of glycine and serine (GGS linker or GS linker). The GGS linker and the amino acids that make up the GS linker, glycine and serine, are small in size and difficult to form higher-order structures in the linker. The length of the linker is not particularly limited. For example, a linker having 5 to 25 amino acid residues can be used. The number of amino acid residues constituting the linker is preferably 8 to 25, more preferably 15 to 20.

 標的には、典型的には、腫瘍細胞に特異的な発現が認められる抗原が用いられる。ここでの「特異的な発現」とは、腫瘍以外の細胞に比較して有意ないし顕著な発現が認められることをいい、腫瘍以外の細胞において全く発現がないものに限定する意図はない。標的抗原の例として、CD19抗原、CD20抗原、GD2抗原、CD22抗原、CD30抗原、CD33抗原、CD44variant7/8抗原、CD123抗原、CEA抗原、Her2/neu抗原、MUC1抗原、MUC4抗原、MUC6抗原、IL-13 receptor-alpha2、イムノグロブリン軽鎖、PSMA抗原、VEGF receptor2、mesothelin抗原、EGFRvIII、EphA2抗原、IGFRなどを挙げることができる。 The target is typically an antigen whose expression is specific to tumor cells. The term "specific expression" as used herein means that significant or remarkable expression is observed as compared with cells other than tumor, and there is no intention of limiting the expression to those having no expression in cells other than tumor. Examples of target antigens are CD19 antigen, CD20 antigen, GD2 antigen, CD22 antigen, CD30 antigen, CD33 antigen, CD44variant7 / 8 antigen, CD123 antigen, CEA antigen, Her2 / neu antigen, MUC1 antigen, MUC4 antigen, MUC6 antigen, IL. -13 Receptor-alpha2, immunoglobulin light chain, PSMA antigen, VEGF receptor2, mesothelin antigen, EGFRvIII, EphA2 antigen, IGFR and the like can be mentioned.

 白血病性幹細胞/前駆細胞に発現しているGM-CSF(顆粒球単球コロニー刺激因子)受容体を標的にすることもできる。この場合、CARを構成する細胞外ドメインには、GM-CSF受容体のリガンドであるGM-CSFが用いられる。そして、骨髄系腫瘍の白血病幹細胞、白血病前駆細胞、白血病細胞等がCAR細胞の標的となり、骨髄増殖性腫瘍、骨髄異形成/骨髄増殖性腫瘍(CMML、JMML、CML、MDS/MPN-UC)、骨髄異形成症候群、急性骨髄性白血病等の予防・治療へ適用可能な細胞が調製される。 It is also possible to target the GM-CSF (granulocyte-macricular colony stimulating factor) receptor expressed in leukemic stem cells / progenitor cells. In this case, GM-CSF, which is a ligand for the GM-CSF receptor, is used as the extracellular domain constituting CAR. Leukemia stem cells, leukemia precursor cells, leukemia cells, etc. of myelogenous tumors are targeted by CAR cells, and myeloproliferative tumors, myelodysplastic / myeloproliferative tumors (CMML, JMML, CML, MDS / MPN-UC) Cells applicable to the prevention and treatment of myelodysplastic syndrome, acute myelogenous leukemia, etc. are prepared.

(b)膜貫通ドメイン
 膜貫通ドメインは、細胞外ドメインと細胞内シグナルドメインの間に介在する。膜貫通ドメインとしては、CD28、CD3ε、CD8α、CD3、CD4又は4-1BBなどの膜貫通ドメインを用いることができる。人工的に構築したポリペプチドからなる膜貫通ドメインを用いることにしてもよい。
(B) Transmembrane domain The transmembrane domain intervenes between the extracellular domain and the intracellular signal domain. As the transmembrane domain, a transmembrane domain such as CD28, CD3ε, CD8α, CD3, CD4 or 4-1BB can be used. A transmembrane domain consisting of an artificially constructed polypeptide may be used.

(c)細胞内シグナルドメイン
 細胞内シグナルドメインは、免疫細胞のエフェクター機能の発揮に必要なシグナルを伝達する。即ち、細胞外ドメインが標的の抗原と結合した際、免疫細胞の活性化に必要なシグナルを伝達することが可能な細胞内シグナルドメインが用いられる。細胞内シグナルドメインには、TCR複合体を介したシグナルを伝達するためのドメイン(便宜上、「第1ドメイン」と呼ぶ)と、共刺激シグナルを伝達するためのドメイン(便宜上、「第2ドメイン」と呼ぶ)が含まれる。第1ドメインとして、CD3ζの他、FcεRIγ等の細胞内ドメインを用いることができる。好ましくは、CD3ζが用いられる。また、第2ドメインとしては共刺激分子の細胞内ドメインが用いられる。共刺激分子としてCD28、4-1BB(CD137)、CD2、CD4、CD5、CD134、OX-40又はICOSを例示することができる。好ましくは、CD28又は4-1BBの細胞内ドメインを採用する。
(C) Intracellular signal domain The intracellular signal domain transmits signals necessary for exerting the effector function of immune cells. That is, when the extracellular domain binds to the target antigen, an intracellular signal domain capable of transmitting a signal necessary for activation of immune cells is used. The intracellular signal domain includes a domain for transmitting a signal via the TCR complex (referred to as "first domain" for convenience) and a domain for transmitting a co-stimulation signal (for convenience, "second domain"). Called) is included. As the first domain, an intracellular domain such as FcεRIγ can be used in addition to CD3ζ. Preferably, CD3ζ is used. The intracellular domain of the co-stimulating molecule is used as the second domain. Examples of co-stimulatory molecules include CD28, 4-1BB (CD137), CD2, CD4, CD5, CD134, OX-40 or ICOS. Preferably, the intracellular domain of CD28 or 4-1BB is adopted.

 第1ドメインと第2ドメインの連結態様は特に限定されないが、好ましくは、過去の事例においてCD3ζを遠位につないだ場合に共刺激が強く伝わったことが知られていることから、膜貫通ドメイン側に第2ドメインを配置する。同一又は異種の複数の細胞内ドメインをタンデム状に連結して第1ドメインを構成してもよい。第2ドメインについても同様である。 The mode of connection between the first domain and the second domain is not particularly limited, but preferably, the transmembrane domain is preferably transmitted because it is known that the co-stimulation was strongly transmitted when the CD3ζ was connected distally in the past cases. Place the second domain on the side. A plurality of intracellular domains of the same or different species may be linked in a tandem manner to form a first domain. The same applies to the second domain.

 第1ドメインと第2ドメインは、これらを直接連結しても、これらの間にリンカーを介在させてもよい。リンカーとしては例えばペプチドリンカーを用いることができる。ペプチドリンカーとは、直鎖状にアミノ酸が連結したペプチドからなるリンカーである。ペプチドリンカーの構造、特徴等は前述の通りである。但し、ここでのリンカーとしては、グリシンのみから構成されるものを用いてもよい。リンカーの長さは特に限定されない。例えば、アミノ酸残基数が2~15個のリンカーを用いることができる。 The first domain and the second domain may be directly linked to each other, or a linker may be interposed between them. As the linker, for example, a peptide linker can be used. A peptide linker is a linker composed of peptides in which amino acids are linearly linked. The structure, characteristics, etc. of the peptide linker are as described above. However, as the linker here, a linker composed of only glycine may be used. The length of the linker is not particularly limited. For example, a linker having 2 to 15 amino acid residues can be used.

(d)その他の要素
 CARの細胞膜上への輸送を促すために、リーダー配列(シグナルペプチド)が用いられる。例えば、GM-CSFレセプターのリーダー配列を用いることができる。また、細胞外ドメインと膜貫通ドメインがスペーサードメインを介して連結した構造にするとよい。即ち、好ましい態様のCARは、細胞外ドメインと膜貫通ドメインの間にスペーサードメインを含む。スペーサードメインは、CARと標的抗原との結合を促進させるために用いられる。例えば、ヒトIgG(例えばヒトIgG1、ヒトIgG4)のFc断片をスペーサードメインとして用いることがきる。その他、CD28の細胞外ドメインの一部やCD8αの細胞外ドメインの一部等をスペーサードメインとして用いることもできる。尚、膜貫通ドメインと細胞内シグナルドメインの間にもスペーサードメインを設けることもできる。
(D) Other elements Leader sequences (signal peptides) are used to facilitate the transport of CAR onto the cell membrane. For example, the leader sequence of the GM-CSF receptor can be used. In addition, it is preferable to have a structure in which the extracellular domain and the transmembrane domain are linked via a spacer domain. That is, the CAR of the preferred embodiment comprises a spacer domain between the extracellular domain and the transmembrane domain. The spacer domain is used to promote the binding of CAR to the target antigen. For example, an Fc fragment of human IgG (eg, human IgG1, human IgG4) can be used as a spacer domain. In addition, a part of the extracellular domain of CD28, a part of the extracellular domain of CD8α, and the like can also be used as the spacer domain. A spacer domain can also be provided between the transmembrane domain and the intracellular signal domain.

 尚、これまでにCARを利用した実験、臨床研究などの報告がいくつかあり(例えばRossig C, et al. Mol Ther 10:5-18, 2004; Dotti G, et al. Hum Gene Ther 20:1229-1239, 2009; Ngo MC, et al. Hum Mol Genet 20 (R1):R93-99, 2011; Ahmed N, et al. Mol Ther 17:1779-1787, 2009; Pule MA, et al. Nat Med 14:1264-1270, 2008; Louis CU, et al. Blood 118:6050-6056, 2011; Kochenderfer JN, et al. Blood 116:4099-4102, 2010; Kochenderfer JN, et al. Blood 119 :2709-2720, 2012; Porter DL, et al. N Engl J Med 365:725-733, 2011; Kalos M, et al. Sci Transl Med 3:95ra73,2011; Brentjens RJ, et al. Blood 118:4817-4828, 2011; Brentjens RJ, et al. Sci Transl Med 5:177 ra38, 2013)、これらの報告を参考にして本発明におけるCARを構築することができる。 There have been some reports on experiments and clinical studies using CAR (for example, Rossig C, et al. Mol Ther 10: 5-18, 2004; Dotti G, et al. Hum Gene Ther 20: 1229. -1239, 2009; Ngo MC, et al. Hum Mol Genet 20 (R1): R93-99, 2011; Ahmed N, et al. Mol Ther 17: 1779-1787, 2009; Pule MA, et al. Nat Med 14 : 1264-1270, 2008; Louis CU, et al. Blood 118: 6050-6056, 2011; Kochenderfer JN, et al. Blood 116: 4099-4102, 2010; Kochenderfer JN, et al. Blood 119: 2709-2720, 2012; Porter DL, et al. N Engl J Med 365: 725-733, 2011; Kalos M, et al. Sci Transl Med 3: 95ra73, 2011; Brentjens RJ, et al. Blood 118: 4817-4828, 2011; Brentjens RJ, et al. Sci Transl Med 5: 177 ra38, 2013), the CAR in the present invention can be constructed with reference to these reports.

 CAR遺伝子の導入には、各種遺伝子導入法を利用することができる。遺伝子導入法はウイルスベクターを利用した方法と非ウイルスベクターを利用した方法に大別される。前者はウイルスが細胞へと感染する現象を巧みに利用するものであり、高い遺伝子導入効率が得られる。ウイルスベクターとしてレトロウイルスベクター、レンチウイルスベクター、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、センダイウイルスベクター等が開発されている。この中でレトロウイルスベクター、レンチウイルスベクター、アデノ随伴ウイルスベクターではベクターに組み込んだ目的遺伝子が宿主染色体へと組み込まれ、安定かつ長期的な発現が期待できる。各ウイルスベクターは既報の方法に従い又は市販される専用のキットを用いて作製することができる。非ウイルスベクターの例としては、プラスミドベクター、リポソームベクター、正電荷型リポソームベクター(Felgner, P.L., Gadek, T.R., Holm, M. et al., Proc. Natl. Acad. Sci., 84:7413-7417, 1987)、YACベクター、BACベクターを挙げることができる。 Various gene transfer methods can be used to introduce the CAR gene. The gene transfer method is roughly classified into a method using a viral vector and a method using a non-viral vector. The former skillfully utilizes the phenomenon that the virus infects cells, and high gene transfer efficiency can be obtained. As viral vectors, retroviral vectors, lentiviral vectors, adenoviral vectors, adeno-associated virus vectors, herpesvirus vectors, Sendai viral vectors and the like have been developed. Among these, in the retroviral vector, lentiviral vector, and adeno-associated virus vector, the target gene incorporated into the vector is integrated into the host chromosome, and stable and long-term expression can be expected. Each viral vector can be prepared according to previously reported methods or using commercially available dedicated kits. Examples of non-viral vectors include plasmid vectors, liposome vectors, and positively charged liposome vectors (Felgner, PL, Gadek, TR, Holm, M. et al., Proc. Natl. Acad. Sci., 84: 7413-7417. , 1987), YAC vector, BAC vector can be mentioned.

 好ましくは、トランスポゾン法による遺伝子導入を行う。トランスポゾン法とは、非ウイルス遺伝子導入法の一つである。トランスポゾンは、進化の過程で保存されてきた、遺伝子転位を引き起こす短い遺伝子配列の総称である。遺伝子酵素(トランスポザーゼ)とその特異認識配列のペアで遺伝子転位を引き起こす。トランスポゾン法として、例えば、piggyBacトランスポゾン法を用いることができる。PiggyBacトランスポゾン法は、昆虫から単離されたトランスポゾンを利用するものであり(Fraser MJ et al., Insect Mol Biol. 1996 May;5(2):141-51.; Wilson MH et al., Mol Ther. 2007 Jan;15(1):139-45.)、哺乳類染色体への高効率な組込みを可能にする。PiggyBacトランスポゾン法は実際にCAR遺伝子の導入に利用されている(例えばNakazawa Y, et al., J Immunother 32:826-836, 2009;Nakazawa Y et al., J Immunother 6:3-10, 2013等を参照)。本発明に適用可能なトランスポゾン法はpiggyBacを利用したものに限定されるものではなく、例えば、Sleeping Beauty(Ivics Z, Hackett PB, Plasterk RH, Izsvak Z (1997) Cell 91: 501-510.)、Frog Prince(Miskey C, Izsvak Z, Plasterk RH, Ivics Z (2003) Nucleic Acids Res 31: 6873-6881.)、Tol1(Koga A, Inagaki H, Bessho Y, Hori H. Mol Gen Genet. 1995 Dec 10;249(4):400-5.;Koga A, Shimada A, Kuroki T, Hori H, Kusumi J, Kyono-Hamaguchi Y, Hamaguchi S. J Hum Genet. 2007;52(7):628-35. Epub 2007 Jun 7.)、Tol2(Koga A, Hori H, Sakaizumi M (2002) Mar Biotechnol  4: 6-11.;Johnson Hamlet MR, Yergeau DA, Kuliyev E, Takeda M, Taira M, Kawakami K, Mead PE (2006) Genesis 44: 438-445.;Choo BG, Kondrichin I, Parinov S, Emelyanov A, Go W, Toh WC, Korzh V (2006) BMC Dev Biol 6: 5.)等のトランスポゾンを利用した方法を採用することにしてもよい。 Preferably, gene transfer is performed by the transposon method. The transposon method is one of the non-viral gene transfer methods. Transposon is a general term for short gene sequences that cause gene translocations that have been conserved during evolution. A pair of gene enzyme (transposase) and its specific recognition sequence causes gene translocation. As the transposon method, for example, the piggyBac transposon method can be used. The PiggyBac transposon method utilizes transposons isolated from insects (Fraser MJ et al., Insect Mol Biol. 1996 May; 5 (2): 141-51 .; Wilson MH et al., Mol Ther. . 2007 Jan; 15 (1): 139-45.), Enables highly efficient integration into mammalian chromosomes. The PiggyBac transposon method is actually used for the introduction of CAR genes (for example, Nakazawa Y, et al., J Immunother 32: 826-836, 2009; Nakazawa Y et al., J Immunother 6: 3-10, 2013, etc. See). The transposon method applicable to the present invention is not limited to the one using piggyBac, for example, Sleeping Beauty (Ivics Z, Hackett PB, Plasterk RH, Izsvak Z (1997) Cell 91: 501-510.), Frog Prince (Miskey C, Izsvak Z, Plasterk RH, Ivics Z (2003) Nucleic Acids Res 31: 6873-6881.), Tol1 (Koga A, Inagaki H, Bessho Y, Hori H. 249 (4): 400-5 .; Koga A, Shimada A, Kuroki T, Hori H, Kusumi J, Kyono-Hamaguchi Y, Hamaguchi S. J Hum Genet. 2007; 52 (7): 628-35. Epub 2007 Jun 7.), Tol2 (Koga A, Hori H, Sakaizumi M (2002) Mar Biotechnol 4: 6-11 .; Johnson Hamlet MR, Yergeau DA, Kulyev E, Takeda M, Taira M, Kawakami K, Mead PE ) Genesis 44: 438-445 .; Choo BG, Kondrichin I, Parinov S, Emelyanov A, Go W, Toh WC, Korzh V (2006) BMC Dev Biol 6: 5.) You may decide.

 トランスポゾン法によるCAR遺伝子導入操作は常法で行えばよく、過去の文献(例えばpiggyBacトランスポゾン法についてはNakazawa Y, et al., J Immunother 32:826-836, 2009、Nakazawa Y et al., J Immunother 6:3-10, 2013、Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5;(69):e4235、Saito S, Nakazawa Y, Sueki A, et al. Anti-leukemic potency of piggyBac-mediated CD19-specific T cells against refractory Philadelphia chromosome-positive acute lymphoblastic leukemia. Cytotherapy. 2014;16:1257-69.)が参考になる。 The CAR gene transfer operation by the transposon method may be performed by a conventional method, and past literature (for example, for the piggyBac transposon method, Nakazawa Y, et al., J Immunother 32: 826-836, 2009, Nakazawa Y et al., J Immunother 6: 3-10, 2013, Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5; (69): e4235, Saito S, Nakazawa Y, Sueki A, et al. Anti-leukemic potential of piggyBac-mediated CD19-specific T cells against refractory Philadelphia chromasome-positive acute lymphoblastic leukemia. Cytotherapy. 2014; 16: 1257-69.) Is helpful.

 本発明の好ましい一態様では、piggyBacトランスポゾン法が採用される。典型的には、piggyBacトランスポゾン法ではpiggyBacトランスポザーゼをコードする遺伝子を搭載したベクター(トランスポザーゼプラスミド)と、所望の核酸コンストラクト(CAR発現カセット及び/又はsiRNA発現カセット)がpiggyBac逆向き反復配列に挟まれた構造を備えるベクター(トランスポゾンプラスミド)を用意し、これらのベクターを標的細胞に導入(トランスフェクション)する。トランスフェクションには、エレクトロポレーション、ヌクレオフェクション、リポフェクション、リン酸カルシウム法など、各種手法を利用できる。 In a preferred embodiment of the present invention, the piggyBac transposon method is adopted. Typically, in the piggyBac transposon method, a vector carrying a gene encoding the piggyBac transposase (transposase plasmid) and a desired nucleic acid construct (CAR expression cassette and / or siRNA expression cassette) are sandwiched between piggyBac reverse repeat sequences. Vectors having a structure (transposon plasmid) are prepared, and these vectors are introduced (transfected) into target cells. For transfection, various methods such as electroporation, nucleofection, lipofection, and calcium phosphate method can be used.

 トランスポゾンプラスミド内において、CAR遺伝子の下流にはポリA付加シグナル配列を配置する。ポリA付加シグナル配列の使用によって転写を終了させる。ポリA付加シグナル配列としてはSV40のポリA付加配列、ウシ由来成長ホルモン遺伝子のポリA付加配列等を用いることができる。 In the transposon plasmid, a poly A addition signal sequence is placed downstream of the CAR gene. Transcription is terminated by the use of the poly A addition signal sequence. As the poly A addition signal sequence, a poly A addition sequence of SV40, a poly A addition sequence of a bovine growth hormone gene, or the like can be used.

 トランスポゾンプラスミドに検出用遺伝子(レポーター遺伝子、細胞又は組織特異的な遺伝子、選択マーカー遺伝子など)、エンハンサー配列、WRPE配列等を含めることにしてもよい。検出用遺伝子は、発現カセットの導入の成否や効率の判定、CAR遺伝子の発現の検出又は発現効率の判定、CAR遺伝子が発現した細胞の選択や分取、等に利用される。一方、エンハンサー配列の使用によって発現効率の向上が図られる。検出用遺伝子としては、ネオマイシンに対する耐性を付与するneo遺伝子、カナマイシン等に対する耐性を付与するnpt遺伝子(Herrera Estrella、EMBO J. 2(1983)、987-995)やnptII遺伝子(Messing & Vierra.Gene 1 9:259-268(1982))、ハイグロマイシンに対する耐性を付与するhph遺伝子(Blochinger & Diggl mann,Mol Cell Bio 4:2929-2931)、メタトレキセートに対する耐性を付与するdhfr遺伝子(Bourouis et al.,EMBO J.2(7))等(以上、マーカー遺伝子)、ルシフェラーゼ遺伝子(Giacomin、P1. Sci. 116(1996)、59~72;Scikantha、J. Bact. 178(1996)、121)、β-グルクロニダーゼ(GUS)遺伝子、GFP(Gerdes、FEBS Lett. 389(1996)、44-47)やその改変体(EGFPやd2EGFPなど)等の蛍光タンパク質の遺伝子(以上、レポーター遺伝子)、細胞内ドメインを欠く上皮成長因子受容体(EGFR)遺伝子等の遺伝子を用いることができる。検出用遺伝子は、例えば、バイシストロニック性制御配列(例えば、リボソーム内部認識配列(IRES))や自己開裂ペプチドをコードする配列を介してCAR遺伝子に連結している。自己開裂ペプチドの例はThosea asigna virus由来の2Aペプチド(T2A)であるが、これに限定されるものではない。自己開裂ペプチドとして蹄疫ウイルス(FMDV)由来の2Aペプチド(F2A)、ウマ鼻炎Aウイルス(ERAV)由来の2Aペプチド(E2A)、porcine teschovirus(PTV-1)由来の2Aペプチド(P2A)等が知られている。 The transposon plasmid may include a detection gene (reporter gene, cell or tissue-specific gene, selectable marker gene, etc.), enhancer sequence, WRPE sequence, and the like. The detection gene is used for determining the success or failure and efficiency of the introduction of the expression cassette, detecting the expression of the CAR gene or determining the expression efficiency, selecting and sorting the cells expressing the CAR gene, and the like. On the other hand, the expression efficiency can be improved by using the enhancer sequence. The genes for detection include the neo gene that imparts resistance to neomycin, the npt gene that imparts resistance to kanamycin, etc. (Herrera Estrella, EMBO J. 2 (1983), 987-995) and the npt II gene (Messing & Vierra. Gene 1). 9: 259-268 (1982)), hph gene conferring resistance to neomycin (Blochinger & Digglmann, Mol Cell Bio 4: 2929-2931), dhfr gene conferring resistance to metatrexate (Bourouis et al. , EMBO J.2 (7)), etc. (marker gene), luciferase gene (Giacomin, P1. Sci. 116 (1996), 59-72; Scikantha, J. Bact. 178 (1996), 121), β -Glucronidase (GUS) gene, genes for fluorescent proteins such as GFP (Gerdes, FEBS Lett. 389 (1996), 44-47) and their variants (EGFP, d2EGFP, etc.), intracellular domains Genes such as the missing epithelial growth factor receptor (EGFR) gene can be used. The detection gene is linked to the CAR gene via, for example, a bicistronic control sequence (eg, ribosome internal recognition sequence (IRES)) or a sequence encoding a self-cleaving peptide. An example of a self-cleaving peptide is, but is not limited to, a 2A peptide (T2A) derived from Thosea signa virus. 2A peptide (F2A) derived from foot-and-mouth disease virus (FMDV), 2A peptide (E2A) derived from horse rhinitis A virus (ERAV), 2A peptide (P2A) derived from porcine teschovirus (PTV-1), etc. are known as self-cleaving peptides. Has been done.

 標的細胞(CAR遺伝子を導入する細胞)として、CD4陽性CD8陰性T細胞、CD4陰性CD8陽性T細胞、iPS細胞から調製されたT細胞、αβ-T細胞、γδ-T細胞、NK細胞、NKT細胞を挙げることができる。上記の如きリンパ球又は前駆細胞を含むものであれば、様々な細胞集団を用いることができる。末梢血から採取される末梢血単核細胞(PBMCs)は好ましい標的細胞の一つである。即ち、好ましい一態様では、T細胞又は前駆細胞を含む細胞集団であるPBMCsに対して遺伝子導入操作を行う。PBMCsは常法で調製すればよい。尚、PBMCsの調製方法については、例えば、Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5;(69):e4235を参照することができる。 Target cells (cells into which the CAR gene is introduced) include CD4-positive CD8-negative T cells, CD4-negative CD8-positive T cells, T cells prepared from iPS cells, αβ-T cells, γδ-T cells, NK cells, and NKT cells. Can be mentioned. Various cell populations can be used as long as they contain lymphocytes or progenitor cells as described above. Peripheral blood mononuclear cells (PBMCs) collected from peripheral blood are one of the preferred target cells. That is, in a preferred embodiment, a gene transfer operation is performed on PBMCs, which are a cell population containing T cells or progenitor cells. PBMCs may be prepared by a conventional method. For the method of preparing PBMCs, refer to, for example, Saha S, Nakazawa Y, Huye LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5; (69): e4235. it can.

ステップ(2)
 ステップ(1)で用意した遺伝子改変リンパ球(CAR細胞)は、特定の条件下での培養に供される(ステップ(2))。一態様(第1態様)では、それが発現するキメラ抗原受容体の抗原認識領域に対する抗イディオタイプ抗体の存在下、ステップ(1)で用意した遺伝子改変リンパ球を培養する。別の態様(第2態様)では、それが発現するキメラ抗原受容体の標的抗原を発現する細胞(標的抗原発現細胞)の存在下、ステップ(1)で用意した遺伝子改変リンパ球を培養する。本発明では、抗イディオタイプ抗体又は標的抗原発現細胞の存在下で培養することにより、目的のCAR細胞(即ち、標的抗原特異的キメラ抗原受容体遺伝子が導入されたもの)を特異的に刺激し、増殖率の向上を図る。
Step (2)
The genetically modified lymphocytes (CAR cells) prepared in step (1) are subjected to culturing under specific conditions (step (2)). In one aspect (first aspect), the genetically modified lymphocyte prepared in step (1) is cultured in the presence of an anti-iditope antibody against the antigen recognition region of the chimeric antigen receptor on which it is expressed. In another aspect (second aspect), the genetically modified lymphocyte prepared in step (1) is cultured in the presence of cells expressing the target antigen of the chimeric antigen receptor on which it is expressed (target antigen expressing cells). In the present invention, the target CAR cells (that is, those into which the target antigen-specific chimeric antigen receptor gene has been introduced) are specifically stimulated by culturing in the presence of anti-iditope antibody or target antigen-expressing cells. , To improve the proliferation rate.

 第1態様に使用する抗イディオタイプ抗体は免疫学的手法、ファージディスプレイ法、リボソームディスプレイ法などを利用して調製すればよい。具体的には例えばPLoS One. 2013; 8(3): e57838に記載の方法などの公知の方法に準じて作製することができる。また、受託サービス(例えば、GenScript社、Creative Biolabs社、株式会社免疫生物研究所、ジーンフロンティア株式会社)を利用して抗イディオタイプ抗体を用意することにしてもよい。抗イディオタイプ抗体はポリクローナル抗体、モノクローナル抗体のいずれであってもよい。尚、抗イディオタイプ抗体は、抗体分子の抗原認識領域(可変ドメイン)を認識する抗体であり、特定のイディオタイプ中のエピトープ(イディオトープ)に結合する。抗イディオタイプ抗体として具体的には、CD19 CARに対する抗イディオタイプ抗体、GD2 CARに対する抗イディオタイプ抗体、CD22 CARに対する抗イディオタイプ抗体、BCMA CARに対する抗イディオタイプ抗体、CD5 CARに対する抗イディオタイプ抗体、CD123 CARに対する抗イディオタイプ抗体、HER2 CARに対する抗イディオタイプ抗体などが挙げられる。好ましくはCD19 CARに対する抗イディオタイプ抗体及びGD2 CARに対する抗イディオタイプ抗体が挙げられる。 The anti-iditope antibody used in the first aspect may be prepared by using an immunological method, a phage display method, a ribosome display method, or the like. Specifically, it can be produced according to a known method such as the method described in PLoS One. 2013; 8 (3): e57838. Further, an anti-iditope antibody may be prepared by using a contract service (for example, GenScript, Creative Biolabs, Immuno-Biological Laboratories, Inc., GeneFrontier Corp.). The anti-iditope antibody may be either a polyclonal antibody or a monoclonal antibody. The anti-iditope antibody is an antibody that recognizes the antigen recognition region (variable domain) of an antibody molecule and binds to an epitope (idiotope) in a specific idiotype. Specifically, the anti-iditope antibody includes an anti-iditope antibody against CD19CAR, an anti-iditopes antibody against GD2CAR, an anti-iditopes antibody against CD22CAR, an anti-iditopes antibody against BCMACAR, and an anti-iditopes antibody against CD5CAR. Examples thereof include an anti-iditope antibody against CD123CAR and an anti-iditopes antibody against HER2CAR. Preferred examples thereof include an anti-iditope antibody against CD19CAR and an anti-iditopes antibody against GD2CAR.

 抗イディオタイプ抗体は、イムノグロブリンの定常領域を含むものであってもよいし、イムノグロブリンの定常領域を含まないものであってもよい。定常領域を含む場合、重鎖の定常領域(CH1、CH2、及びCH3)並びに軽鎖の定常領域(CL)の全てを含んでいてもよいし、これらの内の任意の1種又は2種以上の組み合わせを含んでいてもよい。抗イディオタイプ抗体の具体例としては、例えばイムノグロブリン構造、Fab構造、F(ab’)2構造、ミニボディ(minibody)構造、scFv‐Fc構造、Fv構造、scFv構造、ディアボディ(diabody)構造、トリアボディ(triabody)構造、テトラボディ(tetrabody)構造などが挙げられる。また、抗イディオタイプ抗体には、ナノボディ、モノボディ等の低分子抗体も包含される。 The anti-iditope antibody may contain a constant region of immunoglobulin or may not contain a constant region of immunoglobulin. When the constant region is included, the constant region of the heavy chain (CH1, CH2, and CH3) and the constant region of the light chain (CL) may be all included, and any one or more of these may be included. May include a combination of. Specific examples of anti-iditope antibodies include immunoglobulin structure, Fab structure, F (ab') 2 structure, minibody structure, scFv-Fc structure, Fv structure, scFv structure, and diabody structure. , Triabody structure, tetrabody structure and the like. In addition, anti-iditope antibodies also include low molecular weight antibodies such as Nanobodies and monobodies.

 抗イディオタイプ抗体の存在下で培養することにより、CAR細胞と抗イディオタイプ抗体が接触可能な状態となる。例えば、培養容器の培養面に抗イディオタイプ抗体を固相化しておけば、培養中に当該接触状態を形成することができる。固相化に使用される基板としては、例えばポリスチレン等のプラスチック、ガラス、ニトロセルロースなどを主成分として含む基板が挙げられる。「固相化」とは、抗イディオタイプ抗体が基板に直接又は間接的に結合することにより、固定されていることを意味する。抗イディオタイプ抗体の基板への固定は、定法に従って又は準じて行うことができる。固定の具体的態様としては、特に制限されないが、例えば共有結合を介した固定、アビジンまたはストレプトアビジンとビオチンとの結合を介した固定、物理吸着による固定等が挙げられる。また、ビーズ(例えば磁気ビーズ)に結合した抗イディオタイプ抗体(ビース化抗体)を培養液中に添加することによっても、CAR細胞と抗イディオタイプ抗体が接触可能な状態を形成することができる。ビースの材質としては、特に限定されず、例えば金、銀、銅、鉄、アルミ、ニッケル、マンガン、チタン、これらの酸化物等の金属製粒子; ポリスチレン、ラテックス等の樹脂製粒子; シリカ粒子等が挙げられる。ビースの形状としては、特に制限されないが、例えば球体、直方体、立方体、三角錐等、又はこれらに近い形状が挙げられる。ビースは、好ましくは、抗イディオタイプ抗体の結合をより容易に及び/又はより強固にするための物質を表面に有する。このような物質としては、例えば、エポキシ基、アミノ基、カルボキシ基、アジド基等の反応性基を有する物質; アビジン、プロテインA、プロテインB等の、他の分子に親和性を有する物質等が挙げられる。 By culturing in the presence of anti-iditope antibody, CAR cells and anti-iditope antibody can be brought into contact with each other. For example, if an anti-iditopes antibody is immobilized on the culture surface of a culture vessel, the contact state can be formed during the culture. Examples of the substrate used for solid phase formation include a substrate containing plastic such as polystyrene, glass, nitrocellulose and the like as a main component. By "solid phase" is meant that the anti-iditopes antibody is immobilized by binding directly or indirectly to the substrate. Fixation of the anti-iditopes antibody to the substrate can be performed according to or according to a conventional method. Specific embodiments of fixation are not particularly limited, and examples thereof include fixation via a covalent bond, fixation via a bond between avidin or streptavidin and biotin, and fixation by physical adsorption. Further, by adding an anti-iditope antibody (beaded antibody) bound to beads (for example, magnetic beads) to the culture medium, it is possible to form a state in which CAR cells and the anti-iditope antibody can be contacted. The material of the beads is not particularly limited, and for example, metal particles such as gold, silver, copper, iron, aluminum, nickel, manganese, titanium, and oxides thereof; resin particles such as polystyrene and latex; silica particles and the like. Can be mentioned. The shape of the bead is not particularly limited, and examples thereof include a sphere, a rectangular parallelepiped, a cube, a triangular pyramid, and the like, or a shape close to these. The bead preferably has a substance on its surface to make the binding of the anti-iditope antibody easier and / or stronger. Examples of such substances include substances having a reactive group such as an epoxy group, an amino group, a carboxy group and an azide group; and substances having an affinity for other molecules such as avidin, protein A and protein B. Can be mentioned.

 第2態様に使用する標的抗原発現細胞は、細胞表面上に抗原(特にCAR認識部分)が露出している細胞である限り、特に制限されない。典型的には、標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製することができる。PBMCsを分画することで特定の細胞集団(例えば、T細胞、B細胞、NK細胞、樹状細胞、或いはこれらの中の二つ以上の組合せ)を用意し、当該細胞集団に対して、標的抗原をコードする遺伝子を導入することにしてもよい。また、標的抗原をコードする遺伝子を導入する細胞として、K562等の細胞を用いることもできる。標的抗原の発現は一過性、恒常的のいずれもよい。遺伝子導入には、例えば、プラスミドをはじめとする各種ベクターを利用すればよい。遺伝子導入は、より具体的には、例えば、上記したCAR遺伝子の導入と同様にして行うことができる。 The target antigen-expressing cell used in the second aspect is not particularly limited as long as it is a cell in which the antigen (particularly the CAR recognition portion) is exposed on the cell surface. Typically, it can be prepared by introducing a gene encoding a target antigen into peripheral blood mononuclear cells (PBMCs). By fractionating PBMCs, a specific cell population (for example, T cells, B cells, NK cells, dendritic cells, or a combination of two or more of these) is prepared and targeted to the cell population. A gene encoding an antigen may be introduced. Further, as a cell into which a gene encoding a target antigen is introduced, a cell such as K562 can also be used. The expression of the target antigen may be transient or constant. For gene transfer, for example, various vectors such as a plasmid may be used. More specifically, gene transfer can be carried out in the same manner as, for example, the above-mentioned CAR gene transfer.

 好ましくは、ステップ(1)で用意する遺伝子改変リンパ球(CAR細胞)と標的抗原発現細胞の由来を同一とする。換言すれば、同一の個体から単離した細胞を用いてCAR細胞と標的抗原発現細胞を調製する。このようにすれば、意図しない刺激がCAR細胞に加わることや免疫拒絶の問題、感染症等を回避できる。この態様における、CAR細胞と標的抗原発現細胞の由来である個体は、本発明の調製方法で得られるCAR細胞の投与を受ける患者(レシピエント)(自家移植)又は別の者(同種移植)である。後者(即ち同種移植の場合)は、例えば、血液中のリンパ球が少ない、或いはリンパ球の活性が低い等、CAR細胞の調製に必要な細胞を用意し難い患者に対して適用されるCAR細胞を調製することに好適といえる。 Preferably, the origin of the genetically modified lymphocyte (CAR cell) prepared in step (1) and the target antigen-expressing cell are the same. In other words, CAR cells and target antigen-expressing cells are prepared using cells isolated from the same individual. In this way, it is possible to avoid the addition of unintended stimuli to CAR cells, the problem of immune rejection, infectious diseases, and the like. In this embodiment, the individual from which the CAR cells and the target antigen-expressing cells are derived is a patient (recipient) (autologous transplantation) or another person (allogeneic transplantation) who receives the CAR cells obtained by the preparation method of the present invention. is there. The latter (ie, in the case of allogeneic transplantation) is applied to patients who have difficulty in preparing the cells necessary for the preparation of CAR cells, for example, when there are few lymphocytes in the blood or the activity of lymphocytes is low. It can be said that it is suitable for preparing.

 CAR細胞を用意する際の遺伝子導入操作による損傷/傷害からの回復を促すために、ステップ(1)における、遺伝子導入操作直後ではなく、遺伝子導入操作から2時間~72時間(好ましくは8時間~48時間、さらに好ましくは16時間~24時間)程度経過した後にステップ(2)を実施するとよい。 In order to promote recovery from damage / injury caused by the gene transfer operation when preparing CAR cells, 2 hours to 72 hours (preferably 8 hours to 8 hours) after the gene transfer operation, not immediately after the gene transfer operation in step (1). Step (2) may be performed after 48 hours, more preferably 16 to 24 hours) have elapsed.

 本発明では、非特異的な細胞増殖を抑え、目的のCAR細胞を特異的且つ効率的に増殖させるため、原則として、ステップ(2)の培養の際に抗CD3抗体及び抗CD28抗体による刺激を加えない。一方、細胞の生存率/増殖率を高めるために、ステップ(2)の培養の際、T細胞増殖因子が添加された培養液を使用するとよい。T細胞増殖因子としてはIL-2、IL-7及びIL-15が挙げられるが、IL-15が特に有用である。好ましくは、IL-15に加えIL-7が添加された培養液を用いる。IL-15の添加量は例えば5ng/ml~10ng/mlとする。同様にIL-7の添加量は例えば5ng/ml~10ng/mlとする。IL-15、IL-7等のT細胞増殖因子は常法に従って調製することができる。また、市販品を利用することもできる。ヒト以外の動物種のT細胞増殖因子の使用を排除するものではないが、通常、T細胞増殖因子はヒト由来のもの(組換え体であってもよい)を用いる。ヒトIL-15、ヒトIL-7等の増殖因子は用意に入手することができる(例えばミルテニーバイオテク社、R&Dシステムズ社等が提供する)。 In the present invention, in order to suppress non-specific cell proliferation and proliferate the target CAR cells specifically and efficiently, in principle, stimulation with anti-CD3 antibody and anti-CD28 antibody is performed during the culture in step (2). Do not add. On the other hand, in order to increase the cell viability / proliferation rate, it is advisable to use a culture medium to which a T cell growth factor has been added during the culture in step (2). Examples of T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful. Preferably, a culture solution to which IL-7 is added in addition to IL-15 is used. The amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml. Similarly, the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml. T cell growth factors such as IL-15 and IL-7 can be prepared according to a conventional method. In addition, a commercially available product can also be used. Although the use of T cell growth factors in non-human animal species is not excluded, T cell growth factors are usually derived from humans (may be recombinants). Growth factors such as human IL-15 and human IL-7 can be easily obtained (for example, provided by Miltenyi Biotec, R & D Systems, etc.).

 血清(ヒト血清、ウシ胎仔血清など)を添加した培地を用いてもよいが、無血清培地を採用することにより、臨床応用する際の安全性が高く、且つ血清ロット間の差による培養効率の違いが出にくいという利点を有する細胞を調製することが可能になる。リンパ球用の無血清培地の具体例はTexMACSTM(ミルテニーバイオテク社)、AIM V(登録商標)(Thermo Fisher Scientific社)である。本発明の調製方法で得られるCAR細胞を自己移植に用いる場合には、自己血清(レシピエントの血清)を添加した培地を用いることにしてもよい。基本培地にはリンパ球の培養に適したものを用いればよく、具体例を挙げれば、上掲のTexMACSTM、AIM V(登録商標)である。その他の培養条件は、リンパ球の生存、増殖に適したものであればよく、一般的なものを採用すればよい。例えば、37℃に設定したCO2インキュベーター(CO2濃度5%)内で培養すればよい。 A medium supplemented with serum (human serum, fetal bovine serum, etc.) may be used, but by adopting a serum-free medium, it is highly safe for clinical application and the culture efficiency due to the difference between serum lots is high. It is possible to prepare cells that have the advantage of being less likely to make a difference. Specific examples of serum-free media for lymphocytes are TexMACS TM (Miltenyi Biotec) and AIM V® (Thermo Fisher Scientific). When the CAR cells obtained by the preparation method of the present invention are used for autotransplantation, a medium to which autologous serum (recipient's serum) is added may be used. A medium suitable for lymphocyte culture may be used as the basal medium, and specific examples thereof are TexMACS TM and AIM V (registered trademark) described above. Other culture conditions may be any one suitable for the survival and proliferation of lymphocytes, and general ones may be adopted. For example, the cells may be cultured in a CO 2 incubator (CO 2 concentration 5%) set at 37 ° C.

 ステップ(2)の培養期間は、例えば1日~14日、好ましくは1日~7日、更に好ましくは2日~7日である。培養期間が短すぎると十分な効果が望めず、培養期間が長すぎると細胞の活性(生命力)の低下等のおそれがある。 The culture period of step (2) is, for example, 1 to 14 days, preferably 1 to 7 days, and more preferably 2 to 7 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, the cell activity (life force) may decrease.

 好ましい一態様では、ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入にトランスポゾン法が用いられ、かつステップ(2)における、標的抗原を発現する細胞が、該標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製される。より好ましい一態様では、ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入にpiggyBacトランスポゾン法が用いられ、かつステップ(2)における、標的抗原を発現する細胞が、該標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製される。 In a preferred embodiment, the transposon method is used to introduce the target antigen-specific chimeric antigen receptor gene in step (1), and the cells expressing the target antigen in step (2) encode the target antigen. It is prepared by introducing the gene into peripheral blood mononuclear cells (PBMCs). In a more preferred embodiment, the piggyBac transposon method is used to introduce the target antigen-specific chimeric antigen receptor gene in step (1), and the cells expressing the target antigen in step (2) use the target antigen. It is prepared by introducing the encoding gene into peripheral blood mononuclear cells (PBMCs).

ステップ(3)
 ステップ(2)に続くステップ(3)では、培養後の遺伝子改変リンパ球(CAR細胞)を回収する。回収操作は常法で行えばよい。例えば、ピペッティング、遠心処理等によって回収する。
Step (3)
In step (3) following step (2), the genetically modified lymphocytes (CAR cells) after culturing are collected. The collection operation may be performed by a conventional method. For example, it is collected by pipetting, centrifugation, or the like.

 好ましい一態様では、ステップ(2)とステップ(3)の間に、培養後のCAR細胞をT細胞増殖因子の存在下で培養するステップを行う。このステップによれば、効率的な拡大培養が可能になり、また、細胞の生存率を高める利点もある。 In a preferred embodiment, a step of culturing the cultured CAR cells in the presence of T cell growth factor is performed between steps (2) and (3). This step enables efficient expansion culture and also has the advantage of increasing cell viability.

 T細胞増殖因子としてはIL-15、IL-7等を用いることができる。好ましくは、ステップ(2)と同様に、IL-15とIL-7を添加した培地で培養する。培養期間は例えば1日~21日、好ましくは5日~18日、更に好ましくは10日~14日である。培養期間が短すぎると細胞数の十分な増加を望めず、培養期間が長すぎると細胞の活性(生命力)の低下、細胞の疲弊/疲労等のおそれがある。培養の途中で継代してもよい。また、培養中は必要に応じて培地交換をする。例えば3日に1回の頻度で培養液の1/3~2/3程度を新しい培地に交換する。 IL-15, IL-7, etc. can be used as T cell growth factors. Preferably, the cells are cultured in a medium supplemented with IL-15 and IL-7 in the same manner as in step (2). The culture period is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days. If the culture period is too short, a sufficient increase in the number of cells cannot be expected, and if the culture period is too long, there is a risk of a decrease in cell activity (life force), cell exhaustion / fatigue, and the like. It may be subcultured in the middle of culturing. In addition, the medium is changed as necessary during the culture. For example, replace about 1/3 to 2/3 of the culture medium with a new medium once every 3 days.

1-2.活性化T細胞との共培養によるCAR遺伝子導入操作後の細胞の保護
 CAR遺伝子の導入操作(特に、トランスポゾン法の場合のエレクトロポレーション等)は標的細胞に対して傷害(ダメージ)を与え得るものであり、通常、CAR遺伝子が導入された細胞(即ち、CAR細胞)は、遺伝子導入操作後、傷害を受けた状態にある。そこで、本発明の一態様では、ステップ(1)における遺伝子導入操作によって傷害を受けたCAR細胞を保護し、遺伝子導入効率及び/又は生存率ないし増殖率の向上を図るため、ステップ(1)とステップ(2)の間に、以下のステップ(a)を行う。
 (a)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、抗CD3抗体及び抗CD28抗体で刺激しつつ共培養するステップ
1-2. Protection of cells after CAR gene transfer operation by co-culture with activated T cells CAR gene transfer operation (especially electroporation in the case of transposon method) can cause damage to target cells. In general, the cells into which the CAR gene has been introduced (that is, CAR cells) are in a damaged state after the gene transfer operation. Therefore, in one aspect of the present invention, in order to protect CAR cells damaged by the gene transfer operation in step (1) and improve gene transfer efficiency and / or survival rate or proliferation rate, step (1) and During step (2), the following step (a) is performed.
(A) Non-proliferative cells obtained by stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody and then performing a treatment for losing proliferative ability, and genetically modified lymphocytes prepared in step (1). Steps of mixing lymphocytes and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody

 ステップ(a)では、標的抗原特異的キメラ抗原受容体遺伝子の導入操作後のCAR細胞(ステップ(2)に使用するCAR細胞)の保護に用いる非増殖性細胞を用意し、ステップ(1)で用意したCAR細胞との共培養に供する。 In step (a), non-proliferative cells used for protection of CAR cells (CAR cells used in step (2)) after the transfer operation of the target antigen-specific chimeric antigen receptor gene are prepared, and in step (1). It is used for co-culture with the prepared CAR cells.

 非増殖性細胞を用意するため、まず、T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激する。この処理によって、活性化T細胞が得られる。「T細胞を含む細胞集団」として、好ましくは、末梢血から採取される末梢血単核細胞(PBMCs)を用いる。PBMCsを精製し、T細胞の含有率を高めたものや、末梢血からフェレーシスによって採取した単核球等を、ここでの「T細胞を含む細胞集団」として用いることも可能である。 To prepare non-proliferative cells, first stimulate the cell population containing T cells with anti-CD3 antibody and anti-CD28 antibody. This treatment gives activated T cells. As the "cell population containing T cells", peripheral blood mononuclear cells (PBMCs) collected from peripheral blood are preferably used. It is also possible to use PBMCs purified to increase the T cell content, mononuclear cells collected from peripheral blood by feresis, or the like as the "cell population containing T cells" here.

 例えば、抗CD3抗体と抗CD28抗体で培養面をコートした培養容器(例えば培養皿)で3時間~3日、好ましくは6時間~2日、更に好ましくは12時間~1日、培養することによって、細胞集団内のT細胞に対して抗CD3抗体及び抗CD28抗体による刺激を加えることができる。抗CD3抗体(例えばミルテニーバイオテク社が提供する商品名CD3pure抗体を用いることができる)と抗CD28抗体(例えばミルテニーバイオテク社が提供する商品名CD28pure抗体を用いることができる)は市販もされており、容易に入手可能である。抗CD3抗体と抗CD28抗体がコートされた磁気ビーズ(例えば、VERITAS社が提供するDynabeads T-Activator CD3/CD28)を利用してステップ(a)の刺激を行うことも可能である。尚、抗CD3抗体として「OKT3」クローンを用いることが好ましい。 For example, by culturing in a culture vessel (for example, a culture dish) whose culture surface is coated with anti-CD3 antibody and anti-CD28 antibody for 3 hours to 3 days, preferably 6 hours to 2 days, and more preferably 12 hours to 1 day. , T cells in the cell population can be stimulated with anti-CD3 antibody and anti-CD28 antibody. Anti-CD3 antibody (for example, the trade name CD3pure antibody provided by Miltenyi Biotec) and anti-CD28 antibody (for example, the trade name CD28pure antibody provided by Miltenyi Biotec can be used) are also commercially available. It is easily available. It is also possible to perform the stimulation in step (a) using magnetic beads coated with anti-CD3 antibody and anti-CD28 antibody (for example, Dynabeads T-Activator CD3 / CD28 provided by VERITAS). It is preferable to use an "OKT3" clone as the anti-CD3 antibody.

 抗CD3抗体と抗CD28抗体による刺激を行った細胞は、増殖能を喪失させる処理に供されるが、その前に、T細胞増殖因子の存在下で培養することにするとよい。この培養によって、刺激処理後の細胞の活性が高められる。ここでの培養の期間は例えば1日~10日、好ましくは2日~7日、更に好ましくは3日~4日である。培養期間が短すぎると十分な活性化を望めず、培養期間が長すぎると共刺激分子減弱のおそれがある。培養後の細胞を一旦、凍結保存することにしてもよい。この場合には、使用時に細胞を融解し、再度、抗CD3抗体及びCD28抗体による刺激(条件は上記に準ずる)を行った後に「増殖能を喪失させる処理」に供するとよい。 Cells stimulated with anti-CD3 antibody and anti-CD28 antibody are subjected to a treatment to lose proliferative ability, but before that, it is advisable to culture in the presence of T cell growth factor. This culture enhances the activity of the cells after the stimulation treatment. The culture period here is, for example, 1 to 10 days, preferably 2 to 7 days, and more preferably 3 to 4 days. If the culture period is too short, sufficient activation cannot be expected, and if the culture period is too long, co-stimulatory molecules may be attenuated. The cultured cells may be cryopreserved once. In this case, the cells may be thawed at the time of use, stimulated again with anti-CD3 antibody and CD28 antibody (conditions conform to the above), and then subjected to "treatment to lose proliferative ability".

 「増殖能を喪失させる処理」を経ることによって、増殖能を喪失した活性化T細胞(非増殖性細胞)が得られる。増殖能を喪失させる処理は、典型的には放射線照射であるが、薬剤を用いることにしてもよい。放射線照射の条件の一例を示すと、ガンマ線を用い、25Gy~50Gyの強度で15~30分間の処理である。 Activated T cells (non-proliferative cells) that have lost their proliferative ability can be obtained by undergoing the "treatment that causes them to lose their proliferative ability". The treatment that causes the loss of proliferative capacity is typically irradiation, but agents may also be used. An example of irradiation conditions is a treatment using gamma rays at an intensity of 25 Gy to 50 Gy for 15 to 30 minutes.

 以上のようにして用意した非増殖性細胞と、ステップ(1)で用意したCAR細胞を混合し、抗CD3抗体及び抗CD28抗体で刺激しつつ共培養する。これによって、非増殖性細胞による共刺激分子を介した刺激と抗CD3抗体及び抗CD28抗体による刺激が加わり、CAR細胞が活性化するとともに、その生存・増殖が促される。この共培養は、CAR細胞を調製する際のCAR遺伝子導入操作の後、速やかに実施することが好ましい。例えば、CAR遺伝子導入操作の直後又は1日以内に共培養を開始する。尚、この共培養(即ちステップ(a))と上記ステップ(2)を同時に行うことにしてもよく、その場合にはステップ(2)の開始時期がステップ(a)の開始時期と同じ(例えば、CAR遺伝子導入操作の直後又は1日以内)になる。 The non-proliferative cells prepared as described above and the CAR cells prepared in step (1) are mixed and co-cultured while stimulating with anti-CD3 antibody and anti-CD28 antibody. As a result, stimulation by co-stimulatory molecules by non-proliferative cells and stimulation by anti-CD3 antibody and anti-CD28 antibody are added, CAR cells are activated, and their survival and proliferation are promoted. This co-culture is preferably carried out immediately after the CAR gene transfer operation when preparing CAR cells. For example, co-culture is started immediately after the CAR gene transfer operation or within 1 day. The co-culture (that is, step (a)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (a) (for example). , Immediately after the CAR gene transfer operation or within 1 day).

 共培養に使用する非増殖性細胞の数と遺伝子改変リンパ球(CAR細胞)の数の比率(非増殖性細胞の数/遺伝子改変リンパ球の数)は特に限定されないが、例えば、0.025~0.5とする。 The ratio of the number of non-proliferative cells to the number of genetically modified lymphocytes (CAR cells) used for co-culture (number of non-proliferative cells / number of genetically modified lymphocytes) is not particularly limited, but is, for example, 0.025 to 0.5. And.

 細胞の生存率/増殖率を高めるために、共培養の際、T細胞増殖因子が添加された培養液を使用するとよい。T細胞増殖因子としてはIL-2、IL-7及びIL-15が挙げられるが、IL-15が特に有用である。好ましくは、IL-15に加えIL-7が添加された培養液を用いる。IL-15の添加量は例えば5ng/ml~10ng/mlとする。同様にIL-7の添加量は例えば5ng/ml~10ng/mlとする。 In order to increase the cell viability / proliferation rate, it is advisable to use a culture medium to which T cell growth factor has been added during co-culture. Examples of T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful. Preferably, a culture solution to which IL-7 is added in addition to IL-15 is used. The amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml. Similarly, the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml.

 血清(ヒト血清、ウシ胎仔血清など)を添加した培地を用いてもよいが、無血清培地を採用することにより、臨床応用する際の安全性が高く、且つ血清ロット間の差による培養効率の違いが出にくいという利点を有する細胞を調製することが可能になる。リンパ球用の無血清培地の具体例はTexMACSTM(ミルテニーバイオテク社)、AIM V(登録商標)(Thermo Fisher Scientific社)である。本発明の調製方法で得られるCAR細胞を自己移植に用いる場合には、自己血清(レシピエントの血清)を添加した培地を用いることにしてもよい。基本培地にはリンパ球の培養に適したものを用いればよく、具体例を挙げれば、上掲のTexMACSTM、AIM V(登録商標)である。その他の培養条件は、リンパ球の生存、増殖に適したものであればよく、一般的なものを採用すればよい。例えば、37℃に設定したCO2インキュベーター(CO2濃度5%)内で培養すればよい。 A medium supplemented with serum (human serum, fetal bovine serum, etc.) may be used, but by adopting a serum-free medium, it is highly safe for clinical application and the culture efficiency due to the difference between serum lots is high. It is possible to prepare cells that have the advantage of being less likely to make a difference. Specific examples of serum-free media for lymphocytes are TexMACS TM (Miltenyi Biotec) and AIM V® (Thermo Fisher Scientific). When the CAR cells obtained by the preparation method of the present invention are used for autotransplantation, a medium to which autologous serum (recipient's serum) is added may be used. A medium suitable for lymphocyte culture may be used as the basal medium, and specific examples thereof are TexMACS TM and AIM V (registered trademark) described above. Other culture conditions may be any one suitable for the survival and proliferation of lymphocytes, and general ones may be adopted. For example, the cells may be cultured in a CO 2 incubator (CO 2 concentration 5%) set at 37 ° C.

 共培養の期間は、例えば1日~10日、好ましくは1日~7日、更に好ましくは2日~4日である。培養期間が短すぎると十分な効果が望めず、培養期間が長すぎると細胞の活性(生命力)の低下等のおそれがある。 The co-culture period is, for example, 1 to 10 days, preferably 1 to 7 days, and more preferably 2 to 4 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, the cell activity (life force) may decrease.

1-3.非増殖性PBMCsとの共培養によるCAR遺伝子導入操作後の細胞の保護
 本発明の別の一態様では、上記1-2.の別法として、ステップ(1)とステップ(2)の間に、以下のステップ(b)を行う。
 (b)PBMCsをそのまま、又は抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる非増殖性PBMCsと、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ
1-3. Protection of cells after CAR gene transfer operation by co-culture with non-proliferative PBMCs In another aspect of the present invention, the above 1-2. Alternatively, the following step (b) is performed between steps (1) and (2).
(B) Non-proliferative PBMCs obtained by stimulating PBMCs as they are or with anti-CD3 antibody and anti-CD28 antibody and then performing a treatment to lose proliferative ability, and genetically modified lymphocytes prepared in step (1). Steps to mix and co-culture

 ステップ(b)では、標的抗原特異的キメラ抗原受容体遺伝子の導入操作後のCAR細胞(ステップ(2)に使用するCAR細胞)の保護に用いる非増殖性PBMCsを用意し、ステップ(1)で用意したCAR細胞との共培養に供する。 In step (b), non-proliferative PBMCs used for protection of CAR cells (CAR cells used in step (2)) after the transfer operation of the target antigen-specific chimeric antigen receptor gene are prepared, and in step (1). It is used for co-culture with the prepared CAR cells.

 PBMCsを抗CD3抗体及び抗CD28抗体で刺激する場合の方法は上記1-2.と同様である。増殖能を喪失させる処理、並びにその前にT細胞増殖因子の存在下で培養すること及びその方法も上記1-2.と同様である。尚、「PBMCsをそのまま」とは、PBMCsに抗CD3抗体及び抗CD28抗体による刺激を加えないことを意味する。 The method for stimulating PBMCs with anti-CD3 antibody and anti-CD28 antibody is as described in 1-2 above. Is similar to. The treatment for losing the proliferative ability, and prior to that, culturing in the presence of T cell growth factor and the method thereof are also described in 1-2. Is similar to. In addition, "as is PBMCs" means that PBMCs are not stimulated by anti-CD3 antibody and anti-CD28 antibody.

 以上のようにして用意した非増殖性PBMCsと、ステップ(1)で用意した遺伝子改変リンパ球(CAR細胞)を混合し、共培養する。抗CD3抗体及び抗CD28抗体で刺激しつつ共培養してもよい。この共培養は、CAR細胞を調製する際のCAR遺伝子導入操作の後、速やかに実施することが好ましい。例えば、CAR遺伝子導入操作の直後又は1日以内に共培養を開始する。尚、この共培養(即ちステップ(b))と上記ステップ(2)を同時に行うことにしてもよく、その場合にはステップ(2)の開始時期がステップ(b)の開始時期と同じ(例えば、CAR遺伝子導入操作の直後又は1日以内)になる。 The non-proliferative PBMCs prepared as described above are mixed with the genetically modified lymphocytes (CAR cells) prepared in step (1) and co-cultured. Co-culture may be performed while stimulating with an anti-CD3 antibody and an anti-CD28 antibody. This co-culture is preferably carried out immediately after the CAR gene transfer operation when preparing CAR cells. For example, co-culture is started immediately after the CAR gene transfer operation or within 1 day. The co-culture (that is, step (b)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (b) (for example). , Immediately after the CAR gene transfer operation or within 1 day).

 共培養に使用する非増殖性PBMCsの数と遺伝子改変リンパ球(CAR細胞)の数の比率(非増殖性PBMCsの数/遺伝子改変リンパ球の数)は特に限定されないが、例えば、0.025~0.5、好ましくは0.05~1.0とする。 The ratio of the number of non-proliferative PBMCs used for co-culture to the number of genetically modified lymphocytes (CAR cells) (number of non-proliferative PBMCs / number of genetically modified lymphocytes) is not particularly limited, but is, for example, 0.025 to 0.5. , Preferably 0.05 to 1.0.

 細胞の生存率/増殖率を高めるために、共培養の際、T細胞増殖因子が添加された培養液を使用するとよい。T細胞増殖因子としてはIL-2、IL-7及びIL-15が挙げられるが、IL-15が特に有用である。好ましくは、IL-15に加えIL-7が添加された培養液を用いる。IL-15の添加量は例えば5ng/ml~10ng/mlとする。同様にIL-7の添加量は例えば5ng/ml~10ng/mlとする。尚、言及しない条件(血清の利用の可能性、基本培地、培養温度など)は、1-2.の態様の場合と同様である。 In order to increase the cell viability / proliferation rate, it is advisable to use a culture medium to which T cell growth factor has been added during co-culture. Examples of T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful. Preferably, a culture solution to which IL-7 is added in addition to IL-15 is used. The amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml. Similarly, the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml. Conditions not mentioned (possibility of serum utilization, basal medium, culture temperature, etc.) are described in 1-2. Is the same as in the case of the above aspect.

 非増殖性PBMCsを共培養の途中で(例えば共培養開始後3~11日目に、好ましくは5~9日目に、さらに好ましくは7日目に)追加してもよい。或いは、共培養後の細胞を回収し、別の非増殖性PBMCsと混合した後に再度、培養を行うことにしてもよい。これらの操作を2回以上繰り返すことにしてもよい。このように、非増殖性PBMCsを利用した刺激ないし活性化を複数回行うことにすれば、CAR細胞の誘導率の向上、CAR細胞数の増加を望める。尚、改めて用意したもの、又は最初に用意しておいた細胞の一部を培養しておいたもの若しくは凍結保存しておいたものを、ここでの非増殖性PBMCsとして使用することができる。 Non-proliferative PBMCs may be added in the middle of co-culture (for example, 3 to 11 days after the start of co-culture, preferably 5 to 9 days, and more preferably 7 days). Alternatively, the cells after co-culture may be collected, mixed with other non-proliferative PBMCs, and then cultured again. These operations may be repeated twice or more. In this way, if stimulation or activation using non-proliferative PBMCs is performed a plurality of times, it is expected that the induction rate of CAR cells will be improved and the number of CAR cells will be increased. In addition, those prepared again, or those prepared by culturing a part of the cells prepared at the beginning or those stored in a cryopreservation can be used as the non-proliferative PBMCs here.

 尚、1回の採血で得た末梢血から分離したPBMCsの一部を用いて非増殖性PBMCsを調製するとともに、他の一部からCAR細胞を調製することにすれば、本発明の実施に伴う採血回数を低減することができ、臨床応用上、極めて大きな利点となる。特に、残りのPBMCsを用いて追加用の非増殖性PBMCsを調製すること、或いはPBMCsの一部を用いて調製した非増殖性PBMCsの一部を保存(典型的には凍結保存)しておき、それを追加用の非増殖性PBMCsとして使用すること、にすれば、必要な3種類の細胞、即ち、CAR細胞、当該細胞との共培養に使用する非増殖性PBMCs、共培養の途中で追加するための非増殖性PBMCsを1回の採血によって用意することができることから、本発明で得られるCAR細胞を用いた治療における、患者の負担は大幅に軽減される。 If non-proliferative PBMCs are prepared using a part of PBMCs isolated from peripheral blood obtained by one blood sampling and CAR cells are prepared from the other part, the present invention can be carried out. The number of blood samplings involved can be reduced, which is an extremely great advantage in clinical application. In particular, prepare additional non-proliferative PBMCs using the remaining PBMCs, or store some of the non-proliferative PBMCs prepared using some of the PBMCs (typically cryopreservation). , If it is used as additional non-proliferative PBMCs, then the three required cells, namely CAR cells, non-proliferative PBMCs used for co-culture with the cells, in the middle of co-culture Since non-proliferative PBMCs for addition can be prepared by a single blood collection, the burden on the patient in the treatment using the CAR cells obtained in the present invention is greatly reduced.

1-4.ウイルスペプチドを保持したT細胞との共培養によるウイルス特異的なキメラ抗原受容体遺伝子改変リンパ球の調製
 本発明の別の一態様では、ウイルス特異的なキメラ抗原受容体遺伝子改変リンパ球(ウイルス特異的CAR細胞)が調製される。ウイルス特異的CAR細胞は、自家移植に利用する場合にはウイルスT細胞受容体からの刺激による体内持続性の向上が望めること、同種移植に利用する場合には更に同種免疫反応(GVHD)の軽減により移植ドナーからのCAR細胞作製が可能になり、しかも第3者ドナーからのCAR細胞を製剤化できる可能性があることなど、臨床応用上、重要な利点を有する。実際、ウイルス特異的CAR細胞がより長期に体内に持続することが報告されている(Pule MA, et al. Nat Med. 2008 Nov;14(11):1264-70.)。また、第3者由来EBV特異的CTL臨床研究の報告(Annual Review血液2015、2015年1月発行、中外医学社)により、ウイルス特異的細胞傷害性T細胞(CTL)の安全性が高いことが裏づけられている。
1-4. Preparation of virus-specific chimeric antigen receptor gene-modified lymphocytes by co-culture with T cells carrying a virus peptide In another aspect of the present invention, virus-specific chimeric antigen receptor gene-modified lymphocytes (virus-specific). CAR cells) are prepared. Virus-specific CAR cells can be expected to improve internal persistence by stimulation from viral T cell receptors when used for autologous transplantation, and further reduce allogeneic immune response (GVHD) when used for allogeneic transplantation. This has important advantages in clinical application, such as the possibility of producing CAR cells from a transplant donor and the possibility of formulating CAR cells from a third-party donor. In fact, virus-specific CAR cells have been reported to persist in the body for longer periods of time (Pule MA, et al. Nat Med. 2008 Nov; 14 (11): 1264-70.). In addition, according to a report of a third-party EBV-specific CTL clinical study (Annual Review Blood 2015, published in January 2015, Chugai Medical Co., Ltd.), the safety of virus-specific cytotoxic T cells (CTL) is high. It is backed up.

 この態様では、ウイルス特異的CAR細胞を調製するため、以下のステップ(c)を行う。尚、言及しない事項(例えば、T細胞を含む細胞集団の調製方法、抗CD3抗体及び抗CD28抗体による刺激の基本的な操作、増殖能を喪失させる処理の方法、共培養の基本的な操作等)については上記の1-2.の態様と同様であるため、重複する説明を省略し、対応する説明を援用する。
 (c)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、ウイルスペプチド抗原存在下での培養及び増殖能を喪失させる処理を行うことによって得られる、ウイルスペプチド抗原を保持した非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ
In this aspect, the following step (c) is performed to prepare virus-specific CAR cells. Matters not mentioned (for example, method for preparing a cell population containing T cells, basic operation for stimulation with anti-CD3 antibody and anti-CD28 antibody, method for processing to lose proliferative ability, basic operation for co-culture, etc. ) Is described in 1-2. Since it is the same as the aspect of the above, duplicate explanations are omitted and the corresponding explanations are used.
(C) A cell population containing T cells was stimulated with an anti-CD3 antibody and an anti-CD28 antibody, and then cultured in the presence of the viral peptide antigen and treated to lose the proliferative ability to retain the viral peptide antigen. The step of mixing non-proliferative cells with the genetically modified lymphocytes prepared in step (1) and co-culturing them.

 ステップ(c)では、まず、T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激し、活性化T細胞を得る。その後、ウイルスペプチド抗原存在下での培養と増殖能を喪失させる処理を行う。これによって、非増殖性の「ウイルスペプチド抗原を細胞表面に保持した活性化T細胞」(以下、「ウイルスペプチド保持非増殖性細胞」と呼ぶ)が得られる。ウイルスペプチド抗原存在下での培養と、増殖能を喪失させる処理の順序は特に限定されない。従って、ウイルスペプチド抗原存在下で培養した後に増殖能を喪失させても、或いは増殖能を喪失させた後にウイルスペプチド抗原存在下で培養することにしてもよい。好ましくは、増殖能を喪失する前の方がウイルスペプチド抗原の取り込みがより良好であろうという期待から前者の順序を採用する。ウイルスペプチド抗原存在下で培養するためには、例えば、ウイルスペプチド抗原が添加された培地を用いればよい。或いは、培養中にウイルスペプチド抗原を培地に添加すればよい。ウイルスペプチド抗原の添加濃度は例えば0.5μg/ml~1μg/mlとする。培養期間は例えば10分~5時間、好ましくは20分~3時間とする。本明細書における「ウイルスペプチド抗原」とは、特定のウイルスに特異的な細胞傷害性T細胞(CTL)を誘導しうるエピトープペプチドまたはエピトープを含むロングペプチドをいう。ウイルスペプチド抗原としては、これらに限定されるものではないが、例えばアデノウイルス(AdV)の抗原ペプチド(例えば、WO 2007015540 A1を参照)、サイトメガロウイルス(CMV)の抗原ペプチド(例えば、特開2002-255997号公報、特開2004-242599号公報、特開2012-87126号公報を参照)、エプスタインバールウイルス(EBV)の抗原ペプチド(例えば、WO 2007049737 A1、特願2011-177487号公報、特開2006-188513号公報を参照)、等を用いることができる。ウイルスペプチド抗原は配列情報に基づき常法(例えば液相合成法、固相合成法)で調製することができる。また、ウイルスペプチド抗原の中には市販されているものもある(例えば株式会社医学生物学研究所、タカラバイオ、ミルテニーバイオテクなどが提供する。) In step (c), first, a cell population containing T cells is stimulated with an anti-CD3 antibody and an anti-CD28 antibody to obtain activated T cells. Then, the cells are cultured in the presence of the viral peptide antigen and treated to lose their proliferative ability. As a result, non-proliferative "activated T cells holding a viral peptide antigen on the cell surface" (hereinafter referred to as "viral peptide-retaining non-proliferative cells") are obtained. The order of culturing in the presence of the viral peptide antigen and the treatment for losing the proliferative ability is not particularly limited. Therefore, the growth ability may be lost after culturing in the presence of the virus peptide antigen, or the culture may be performed in the presence of the virus peptide antigen after the loss of the growth ability. Preferably, the former order is adopted because of the expectation that the uptake of the viral peptide antigen will be better before the loss of proliferative capacity. In order to culture in the presence of the viral peptide antigen, for example, a medium to which the viral peptide antigen has been added may be used. Alternatively, the viral peptide antigen may be added to the medium during culturing. The concentration of the viral peptide antigen added is, for example, 0.5 μg / ml to 1 μg / ml. The culture period is, for example, 10 minutes to 5 hours, preferably 20 minutes to 3 hours. As used herein, the term "viral peptide antigen" refers to an epitope peptide or a long peptide containing an epitope capable of inducing cytotoxic T cells (CTL) specific to a specific virus. The viral peptide antigen is not limited to these, but is, for example, an adenovirus (AdV) antigen peptide (see, for example, WO2007015540 A1) and a cytomegalovirus (CMV) antigen peptide (for example, JP-A-2002). -255997, 2004-242599, 2012-87126), Epstein-Barr virus (EBV) antigen peptide (for example, WO 2007049737 A1, Japanese Patent Application No. 2011-177487, Japanese Patent Application Laid-Open No. (See 2006-188513), etc. can be used. The viral peptide antigen can be prepared by a conventional method (for example, liquid phase synthesis method, solid phase synthesis method) based on the sequence information. In addition, some viral peptide antigens are commercially available (for example, provided by Medical & Biological Laboratories, Ltd., Takara Bio, Miltenyi Biotec, etc.).

 1種類の抗原ペプチドを用いることも可能であるが、通常は2種類以上の抗原ペプチド(抗原ペプチド混合物)を使用する。例えば、AdV抗原ペプチド混合物、CMV抗原ペプチド混合物又はEBV抗原ペプチド混合物、或いはこれら抗原ペプチド混合物の中の二つ以上を組み合わせたもの(例えば、AdV抗原ペプチド混合物、CMV抗原ペプチド混合物及びEBV抗原ペプチド混合物を混合したもの)を用いる。2種類以上の抗原ペプチドを併用することにより、標的(抗原ペプチド)が異なる複数の活性化T細胞を得ることができ、本発明の調製方法で得られるウイルス特異的CAR細胞が有効な治療対象(患者)の増大(カバー率の向上)を望める。いずれのウイルスに由来する抗原ペプチドを使用するかを決定するにあたっては、本発明の調製方法で得られるウイルス特異的CAR細胞の用途、具体的には治療対象となる疾患や患者の病態を考慮するとよい。例えば、造血幹細胞移植後の再発性白血病の治療を目的とする場合には、EBVウイルスの抗原ペプチド混合物を単独で又は他のウイルスの抗原ペプチド混合物との併用で用いるとよい。AdV抗原ペプチド混合物、CMV抗原ペプチド混合物、EBV抗原ペプチド混合物については市販もされており(例えば、ミルテニーバイオテク社が提供する、PepTivator(登録商標) AdV5 Hexon、PepTivator(登録商標) CMV pp65、PepTivator(登録商標) EBV EBNA-1、PepTivator(登録商標) EBV BZLF1、JPT Peptide Technologies社が提供するPepMixTM Collection HCMV、PepMixTM EBV (EBNA1)等)、容易に入手することができる。 Although it is possible to use one type of antigen peptide, usually two or more types of antigen peptides (antigen peptide mixture) are used. For example, AdV antigen peptide mixture, CMV antigen peptide mixture or EBV antigen peptide mixture, or a combination of two or more of these antigen peptide mixture (for example, AdV antigen peptide mixture, CMV antigen peptide mixture and EBV antigen peptide mixture). Use a mixture). By using two or more kinds of antigen peptides in combination, a plurality of activated T cells having different targets (antigen peptides) can be obtained, and the virus-specific CAR cells obtained by the preparation method of the present invention are effective therapeutic targets ( We can expect an increase in the number of patients (improvement of coverage). In deciding which virus-derived antigen peptide to use, the use of the virus-specific CAR cells obtained by the preparation method of the present invention, specifically, the disease to be treated and the pathological condition of the patient should be considered. Good. For example, when the purpose is to treat recurrent leukemia after hematopoietic stem cell transplantation, the antigen-peptide mixture of EBV virus may be used alone or in combination with an antigen-peptide mixture of other viruses. AdV antigen-peptide mixture, CMV antigen-peptide mixture, and EBV antigen-peptide mixture are also commercially available (for example, PepTivator (registered trademark) AdV5 Hexon, PepTivator (registered trademark) CMV pp65, PepTivator (for example, provided by Milteny Biotech). Registered trademarks) EBV EBNA-1, PepTivator (registered trademark) EBV BZLF1, PepMix TM Collection HCMV provided by JPT Peptide Technologies, PepMix TM EBV (EBNA1), etc.) can be easily obtained.

 以上のようにして用意したウイルスペプチド保持非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球(CAR細胞)を混合し、共培養する。これによって、ウイルスペプチド保持非増殖性細胞による共刺激分子及びウイルス抗原ペプチドを介した刺激が加わり、ウイルス抗原特異的な遺伝子改変リンパ球が活性化するとともに、その生存・増殖が促される。ステップ(1)における遺伝子導入操作によって傷害を受けたCAR細胞を保護し、遺伝子導入効率及び/又は生存率ないし増殖率の向上を図るため、この共培養は、CAR細胞を調製する際のCAR遺伝子導入操作の後、速やかに実施することが好ましい。例えば、CAR遺伝子導入操作の直後又は1日以内に共培養を開始する。尚、この共培養(即ちステップ(c))と上記ステップ(2)を同時に行うことにしてもよく、その場合にはステップ(2)の開始時期がステップ(c)の開始時期と同じ(例えば、CAR遺伝子導入操作の直後又は1日以内)になる。 The viral peptide-retaining non-proliferative cells prepared as described above and the genetically modified lymphocytes (CAR cells) prepared in step (1) are mixed and co-cultured. As a result, stimulation via a co-stimulating molecule and a virus antigen peptide by a virus peptide-carrying non-proliferative cell is applied, and virus antigen-specific genetically modified lymphocytes are activated and their survival and proliferation are promoted. In order to protect CAR cells damaged by the gene transfer operation in step (1) and improve gene transfer efficiency and / or survival rate or proliferation rate, this co-culture is used for CAR genes in preparing CAR cells. It is preferable to carry out immediately after the introduction operation. For example, co-culture is started immediately after the CAR gene transfer operation or within 1 day. The co-culture (that is, step (c)) and the above step (2) may be performed at the same time, in which case the start time of step (2) is the same as the start time of step (c) (for example). , Immediately after the CAR gene transfer operation or within 1 day).

 共培養に使用するウイルスペプチド保持非増殖性細胞の数と遺伝子改変リンパ球(CAR細胞)の数の比率(ウイルスペプチド保持非増殖性細胞の数/遺伝子改変リンパ球の数)は特に限定されないが、例えば、0.025~0.5、好ましくは0.05~1.0とする。 The ratio of the number of viral peptide-carrying non-proliferative cells used for co-culture to the number of genetically modified lymphocytes (CAR cells) (number of viral peptide-carrying nonproliferative cells / number of genetically modified lymphocytes) is not particularly limited. For example, 0.025 to 0.5, preferably 0.05 to 1.0.

 このステップは、ウイルス特異的CAR細胞を選択的に増殖させるため、強力な刺激を避けてT細胞の疲弊/疲労を防ぐため、等の理由から、原則として、抗CD3抗体及び抗CD28抗体による刺激を加えない。一方、細胞の生存率/増殖率を高めるために、共培養の際、T細胞増殖因子が添加された培養液を使用するとよい。T細胞増殖因子としてはIL-2、IL-7及びIL-15が挙げられるが、IL-15が特に有用である。好ましくは、IL-15に加えIL-7が添加された培養液を用いる。IL-15の添加量は例えば5ng/ml~10ng/mlとする。同様にIL-7の添加量は例えば5ng/ml~10ng/mlとする。尚、言及しない条件(血清の利用の可能性、基本培地、培養温度など)は、1-2.の態様の場合と同様である。 In principle, this step is to stimulate with anti-CD3 antibody and anti-CD28 antibody for reasons such as selectively proliferating virus-specific CAR cells, avoiding strong stimulation and preventing T cell exhaustion / fatigue. Do not add. On the other hand, in order to increase the cell viability / proliferation rate, it is advisable to use a culture medium to which a T cell growth factor has been added during co-culture. Examples of T cell growth factors include IL-2, IL-7 and IL-15, of which IL-15 is particularly useful. Preferably, a culture solution to which IL-7 is added in addition to IL-15 is used. The amount of IL-15 added is, for example, 5 ng / ml to 10 ng / ml. Similarly, the amount of IL-7 added is, for example, 5 ng / ml to 10 ng / ml. Conditions not mentioned (possibility of serum utilization, basal medium, culture temperature, etc.) are described in 1-2. Is the same as in the case of the above aspect.

 ウイルスペプチド保持非増殖性細胞を共培養の途中で追加してもよい。或いは、共培養後の細胞を回収し、別のウイルスペプチド保持非増殖性細胞と混合した後に再度、共培養を行うことにしてもよい。これらの操作を2回以上繰り返すことにしてもよい。このように、ウイルスペプチド保持非増殖性細胞を利用した刺激なしし活性化を複数回行うことにすれば、ウイルス特異的CAR細胞の誘導率の向上、ウイルス特異的CAR細胞数の増加を望める。尚、改めて用意したもの、又は最初に用意しておいた細胞の一部を保存しておいたものを、ここでの「別のウイルスペプチド保持非増殖性細胞」として使用することができる。 Viral peptide-carrying non-proliferative cells may be added during co-culture. Alternatively, the cells after co-culture may be collected, mixed with another virus peptide-carrying non-proliferative cell, and then co-cultured again. These operations may be repeated twice or more. As described above, if the activation without stimulation using the virus peptide-retaining non-proliferative cells is performed a plurality of times, the induction rate of the virus-specific CAR cells can be improved and the number of virus-specific CAR cells can be expected to increase. In addition, a cell prepared again or a cell in which a part of the cell prepared at the beginning is preserved can be used as "another viral peptide-carrying non-proliferative cell" here.

 共培養の期間は、例えば1日~21日、好ましくは5日~18日、更に好ましくは10日~14日である。培養期間が短すぎると十分な効果を望めず、培養期間が長すぎると細胞の活性(生命力)の低下、細胞の疲弊/疲労等のおそれがある。 The co-culture period is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days. If the culturing period is too short, a sufficient effect cannot be expected, and if the culturing period is too long, there is a risk of a decrease in cell activity (life force), cell exhaustion / fatigue, and the like.

 ウイルスペプチド保持非増殖性細胞との共培養の前に、ステップ(1)で用意した遺伝子改変リンパ球(CAR細胞)をウイルスペプチド保持非増殖性末梢血単核細胞(PBMCs)と共培養することにしてもよい。ここでの共培養の期間は、例えば1日~21日、好ましくは5日~18日、更に好ましくは10日~14日である。この態様の場合、ステップ(1)で用意した遺伝子改変リンパ球(CAR細胞)とウイルスペプチド保持非増殖性PBMCsの共培養(1段階目の共培養)で得られた細胞が、上記の方法で用意したウイルスペプチド保持非増殖性細胞と混合され、2段階目の共培養が行われることになる。ここでのウイルスペプチド保持非増殖性PBMCsは、PBMCsをウイルスペプチド抗原存在下での培養及び増殖能を喪失させる処理に供することによって調製することができる。具体的には、例えば、末梢血から分離したPBMCsを放射線処理した後、ウイルスペプチド抗原存在下で培養し、ウイルスペプチド保持非増殖性PBMCsを得る。尚、1回の採血で得た末梢血から分離したPBMCsの一部を用いてウイルスペプチド保持非増殖性PBMCsを調製するとともに、他の一部からCAR細胞を調製することにすれば、本発明の実施に伴う採血回数を低減することができ、臨床応用上、極めて大きな利点となる。特に、残りのPBMCsを用いてウイルスペプチド保持非増殖性細胞(2段階目の共培養に使用する細胞)を調製すること、或いはPBMCsの一部を用いて調製したウイルスペプチド保持非増殖性PBMCsの一部を保存(典型的には凍結保存)しておき、それをウイルスペプチド保持非増殖性細胞(2段階目の共培養に使用する細胞)として使用すること、にすれば、必要な3種類の細胞、即ち、CAR細胞、当該細胞との共培養(1段階目の共培養)に使用するウイルスペプチド保持非増殖性PBMCs、2段階目の共培養に使用するウイルスペプチド保持非増殖性細胞を1回の採血によって用意することができることから、本発明で得られるCAR細胞を用いた治療における、患者の負担は大幅に軽減される。 Prior to co-culturing with viral peptide-carrying non-proliferative cells, the genetically modified lymphocytes (CAR cells) prepared in step (1) are co-cultured with viral peptide-carrying non-proliferative peripheral blood mononuclear cells (PBMCs). It may be. The period of co-culture here is, for example, 1 to 21 days, preferably 5 to 18 days, and more preferably 10 to 14 days. In the case of this embodiment, the cells obtained by co-culturing the genetically modified lymphocytes (CAR cells) prepared in step (1) and the viral peptide-carrying non-proliferative PBMCs (co-culture in the first step) are obtained by the above method. It will be mixed with the prepared viral peptide-carrying non-proliferative cells and co-cultured in the second stage. The viral peptide-retaining non-proliferative PBMCs here can be prepared by subjecting the PBMCs to a culture in the presence of a viral peptide antigen and a treatment for losing the ability to proliferate. Specifically, for example, PBMCs isolated from peripheral blood are radiation-treated and then cultured in the presence of a viral peptide antigen to obtain viral peptide-retaining non-proliferative PBMCs. In addition, if it is decided to prepare viral peptide-retaining non-proliferative PBMCs using a part of PBMCs isolated from peripheral blood obtained by one blood sampling and to prepare CAR cells from the other part, the present invention. It is possible to reduce the number of blood samplings associated with the implementation of the above, which is an extremely great advantage in clinical application. In particular, the remaining PBMCs are used to prepare viral peptide-retaining non-proliferative cells (cells used for the second stage co-culture), or the viral peptide-retaining non-proliferative PBMCs prepared using a part of PBMCs. If a part is preserved (typically cryopreserved) and used as a viral peptide-carrying non-proliferative cell (cell used for the second stage co-culture), the necessary three types Cells, that is, CAR cells, viral peptide-retaining non-proliferative PBMCs used for co-culture with the cells (first-stage co-culture), and viral peptide-retaining non-proliferative cells used for second-stage co-culture. Since it can be prepared by collecting blood once, the burden on the patient in the treatment using the CAR cells obtained in the present invention is greatly reduced.

2.キメラ抗原受容体を発現する遺伝子改変リンパ球及びその用途
 本発明の更なる局面は、本発明の調製方法で得られた、キメラ抗原受容体を発現する遺伝子改変リンパ球(以下、「本発明のCAR細胞」と呼ぶ)及びその用途に関する。本発明のCAR細胞はCAR療法が有効と考えられる各種疾患(以下、「標的疾患」と呼ぶ)の治療、予防又は改善に利用され得る。標的疾患の代表はがんであるが、これに限定されるものではない。標的疾患の例を挙げると、各種B細胞リンパ腫(濾胞性悪性リンパ腫、びまん性悪性リンパ腫、マントル細胞リンパ腫、MALTリンパ腫、血管内B細胞性リンパ腫、CD20陽性ホジキンリンパ腫など)、骨髄増殖性腫瘍、骨髄異形成/骨髄増殖性腫瘍(CMML、JMML、CML、MDS/MPN-UC)、骨髄異形成症候群、急性骨髄生白血病、神経芽腫、脳腫瘍、ユーイング肉腫、骨肉腫、網膜芽細胞腫、肺小細胞腫、メラノーマ、卵巣がん、横紋筋肉腫、腎臓がん、膵臓がん、悪性中皮腫、前立腺がん等である。「治療」とは、標的疾患に特徴的な症状又は随伴症状を緩和すること(軽症化)、症状の悪化を阻止ないし遅延すること等が含まれる。「予防」とは、疾病(障害)又はその症状の発症/発現を防止又は遅延すること、或いは発症/発現の危険性を低下させることをいう。一方、「改善」とは、疾病(障害)又はその症状が緩和(軽症化)、好転、寛解、又は治癒(部分的な治癒を含む)することをいう。
2. Gene-modified lymphocytes expressing chimeric antigen receptor and their uses Further aspects of the present invention are the genetically modified lymphocytes expressing chimeric antigen receptor obtained by the preparation method of the present invention (hereinafter, "the present invention". "CAR cells") and their uses. The CAR cells of the present invention can be used for the treatment, prevention or amelioration of various diseases (hereinafter referred to as "target diseases") for which CAR therapy is considered to be effective. The representative of the target disease is cancer, but it is not limited to this. Examples of target diseases include various B-cell lymphomas (follicle malignant lymphoma, diffuse malignant lymphoma, mantle cell lymphoma, MALT lymphoma, intravascular B-cell lymphoma, CD20-positive hodgkin lymphoma, etc.), myeloproliferative neoplasm, bone marrow. Hypoplastic / myeloproliferative neoplasms (CMML, JMML, CML, MDS / MPN-UC), myelodysplastic syndrome, acute myeloproliferative leukemia, neuroblastoma, brain tumor, Ewing sarcoma, osteosarcoma, retinoblastoma, small lung Celloma, melanoma, ovarian cancer, horizontal print myeloma, kidney cancer, pancreatic cancer, malignant mesoderma, prostate cancer, etc. "Treatment" includes alleviating (mitigating) the symptoms characteristic of the target disease or associated symptoms, preventing or delaying the exacerbation of the symptoms, and the like. "Prevention" means preventing or delaying the onset / delay of a disease (disorder) or its symptoms, or reducing the risk of onset / onset. On the other hand, "improvement" means that the disease (disorder) or its symptoms are alleviated (mild), improved, ameliorated, or cured (including partial cure).

 本発明のCAR細胞を細胞製剤の形態で提供することもできる。本発明の細胞製剤には、本発明のCAR細胞が治療上有効量含有される。例えば1回の投与用として、104個~1010個の細胞を含有させる。細胞の保護を目的としてジメチルスルフォキシド(DMSO)や血清アルブミン等、細菌の混入を阻止する目的で抗生物質等、細胞の活性化、増殖又は分化誘導などを目的とした各種の成分(ビタミン類、サイトカイン、成長因子、ステロイド等)等の成分を細胞製剤に含有させてもよい。 The CAR cells of the present invention can also be provided in the form of a cell preparation. The cell preparation of the present invention contains a therapeutically effective amount of the CAR cells of the present invention. For example, it contains 10 4 to 10 10 cells for a single dose. Various components (vitamins) such as dimethylsulfoxide (DMSO) and serum albumin for the purpose of protecting cells, antibiotics for the purpose of preventing bacterial contamination, and various components (vitamins) for the purpose of activating, proliferating or inducing differentiation of cells. , Cytokines, growth factors, steroids, etc.) may be contained in the cell preparation.

 本発明のCAR細胞又は細胞製剤の投与経路は特に限定されない。例えば、静脈内注射、動脈内注射、門脈内注射、皮内注射、皮下注射、筋肉内注射、又は腹腔内注射によって投与する。全身投与によらず、局所投与することにしてもよい。局所投与として、目的の組織・臓器・器官への直接注入を例示することができる。投与スケジュールは、対象(患者)の性別、年齢、体重、病態などを考慮して作成すればよい。単回投与の他、連続的又は定期的に複数回投与することにしてもよい。 The administration route of the CAR cell or cell preparation of the present invention is not particularly limited. For example, it is administered by intravenous injection, intraarterial injection, intraportal injection, intradermal injection, subcutaneous injection, intramuscular injection, or intraperitoneal injection. Topical administration may be used instead of systemic administration. As local administration, direct injection into a target tissue / organ / organ can be exemplified. The administration schedule may be prepared in consideration of the gender, age, body weight, pathological condition, etc. of the subject (patient). In addition to a single dose, multiple doses may be administered continuously or periodically.

<遺伝子導入効率/細胞増殖率の向上をもたらす新規CAR-T調製法>
 CAR-T細胞を調製する際の遺伝子導入効率/細胞増殖率の向上を目指し、以下の新規調製法(培養法2、3)を創出した。尚、比較のため、従来の調製法(培養法1)も記載する。
<New CAR-T preparation method that improves gene transfer efficiency / cell proliferation rate>
The following new preparation methods (culture methods 2 and 3) were created with the aim of improving the gene transfer efficiency / cell proliferation rate when preparing CAR-T cells. For comparison, the conventional preparation method (culture method 1) is also described.

1.材料
(1)抗体
 抗CD3抗体(ミルテニーバイオテク社)
 抗CD28抗体(ミルテニーバイオテク社)
 抗イディオタイプ抗体(公知の方法(例えばPLoS One. 2013; 8(3): e57838.に記載の方法)に準じて作製したL細胞由来のCD19CARに対する抗イディオタイプ抗体)
(2)培地
 TexMACSTM(ミルテニーバイオテク社)
(3)サイトカイン
 リコンビナントヒトIL-7(ミルテニーバイオテク社)
 リコンビナントヒトIL-15(ミルテニーバイオテク社)
(4)ウイルスペプチドミックス
 PepTivator(登録商標)CMV pp65-premium grade, ヒト(ミルテニーバイオテク社社) PepTivator(登録商標)AdV5 Hexon-premium grade, ヒト(ミルテニーバイオテク社) PepTivator(登録商標)EBV EBNA-1-premium grade, ヒト(ミルテニーバイオテク社) PepTivator(登録商標)EBV BZLF1-premium grade, ヒト(ミルテニーバイオテク社)
(5)プラスミド
 pIRII-CD19CARベクター(CARを発現する)
 pCMV-piggyBacベクター(piggyBacトランスポサーゼを発現する)
(6)細胞培養容器
 24ウェル非コート組織培養プレート(Falcon)
 24ウェル組織培養プレート(Falcon)
 G-Rex10 (Wilson Wolf)
1. 1. Material (1) Antibody Anti-CD3 antibody (Miltenyi Biotec)
Anti-CD28 antibody (Miltenyi Biotec)
Anti-iditope antibody (anti-iditope antibody against CD19CAR derived from L cells prepared according to a known method (for example, the method described in PLoS One. 2013; 8 (3): e57838.))
(2) Medium TexMACS TM (Miltenyi Biotec)
(3) Cytokine recombinant human IL-7 (Miltenyi Biotec)
Recombinant Human IL-15 (Miltenyi Biotec)
(4) Viral Peptide Mix PepTivator® CMV pp65-premium grade, Human (Miltenyi Biotec) PepTivator® AdV5 Hexon-premium grade, Human (Miltenyi Biotec) PepTivator® EBV EBNA -1-premium grade, human (Miltenyi Biotec) PepTivator® EBV BZLF1-premium grade, human (Miltenyi Biotec)
(5) plasmid pIRII-CD19CAR vector (expressing CAR)
pCMV-piggyBac vector (expressing piggyBac transposase)
(6) Cell culture container 24-well uncoated tissue culture plate (Falcon)
24-well tissue culture plate (Falcon)
G-Rex10 (Wilson Wolf)

2.方法
(1)抗CD3抗体/抗CD28抗体コート(感作)プレートの作製(従来法の培養法1に使用) 抗CD3抗体と抗CD28抗体を1mg/mlとなるようにPBSで希釈し、24ウェルの非コートプレートに0.5ml/ウェルになるように加える。プレートは37℃のインキュベーターにて2~4時間静置する。抗体希釈PBSを吸引し、1ウェルあたり1mlのPBSで1回洗浄する。
2. Method (1) Preparation of anti-CD3 antibody / anti-CD28 antibody coated (sensitized) plate (used in the conventional culture method 1) Anti-CD3 antibody and anti-CD28 antibody were diluted with PBS to 1 mg / ml, and 24 Add 0.5 ml / well to the uncoated plate of the well. The plate is allowed to stand in an incubator at 37 ° C for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.

(2)抗イデオタイプ抗体コート(感作)プレートの作製(培養法2に使用)
 抗イディオタイプ抗体を0.5mg/mlとなるようにPBSで希釈し、24wellの非コートプレートに0.5ml/ウェルになるように加える。プレートは37℃のインキュベーターにて2~4時間静置する。抗体希釈PBSを吸引し、1ウェルあたり1mlのPBSで1回洗浄する。
(2) Preparation of anti-ideotype antibody-coated (sensitized) plate (used in culture method 2)
The anti-iditope antibody is diluted with PBS to 0.5 mg / ml and added to a 24-well uncoated plate to 0.5 ml / well. The plate is allowed to stand in an incubator at 37 ° C for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.

(3)標的抗原遺伝子導入PBMCsの作製(培養法3に使用)
 0日目:末梢血から単核球(PBMCs)を分離する。PBMCsに対し、CARの標的抗原を発現するプラスミドベクター 5μgをエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入し、インキュベーター内で培養を開始する。
 1日目:放射線照射後、フィーダー(feeder)細胞として使用する。
(3) Preparation of target antigen gene-introduced PBMCs (used in culture method 3)
Day 0: Isolate mononuclear cells (PBMCs) from peripheral blood. For PBMCs, 5 μg of a plasmid vector expressing the CAR target antigen is introduced by electroporation (nucleofection), and culture is started in an incubator.
Day 1: After irradiation, it is used as a feeder cell.

(4)従来法によるCAR-Tの作製及び培養(培養法1、図1を参照)
 0日目:末梢血から単核球を分離し、カウントする。1x107個の単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加して、4D nucleofector(ロンザ)を使用してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。その後、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:抗CD3抗体/抗CD28抗体コートプレートで刺激する。
 4日目:G-Rex10に細胞を移し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養する。
 7日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
(4) Preparation and culturing of CAR-T by the conventional method (see culturing method 1 and FIG. 1)
Day 0: Mononuclear cells are separated from peripheral blood and counted. To 1x10 7 monocytes, add 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5), and electroporate using 4D nucleofector (Lonza). Gene transfer by ration (nucleofection). Then, the cells are floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and culture is started in a 37 ° C. incubator on a 24-well plate.
Day 1: Stimulate with anti-CD3 antibody / anti-CD28 antibody coated plate.
Day 4: Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 7: Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

(5)抗イディオタイプ抗体を利用したCAR-Tの作製及び培養(培養法2、図2を参照) 0日目:末梢血から単核球を分離し、1x107個の単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。その後、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:抗イディオタイプ抗体コートプレートで刺激する。
 4日目:G-Rex10に細胞を移し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養する。
 7日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
(5) Preparation and culture of CAR-T using anti-iditope antibody (see culture method 2 and FIG. 2) Day 0: Mononuclear cells were isolated from peripheral blood, and 1x10 7 mononuclear cells were treated. , PIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) are added 5 ng each and gene is introduced by electroporation (nucleofection). Then, the cells are floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and culture is started in a 37 ° C. incubator on a 24-well plate.
Day 1: Stimulate with an anti-iditope antibody coated plate.
Day 4: Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 7: Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

(6)標的抗原発現細胞(標的抗原遺伝子導入PBMCs)を利用したCAR-Tの作製及び培養(培養法3、図3を参照)
 0日目:末梢血から単核球を分離し、1x107個の単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。その後、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:標的抗原遺伝子導入PBMCs(フィーダー細胞)で刺激する(例えば、フィーダー細胞を播種した培養皿で培養する)。
 4日目:G-Rex10に細胞を移し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養する。
 7日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
(6) Preparation and culture of CAR-T using target antigen-expressing cells (target antigen gene-introduced PBMCs) (see culture method 3 and FIG. 3).
Day 0: Mononuclear cells were isolated from peripheral blood, and 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) were added to 1x10 7 mononuclear cells. Then, the gene is introduced by electroporation (nucleofection). Then, the cells are floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and culture is started in a 37 ° C. incubator on a 24-well plate.
Day 1: Stimulate with target antigen gene-introduced PBMCs (feeder cells) (eg, culture in a culture dish inoculated with feeder cells).
Day 4: Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 7: Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

3.結果
 従来法、又は抗イディオタイプ抗体を利用したCAR-Tの作製及び培養(A)
 上述の2.方法に記載した方法と同様の方法により、(1)抗CD3抗体/抗CD28抗体コート(感作)プレートの作製、及び(2)抗イディオタイプ抗体コート(感作)プレートの作製を行った。
 最初に7日間従来法(培養法1)で培養した後に、さらに従来法(培養法1)で7日間培養したもの(A-1)と、最初に7日間従来法(培養法1)で培養した後に、新規方法(培養法2)で7日間培養したもの(A-2)との間で遺伝子導入効率を比較した。
3. 3. Results Preparation and culture of CAR-T using conventional methods or anti-iditope antibodies (A)
2. above. (1) An anti-CD3 antibody / anti-CD28 antibody-coated (sensitized) plate was prepared, and (2) an anti-iditopes antibody-coated (sensitized) plate was prepared by the same method as described in the method.
First, the cells were cultured by the conventional method (culture method 1) for 7 days, then further cultured by the conventional method (culture method 1) for 7 days (A-1), and first cultured by the conventional method (culture method 1) for 7 days. After that, the gene transfer efficiency was compared with that of the one (A-2) cultured for 7 days by the new method (culture method 2).

 培養終了後(14日目)、培養法1(従来法)(A-1)と培養法2(A-2)について、遺伝子導入効率(CAR-T細胞/全生細胞)、CAR-T細胞数を求め、比較した。尚、遺伝子導入効率はフローサイトメトリー解析の結果から算出し、CAR-T細胞数は7日目からの増殖率で評価した。 After the completion of culturing (14th day), for culture method 1 (conventional method) (A-1) and culture method 2 (A-2), gene transfer efficiency (CAR-T cells / whole viable cells), CAR-T cells The numbers were calculated and compared. The gene transfer efficiency was calculated from the results of flow cytometry analysis, and the number of CAR-T cells was evaluated by the proliferation rate from the 7th day.

 遺伝子導入効率は、培養法1(A-1)が8%、培養法2(A-2)が18.1%であり(図6A)、従来法の培養法1に比較し、培養法2は大幅な遺伝子導入効率の改善を認めた。即ち、新規培養法が遺伝子導入効率の向上に有効であることが実証された。また、CAR-T細胞の7日目からの増殖率は、抗イディオタイプ抗体刺激(A-2)では20倍、従来法のOKT3/抗CD28抗体刺激(A-1)では13倍であった(図6B)。新規培養法はトランスポゾン法の弱点(ウイルスベクター法と比較して遺伝子導入効率や細胞生存率/細胞増殖率が低いこと)を克服し、トランスポゾン法を利用したCAR療法の臨床応用の拡大をもたらすといえる。また、新規培養法はウイルスベクター法等でCAR細胞を調製する場合にも適用できる汎用性の高いものであり、その利用価値は大きい。 The gene transfer efficiency was 8% for culture method 1 (A-1) and 18.1% for culture method 2 (A-2) (Fig. 6A), and culture method 2 was significantly higher than that of conventional culture method 1. Improvement of gene transfer efficiency was observed. That is, it was demonstrated that the novel culture method is effective in improving the efficiency of gene transfer. The proliferation rate of CAR-T cells from day 7 was 20-fold with anti-iditopes antibody stimulation (A-2) and 13-fold with conventional OKT3 / anti-CD28 antibody stimulation (A-1). (Fig. 6B). The new culture method overcomes the weaknesses of the transposon method (gene transfer efficiency and cell viability / cell proliferation rate are lower than those of the viral vector method), and will expand the clinical application of CAR therapy using the transposon method. I can say. In addition, the novel culture method is highly versatile and can be applied to the preparation of CAR cells by the viral vector method or the like, and its utility value is great.

 上述の2.方法に記載した方法と同様の方法により、(1)抗CD3抗体/抗CD28抗体コート(感作)プレートの作製、及び(2)抗イデオタイプ抗体コート(感作)プレートの作製を行った。 2. (1) An anti-CD3 antibody / anti-CD28 antibody-coated (sensitized) plate and (2) an anti-ideotype antibody-coated (sensitized) plate were prepared by the same method as described in the method.

 従来法、又は抗イディオタイプ抗体を利用したCAR-Tの作製及び培養(B)
 0日目:末梢血から単核球を分離し、1x107個の単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。その後、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:抗イディオタイプ抗体コートプレート(B-2、培養法2)またはOKT3/抗CD28抗体コートプレート(B-1、培養法1(従来法))で刺激する。
 4日目:24ウェルプレートに細胞を移し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養する。
 7日目:培養を終了
Preparation and culture of CAR-T using conventional methods or anti-iditope antibodies (B)
Day 0: Mononuclear cells were isolated from peripheral blood, and 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) were added to 1x10 7 mononuclear cells. Then, the gene is introduced by electroporation (nucleofection). Then, the cells are floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and culture is started in a 37 ° C. incubator on a 24-well plate.
Day 1: Stimulate with anti-iditope antibody coated plate (B-2, culture method 2) or OKT3 / anti-CD28 antibody coated plate (B-1, culture method 1 (conventional method)).
Day 4: Transfer cells to a 24-well plate and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 7: Culture finished

結果
 培養終了後(7日目)、遺伝子導入効率(CAR-T細胞/全生細胞)、CAR-T細胞数を求め、比較した。尚、遺伝子導入効率はフローサイトメトリー解析の結果から算出し、CAR-T細胞数は7日目の増殖率で評価した。
Results After the completion of culturing (7th day), the gene transfer efficiency (CAR-T cells / whole viable cells) and the number of CAR-T cells were determined and compared. The gene transfer efficiency was calculated from the results of flow cytometry analysis, and the number of CAR-T cells was evaluated by the proliferation rate on the 7th day.

 遺伝子導入効率は、抗イディオタイプ抗体刺激(B-2、培養法2)が20.5%、OKT3/抗CD28抗体刺激(B-1、培養法1(従来法))が3.58%であり(図8A)、従来法のOKT3/抗CD28抗体刺激に比較し、抗イディオタイプ抗体刺激は大幅な遺伝子導入効率の改善を認めた。また、CAR-T細胞数は、抗イディオタイプ抗体刺激(B-2)が55万個、OKT3/抗CD28抗体刺激(B-1)が18万個であり(図8B)、従来法のOKT3/抗CD28抗体刺激に比較し、抗イディオタイプ抗体刺激は大幅なCAR-T細胞数の改善を認めた。 The gene transfer efficiency was 20.5% for anti-iditope antibody stimulation (B-2, culture method 2) and 3.58% for OKT3 / anti-CD28 antibody stimulation (B-1, culture method 1 (conventional method)) (Fig. 8A). ), Compared with the conventional method of OKT3 / anti-CD28 antibody stimulation, anti-iditopes antibody stimulation showed a significant improvement in gene transfer efficiency. The number of CAR-T cells was 550,000 for anti-iditopes antibody stimulation (B-2) and 180,000 for OKT3 / anti-CD28 antibody stimulation (B-1) (Fig. 8B). / Compared with anti-CD28 antibody stimulation, anti-iditope antibody stimulation showed a significant improvement in CAR-T cell number.

<遺伝子導入後のCAR-T細胞の保護とウイルス特異的CAR-T細胞の調製>
 上記の培養法2、3の応用として、活性化T細胞による刺激を組み合わせた方法(培養法4)とウイルスペプチド添加活性化T細胞による刺激を組み合わせた方法(培養法5)を以下に示す。
<Protection of CAR-T cells after gene transfer and preparation of virus-specific CAR-T cells>
As applications of the above culture methods 2 and 3, a method combining stimulation with activated T cells (culture method 4) and a method combining stimulation with virus peptide-added activated T cells (culture method 5) are shown below.

1.材料
(1)抗体
 抗CD3抗体(ミルテニーバイオテク社)
 抗CD28抗体(ミルテニーバイオテク社)
 抗イディオタイプ抗体(公知の方法(例えばPLoS One. 2013; 8(3): e57838.記載の方法)に準じて作製したL細胞由来のCD19CARに対する抗イディオタイプ抗体)
(2)培地
 TexMACS(ミルテニーバイオテク社)
(3)サイトカイン
 リコンビナントヒトIL-7(ミルテニーバイオテク社)
 リコンビナントヒトIL-15(ミルテニーバイオテク社)
(4)ウイルスペプチドミックス
 PepTivator(登録商標)CMV pp65-premium grade, ヒト(ミルテニーバイオテク社社) PepTivator(登録商標)AdV5 Hexon-premium grade, ヒト(ミルテニーバイオテク社) PepTivator(登録商標)EBV EBNA-1-premium grade, ヒト(ミルテニーバイオテク社) PepTivator(登録商標)EBV BZLF1-premium grade, ヒト(ミルテニーバイオテク社)
(5)プラスミド
 pIRII-CD19CARベクター(CARを発現する)
 pCMV-piggyBacベクター(piggyBacトランスポサーゼを発現する)
(6)細胞培養容器
 24ウェル非コート組織培養プレート(Falcon)
 24ウェル組織培養プレート(Falcon)
 G-Rex10 (Wilson Wolf)
1. 1. Material (1) Antibody Anti-CD3 antibody (Miltenyi Biotec)
Anti-CD28 antibody (Miltenyi Biotec)
Anti-iditope antibody (anti-iditope antibody against CD19CAR derived from L cells prepared according to a known method (for example, the method described in PLoS One. 2013; 8 (3): e57838.))
(2) Medium TexMACS (Miltenyi Biotec)
(3) Cytokine recombinant human IL-7 (Miltenyi Biotec)
Recombinant Human IL-15 (Miltenyi Biotec)
(4) Viral Peptide Mix PepTivator® CMV pp65-premium grade, Human (Miltenyi Biotec) PepTivator® AdV5 Hexon-premium grade, Human (Miltenyi Biotec) PepTivator® EBV EBNA -1-premium grade, human (Miltenyi Biotec) PepTivator® EBV BZLF1-premium grade, human (Miltenyi Biotec)
(5) plasmid pIRII-CD19CAR vector (expressing CAR)
pCMV-piggyBac vector (expressing piggyBac transposase)
(6) Cell culture container 24-well uncoated tissue culture plate (Falcon)
24-well tissue culture plate (Falcon)
G-Rex10 (Wilson Wolf)

2.方法
(1)活性化T細胞の準備
(1-1)抗CD3抗体/抗CD28抗体コート(感作)プレートの作製
 抗CD3抗体と抗CD28抗体を1mg/mlとなるようにPBSで希釈し、24ウェルの非コートプレートに0.5ml/ウェルになるように加える。プレートは37℃のインキュベーターにて2~4時間静置する。抗体希釈PBSを吸引し、1ウェルあたり1mlのPBSで1回洗浄する。
2. Method (1) Preparation of activated T cells (1-1) Preparation of anti-CD3 antibody / anti-CD28 antibody coated (sensitized) plate Anti-CD3 antibody and anti-CD28 antibody were diluted with PBS to 1 mg / ml and diluted with PBS. Add to a 24-well uncoated plate to 0.5 ml / well. The plate is allowed to stand in an incubator at 37 ° C for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.

(1-2)抗CD3抗体/抗CD28抗体コートプレートでの培養
 0日目:末梢血から分離したPBMCを、IL-15 5ng/mlになるように添加したTexMACSTMで5x105/mlになるように希釈し、抗CD3抗体/抗CD28抗体コートプレートに1ウェルあたり2mlずつ分注する。
 1日目:24ウェル組織培養プレートへ細胞を移す。培養液は半量交換し、IL-15 5ng/mlとなるように添加する。
 4日目:IL-15を5ng/mlとなるように添加する。
 7日目:細胞を回収し、分注して凍結保存する。
(1-2) Culture on anti-CD3 antibody / anti-CD28 antibody coated plate Day 0: PBMC isolated from peripheral blood was added to IL-15 5 ng / ml to 5 x 10 5 / ml with TexMACS TM. And dispense 2 ml per well into anti-CD3 antibody / anti-CD28 antibody coated plates.
Day 1: Transfer cells to a 24-well tissue culture plate. Replace half of the culture solution and add to IL-15 5 ng / ml.
Day 4: Add IL-15 to 5 ng / ml.
Day 7: Cells are collected, dispensed and cryopreserved.

(1-3)活性化T細胞の再刺激
 0日目:凍結保存したあった細胞を解凍し、2回洗浄後に、1x106個/mlとなるようにIL15 5ng/ml添加TexMACSTMで希釈し、抗CD3抗体/抗CD28抗体コートプレートへ1ウェル 2mlずつ分注する。
 3日目:細胞を回収し、CAR-T培養に使用する。
(1-3) Restimulation of activated T cells Day 0: Thaw the cryopreserved cells, wash twice, and dilute with IL15 5 ng / ml-added TexMACS TM to 1x10 6 cells / ml. , Dispense 1 well 2 ml each into anti-CD3 antibody / anti-CD28 antibody coated plates.
Day 3: Collect cells and use for CAR-T culture.

(2)抗イディオタイプ抗体コート(感作)プレートの作製
 抗イディオタイプ抗体を0.5mg/mlとなるようにPBSで希釈し、24wellの非コートプレートに0.5ml/ウェルになるように加える。プレートは37℃のインキュベーターにて2~4時間静置する。抗体希釈PBSを吸引し、1ウェルあたり1mlのPBSで1回洗浄する。
(2) Preparation of anti-iditope antibody-coated (sensitized) plate Dilute the anti-iditope antibody with PBS to 0.5 mg / ml, and add to a 24-well uncoated plate to 0.5 ml / well. The plate is allowed to stand in an incubator at 37 ° C for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.

(3)標的抗原遺伝子導入PBMCsの作製
 0日目:末梢血から単核球(PBMCs)を分離する。PBMCsに対し、CARの標的抗原を発現するプラスミドベクター 5μgをエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入し、インキュベーター内で培養を開始する。
 1日目:放射線照射後、フィーダー(feeder)細胞として使用する。
(3) Preparation of target antigen gene-introduced PBMCs Day 0: Mononuclear cells (PBMCs) are isolated from peripheral blood. For PBMCs, 5 μg of a plasmid vector expressing the CAR target antigen is introduced by electroporation (nucleofection), and culture is started in an incubator.
Day 1: After irradiation, it is used as a feeder cell.

(4)活性化T細胞による刺激を組み合わせた方法(培養法4)
 0日目:末梢血から単核球を分離し、1x107個の単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。遺伝子導入した細胞と放射線照射した5x105個の活性化T細胞を混合し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:抗イディオタイプ抗体コートプレート又は標的抗原遺伝子導入PBMCs(フィーダー細胞)で刺激する。
 4日目:G-Rex10に細胞を移し、IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養する。
 7日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
(4) Method combining stimulation with activated T cells (culture method 4)
Day 0: Mononuclear cells were isolated from peripheral blood, and 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) were added to 1x10 7 mononuclear cells. Then, the gene is introduced by electroporation (nucleofection). Transgenic cells and irradiated with 5x10 5 cells of activated T cells were mixed, cultured in an incubator of IL-7 10ng / ml and 37 ° C. at floated 24-well plates in IL15 5 ng / ml added TeXmacs TM Start.
Day 1: Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
Day 4: Transfer cells to G-Rex 10 and incubate with TexMACS TM supplemented with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 7: Replace half of the culture with TexMACS TM with IL-7 10 ng / ml and IL 15 5 ng / ml.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

 以上の培養法4では、共刺激分子の発現による細胞刺激(エレクトロポレーションの際にダメージを受けた遺伝子導入細胞の保護作用)、培養微小環境によるサイトカイン刺激などによって、CAR-T細胞数の更なる増加と遺伝子導入効率の更なる上昇を期待できる。 In the above culture method 4, the number of CAR-T cells is increased by cell stimulation (protective action of transgenic cells damaged during electroporation) by expression of co-stimulating molecules, cytokine stimulation by culture microenvironment, and the like. It can be expected that the increase and the efficiency of gene transfer will increase further.

(5)ウイルスペプチド添加活性化T細胞による刺激を組み合わせた方法(培養法5)
 0日目:放射線照射した活性化T細胞5x105個にウイルスペプチド(PepTivator CMV pp65、PepTivator AdV5 Hexon、PepTivator EBV EBNA-1及びPepTivator EBV BZLF1を各50ng)を添加し、37℃で30分インキュベートする。1x107個の末梢血単核球に対し、pIRII-CAR.CD19.28zベクター(図4)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。遺伝子導入した細胞と放射線照射したウイルスペプチド添加活性化T細胞を混合し、IL-7 10ng/mlとIL15 5ng/mlを添加したTexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。
 1日目:抗イディオタイプ抗体コートプレート又は標的抗原遺伝子導入PBMCs(フィーダー細胞)で刺激する。
 2日目~5日目:必要に応じてIL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 7日目:細胞を回収しカウントする。上記と同様の方法で調製したウイルスペプチド添加活性化T細胞2x106個と回収した細胞を、IL-15(5ng/ml)とIL-7(10ng/ml)添加培養液30ml中に浮遊させ、G-Rex10で培養を開始する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
(5) Method combining stimulation with activated T cells added with viral peptide (culture method 5)
Day 0: Irradiated activated T cells 5x10 5 cells to viral peptide (PepTivator CMV pp65, PepTivator AdV5 Hexon , the PepTivator EBV EBNA-1 and PepTivator EBV BZLFl each 50 ng) is added and incubated for 30 minutes at 37 ° C. .. To 1x10 7 peripheral blood mononuclear cells, add 5 ng each of pIRII-CAR.CD19.28z vector (Fig. 4) and pCMV-pigBac vector (Fig. 5) and electroporate (nucleofection). Introduce the gene. The transgenic cells and irradiated viral peptide-added activated T cells are mixed, floated on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and cultured in a 24-well plate in a 37 ° C incubator. To start.
Day 1: Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
Day 2-5: If necessary, replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 7: Cells are collected and counted. 6 virus peptide-added activated T cells prepared in the same manner as above and the recovered cells were suspended in 30 ml of IL-15 (5 ng / ml) and IL-7 (10 ng / ml) -added culture medium. Start culturing with G-Rex10.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

 以上の培養法5では、共刺激分子の発現による細胞刺激、培養微小環境によるサイトカイン刺激、ウイルス特異的T細胞受容体からの比較的緩やかな細胞刺激などによって、CAR-T細胞数の更なる増加と遺伝子導入効率の更なる上昇を期待できる。また、同種反応性の低下(ウイルス特異的CTLに期待できる効果)、そのことによる第三者由来のCAR-T細胞を利用できる可能性、体内ウイルスによりウイルス抗原受容体に刺激が入ることによる体内持続性(persistency)が上昇する可能性などがあり、安全性の更なる向上や治療効果の増大などが期待される。 In the above culture method 5, the number of CAR-T cells is further increased by cell stimulation by the expression of co-stimulating molecules, cytokine stimulation by the culture microenvironment, and relatively gentle cell stimulation from virus-specific T cell receptors. And further increase in gene transfer efficiency can be expected. In addition, a decrease in allogeneic reactivity (effect that can be expected for virus-specific CTL), the possibility of using CAR-T cells derived from a third party due to this, and the stimulation of virus antigen receptors in the body by a virus in the body. Persistency may increase, and further improvement in safety and increase in therapeutic effect are expected.

<培養法6(培養法5の改良)>
1.方法
 0日目:末梢血から単核球(PBMCs)を分離する。一部(PBMC 1x106個)に放射線照射した後、ウイルスペプチド(PepTivator CMV pp65、PepTivator AdV5 Hexon、PepTivator EBV EBNA-1及びPepTivator EBV BZLF1を各50ng)を添加し、37℃で30分インキュベートする。一方、1x107個のPBMCに対し、pIRII-CAR.CD19_optimizedベクター(図7)とpCMV-pigBacベクター(図5)を5 ngずつ添加してエレクトロポレーション(ヌクレオフェクション)にて遺伝子導入する。遺伝子導入した細胞と放射線照射したウイルスペプチド添加PBMCを混合し、IL-7 10ng/mlとIL15 5ng/mlを添加したTexMACSTMに浮かべて24ウェルプレートにて37℃のインキュベーター中で培養を開始する。尚、上記2.(1)の方法に従い、残りのPBMCsから活性化T細胞を調製しておく。
 1日目:抗イディオタイプ抗体コートプレート又は標的抗原遺伝子導入PBMCs(フィーダー細胞)で刺激する。
 2日目~5日目:必要に応じてIL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 7日目:細胞を回収しカウントする。一方、上記の方法で調製した活性化T細胞2x106個に放射線照射した後、ウイルスペプチド(PepTivator CMV pp65、PepTivator AdV5 Hexon、PepTivator EBV EBNA-1及びPepTivator EBV BZLF1を各50ng)を添加し、37℃で30分インキュベートする。このようにして得たウイルスペプチド添加活性化T細胞2x106個と、回収した細胞を、IL-15(5ng/ml)とIL-7(10ng/ml)添加培養液30ml中に浮遊させ、G-Rex10で培養を開始する。
 10日目:IL-7 10ng/mlとIL15 5ng/ml添加TexMACSTMで培養液を半量交換する。
 14日目:培養を終了する。
<Culture method 6 (improvement of culture method 5)>
1. 1. Method Day 0: Isolate mononuclear cells (PBMCs) from peripheral blood. After irradiating a part ( 6 PBMC 1x10), add viral peptides (PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50 ng each) and incubate at 37 ° C for 30 minutes. On the other hand, with respect to 1x10 7 cells of PBMC, gene transfer by pIRII-CAR.CD19_optimized vector (Fig. 7) and pCMV-pigBac vector electroporation by addition of (Fig. 5) by 5 ng (nucleocapsid transfection). Mix the transgenic cells with irradiated viral peptide-added PBMC, float on TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml, and start culturing in a 37 ° C incubator on a 24-well plate. .. In addition, the above 2. Activated T cells are prepared from the remaining PBMCs according to the method (1).
Day 1: Stimulate with anti-iditope antibody coated plates or target antigen gene-introduced PBMCs (feeder cells).
Day 2-5: If necessary, replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 7: Cells are collected and counted. On the other hand, after irradiating 2x10 6 activated T cells prepared by the above method with virus peptides (PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50 ng each), 37 Incubate at ° C for 30 minutes. The virus peptide-added activated T cells 2x10 6 obtained in this manner and the recovered cells were suspended in 30 ml of IL-15 (5 ng / ml) and IL-7 (10 ng / ml) -added culture medium, and G. -Start culturing with Rex10.
Day 10: Replace half of the culture with IL-7 10 ng / ml and IL 15 5 ng / ml TexMACS TM.
Day 14: The culture is finished.

 以上の培養法6は、この培養法は1回の採血によってCAT-T細胞を得ることを可能にするものであり、患者への負担が軽減するという利点を有する。また、培養法6には遺伝子導入効率の更なる向上も期待できる。 The above culture method 6 makes it possible to obtain CAT-T cells by collecting blood once, and has an advantage that the burden on the patient is reduced. Further, the culture method 6 can be expected to further improve the gene transfer efficiency.

 本発明によれば、CAR細胞を調製する際の遺伝子導入効率ないし細胞増殖率が向上する。即ち本発明は、より多くのCAR細胞を効率的に調製することを可能にするものであり、CAR療法の治療成績の向上、CAR療法の適用範囲の拡大等に貢献し得る。 According to the present invention, the gene transfer efficiency or cell proliferation rate when preparing CAR cells is improved. That is, the present invention makes it possible to efficiently prepare a larger number of CAR cells, and can contribute to improving the therapeutic results of CAR therapy, expanding the scope of application of CAR therapy, and the like.

 この発明は、上記発明の実施の形態及び実施例の説明に何ら限定されるものではない。特許請求の範囲の記載を逸脱せず、当業者が容易に想到できる範囲で種々の変形態様もこの発明に含まれる。本明細書の中で明示した論文、公開特許公報、及び特許公報などの内容は、その全ての内容を援用によって引用することとする。 The present invention is not limited to the description of the embodiments and examples of the above invention. Various modifications are also included in the present invention as long as those skilled in the art can easily conceive without departing from the description of the scope of claims. The contents of the papers, published patent gazettes, patent gazettes, etc. specified in this specification shall be cited by reference in their entirety.

Claims (24)

 以下のステップ(1)~(3)を含む、キメラ抗原受容体を発現する遺伝子改変リンパ球の調製方法:
 (1)標的抗原特異的キメラ抗原受容体遺伝子が導入された遺伝子改変リンパ球を用意するステップ;
 (2)ステップ(1)で用意した遺伝子改変リンパ球を、それが発現するキメラ抗原受容体の抗原認識領域に対する抗イディオタイプ抗体の存在下、又はそれが発現するキメラ抗原受容体の標的抗原を発現する細胞の存在下で培養するステップ;及び
 (3)培養後の遺伝子改変リンパ球を回収するステップ。
Method for preparing genetically modified lymphocytes expressing chimeric antigen receptor, including the following steps (1) to (3):
(1) A step of preparing a genetically modified lymphocyte into which a target antigen-specific chimeric antigen receptor gene has been introduced;
(2) The genetically modified lymphocyte prepared in step (1) is used in the presence of an anti-iditope antibody against the antigen recognition region of the chimeric antigen receptor on which it is expressed, or the target antigen of the chimeric antigen receptor on which it is expressed. The step of culturing in the presence of expressing cells; and (3) the step of collecting the genetically modified lymphocytes after culturing.
 ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入にトランスポゾン法が用いられる、請求項1に記載の調製方法。 The preparation method according to claim 1, wherein the transposon method is used for the introduction of the target antigen-specific chimeric antigen receptor gene in step (1).  トランスポゾン法がpiggyBacトランスポゾン法である、請求項2に記載の調製方法。 The preparation method according to claim 2, wherein the transposon method is the piggyBac transposon method.  ステップ(1)における、標的抗原特異的キメラ抗原受容体遺伝子の導入操作から8時間~48時間経過した後にステップ(2)を行う、請求項1~3のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 3, wherein step (2) is performed after 8 to 48 hours have elapsed from the operation of introducing the target antigen-specific chimeric antigen receptor gene in step (1).  ステップ(2)の培養期間が1日~14日である、請求項1~4のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 4, wherein the culture period of step (2) is 1 to 14 days.  ステップ(2)の培養の際、抗CD3抗体及び抗CD28抗体による刺激を加えない、請求項1~5のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 5, wherein stimulation by an anti-CD3 antibody and an anti-CD28 antibody is not applied during culturing in step (2).  抗イディオタイプ抗体が固相化抗体又はビーズ化抗体である、請求項1~6のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 6, wherein the anti-iditope antibody is a solid phase antibody or a beaded antibody.  T細胞又はその前駆細胞を含む細胞集団に対する、標的抗原特異的キメラ抗原受容体遺伝子の導入によって、ステップ(1)の遺伝子改変リンパ球が得られる、請求項1~7のいずれか一項に記載の調製方法。 The invention according to any one of claims 1 to 7, wherein the genetically modified lymphocyte of step (1) is obtained by introducing a target antigen-specific chimeric antigen receptor gene into a cell population containing T cells or progenitor cells thereof. Preparation method.  細胞集団が末梢血単核細胞(PBMCs)である、請求項8に記載の調製方法。 The preparation method according to claim 8, wherein the cell population is peripheral blood mononuclear cells (PBMCs).  ステップ(2)における、標的抗原を発現する細胞が、該標的抗原をコードする遺伝子を末梢血単核細胞(PBMCs)に導入することによって調製される、請求項1~9のいずれか一項に記載の調製方法。 The cell according to any one of claims 1 to 9, wherein the cell expressing the target antigen in step (2) is prepared by introducing a gene encoding the target antigen into peripheral blood mononuclear cells (PBMCs). The preparation method described.  ステップ(2)における、標的抗原を発現する細胞と、遺伝子改変リンパ球が同一の個体に由来する、請求項1~10のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 10, wherein in step (2), the cell expressing the target antigen and the genetically modified lymphocyte are derived from the same individual.  個体が、ステップ(3)で回収した遺伝子改変リンパ球の移植を受ける個体と別の個体である、請求項11に記載の調製方法。 The preparation method according to claim 11, wherein the individual is an individual different from the individual to be transplanted with the genetically modified lymphocyte collected in step (3).  ステップ(2)とステップ(3)の間に、培養後の細胞をT細胞増殖因子の存在下で培養するステップを行う、請求項1~12のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 1 to 12, wherein a step of culturing the cultured cells in the presence of a T cell growth factor is performed between steps (2) and (3).  T細胞増殖因子がIL-15である、請求項13に記載の調製方法。 The preparation method according to claim 13, wherein the T cell growth factor is IL-15.  T細胞増殖因子としてIL-15とIL-7を併用する、請求項13に記載の調製方法。 The preparation method according to claim 13, wherein IL-15 and IL-7 are used in combination as T cell growth factors.  ステップ(1)とステップ(2)の間に、以下のステップ(a)を行う、請求項1~15のいずれか一項に記載の調製方法:
 (a)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、抗CD3抗体及び抗CD28抗体で刺激しつつ共培養するステップ。
The preparation method according to any one of claims 1 to 15, wherein the following step (a) is performed between steps (1) and (2):
(A) Non-proliferative cells obtained by stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody and then performing a treatment for losing proliferative ability, and genetically modified lymphocytes prepared in step (1). A step of mixing lymphocytes and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody.
 ステップ(1)とステップ(2)の間に、以下のステップ(b)を行う、請求項1~15のいずれか一項に記載の調製方法:
 (b)末梢血単核細胞(PBMCs)をそのまま、又は抗CD3抗体及び抗CD28抗体で刺激した後、増殖能を喪失させる処理を行うことによって得られる、非増殖性PBMCsと、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ。
The preparation method according to any one of claims 1 to 15, wherein the following step (b) is performed between steps (1) and (2):
(B) Non-proliferative PBMCs obtained by treating peripheral blood mononuclear cells (PBMCs) as they are or by stimulating them with anti-CD3 antibody and anti-CD28 antibody and then performing a treatment to lose proliferative ability, and step (1). The step of mixing and co-culturing the genetically modified lymphocytes prepared in.
 ステップ(1)とステップ(2)の間に、以下のステップ(c)を行う、請求項1~15のいずれか一項に記載の調製方法:
 (c)T細胞を含む細胞集団を抗CD3抗体及び抗CD28抗体で刺激した後、ウイルスペプチド抗原存在下での培養及び増殖能を喪失させる処理を行うことによって得られる、ウイルスペプチド抗原を保持した非増殖性細胞と、ステップ(1)で用意した遺伝子改変リンパ球を混合し、共培養するステップ。
The preparation method according to any one of claims 1 to 15, wherein the following step (c) is performed between steps (1) and (2):
(C) A cell population containing T cells was stimulated with an anti-CD3 antibody and an anti-CD28 antibody, and then cultured in the presence of the viral peptide antigen and treated to lose the proliferative ability to retain the viral peptide antigen. A step of mixing non-proliferative cells with the genetically modified lymphocytes prepared in step (1) and co-culturing them.
 T細胞を含む細胞集団が末梢血単核細胞(PBMCs)である、請求項16又は18に記載の調製方法。 The preparation method according to claim 16 or 18, wherein the cell population containing T cells is peripheral blood mononuclear cells (PBMCs).  増殖能を喪失させる処理が放射線照射である、請求項16~19のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 16 to 19, wherein the treatment for losing the proliferative ability is irradiation.  非増殖性細胞と、遺伝子改変リンパ球が同一の個体に由来する、請求項16~20のいずれか一項に記載の調製方法。 The preparation method according to any one of claims 16 to 20, wherein the non-proliferative cells and the genetically modified lymphocytes are derived from the same individual.  請求項1~21のいずれか一項に記載の調製方法で得られた、キメラ抗原受容体を発現する遺伝子改変リンパ球。 A genetically modified lymphocyte expressing a chimeric antigen receptor obtained by the preparation method according to any one of claims 1 to 21.  請求項22に記載の遺伝子改変リンパ球を治療上有効量含む、細胞製剤。 A cell preparation containing a therapeutically effective amount of the genetically modified lymphocyte according to claim 22.  請求項22に記載の遺伝子改変リンパ球を、治療上有効量、がん患者に投与するステップを含む、がんの治療法。 A method for treating cancer, which comprises a step of administering the genetically modified lymphocyte according to claim 22 to a cancer patient in a therapeutically effective amount.
PCT/JP2020/042187 2019-11-20 2020-11-12 Method for producing chimeric antigen receptor gene-modified lymphocytes Ceased WO2021100585A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2021558324A JPWO2021100585A1 (en) 2019-11-20 2020-11-12

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2019209891 2019-11-20
JP2019-209891 2019-11-20

Publications (1)

Publication Number Publication Date
WO2021100585A1 true WO2021100585A1 (en) 2021-05-27

Family

ID=75981284

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2020/042187 Ceased WO2021100585A1 (en) 2019-11-20 2020-11-12 Method for producing chimeric antigen receptor gene-modified lymphocytes

Country Status (2)

Country Link
JP (1) JPWO2021100585A1 (en)
WO (1) WO2021100585A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2024024551A1 (en) * 2022-07-26 2024-02-01

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017061615A1 (en) * 2015-10-08 2017-04-13 国立大学法人名古屋大学 Method for preparing genetically-modified t cells which express chimeric antigen receptor
JP2017535284A (en) * 2014-11-05 2017-11-30 ボード・オブ・リージエンツ,ザ・ユニバーシテイ・オブ・テキサス・システム Genetically modified immune effector cells and genetically engineered cells for expansion of immune effector cells
JP2018516092A (en) * 2015-04-06 2018-06-21 サブドメイン リミテッド ライアビリティ カンパニー Novel binding domain-containing polypeptides and uses thereof
JP2018519842A (en) * 2015-07-21 2018-07-26 ノバルティス アーゲー Methods for improving the effectiveness and expansion of immune cells
WO2019027850A1 (en) * 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
JP2019527553A (en) * 2016-07-29 2019-10-03 ジュノー セラピューティクス インコーポレイテッド Anti-idiotype antibody against anti-CD19 antibody

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2017535284A (en) * 2014-11-05 2017-11-30 ボード・オブ・リージエンツ,ザ・ユニバーシテイ・オブ・テキサス・システム Genetically modified immune effector cells and genetically engineered cells for expansion of immune effector cells
JP2018516092A (en) * 2015-04-06 2018-06-21 サブドメイン リミテッド ライアビリティ カンパニー Novel binding domain-containing polypeptides and uses thereof
JP2018519842A (en) * 2015-07-21 2018-07-26 ノバルティス アーゲー Methods for improving the effectiveness and expansion of immune cells
WO2017061615A1 (en) * 2015-10-08 2017-04-13 国立大学法人名古屋大学 Method for preparing genetically-modified t cells which express chimeric antigen receptor
JP2019527553A (en) * 2016-07-29 2019-10-03 ジュノー セラピューティクス インコーポレイテッド Anti-idiotype antibody against anti-CD19 antibody
WO2019027850A1 (en) * 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2024024551A1 (en) * 2022-07-26 2024-02-01
WO2024024551A1 (en) * 2022-07-26 2024-02-01 国立大学法人京都大学 Artificial receptor having shedding structure

Also Published As

Publication number Publication date
JPWO2021100585A1 (en) 2021-05-27

Similar Documents

Publication Publication Date Title
JP6857360B2 (en) Method for preparing gene-modified T cells expressing chimeric antigen receptor
TWI823829B (en) CHIMERIC ANTIGEN RECEPTORS (CARs) TARGETING HEMATOLOGIC MALIGNANCIES, COMPOSITIONS AND METHODS OF USE THEREOF
AU2020204448A1 (en) Compositions and methods for immunotherapy
JP6853514B2 (en) Chimeric antigen receptor genetically modified lymphocytes that suppress the production of inflammatory cytokines
JP6971986B2 (en) Mesenchymal stem cells to enhance the antitumor activity of immunotherapy
WO2024199197A1 (en) Functionally enhanced engineered immune cell, and preparation therefor and use thereof
WO2022236142A2 (en) Chimeric receptors and methods of use thereof
JP7057975B2 (en) Chimeric antigen receptor gene-modified lymphocytes with cell-killing effect
US20240301352A1 (en) Method for producing car-t cells
WO2021100585A1 (en) Method for producing chimeric antigen receptor gene-modified lymphocytes
US20180369282A1 (en) Gene-modified lymphocytes expressing chimeric antigen receptor in which production of inflammatory cytokines is inhibited
CN115960257B (en) Optimized chimeric antigen receptor targeting IL13Rα2 and uses thereof
JP7576547B2 (en) Method for producing cell population containing CAR-expressing immune cells
JP7789342B2 (en) Method for preparing genetically modified T cells expressing chimeric antigen receptors
US20220184124A1 (en) Methods and reagents for characterizing car t cells for therapies
JP7531603B2 (en) CD28H domain-containing chimeric antigen receptors and methods of use
JP7075595B2 (en) Thioprine hypersensitive chimeric antigen receptor gene-modified lymphocytes
CN114269929B (en) Methods for producing cell populations containing CAR-expressing immune cells
WO2020085480A1 (en) High-efficiency method for producing genetically modified cells
JP2022171485A (en) Method for preparing genetically modified T cells expressing chimeric antigen receptors
RU2816370C2 (en) Rituximab-resistant chimeric antigen receptors and ways of use thereof
HK40104781A (en) Method for preparing genetically-modified t cells which express chimeric antigen receptor
WO2025085721A1 (en) Chimeric receptors and uses thereof
JP2023504196A (en) Placenta-derived allogeneic CAR-T cells and uses thereof
JPWO2020004337A1 (en) CD37 specific chimeric antigen receptor

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: 20889865

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021558324

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20889865

Country of ref document: EP

Kind code of ref document: A1