WO2017061615A1 - キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 - Google Patents
キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 Download PDFInfo
- Publication number
- WO2017061615A1 WO2017061615A1 PCT/JP2016/079989 JP2016079989W WO2017061615A1 WO 2017061615 A1 WO2017061615 A1 WO 2017061615A1 JP 2016079989 W JP2016079989 W JP 2016079989W WO 2017061615 A1 WO2017061615 A1 WO 2017061615A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- cell
- preparation
- antibody
- culture
- 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
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/11—T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/31—Chimeric antigen receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/4202—Receptors, cell surface antigens or cell surface determinants
- A61K40/421—Immunoglobulin superfamily
- A61K40/4211—CD19 or B4
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/46—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
- C07K14/01—DNA viruses
- C07K14/075—Adenoviridae
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70521—CD28, CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0646—Natural killers cells [NK], NKT cells
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
- C12N5/12—Fused cells, e.g. hybridomas
- C12N5/16—Animal cells
- C12N5/163—Animal cells one of the fusion partners being a B or a T lymphocyte
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/58—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
- A61K2039/585—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterized by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/38—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
- A61K2239/48—Blood cells, e.g. leukemia or lymphoma
Definitions
- the present invention relates to a method for preparing a genetically modified T cell expressing a chimeric antigen receptor and its use.
- CAR therapy Genetically modified T-cell therapy (CAR therapy) using a chimeric antigen receptor (Chimeric Antigen Receptor, hereinafter also referred to as “CAR”) is being clinically applied.
- CAR typically has a structure in which a single-chain variable region of an antibody is an extracellular domain, which is connected to a transmembrane region, CD3 ⁇ , and an intracellular domain of a molecule that transmits a costimulatory signal.
- CD3 ⁇ transmembrane region
- CD3 ⁇ transmembrane region
- CD3 ⁇ transmembrane region
- intracellular domain of a molecule that transmits a costimulatory signal By binding to an antigen according to the specificity of the antibody, CAR-T cells are activated and damage target cells (such as cancer cells).
- CAR therapy has advantages such as relatively easy cell preparation, high cytotoxic activity, and expected long-lasting effects, and is especially resistant to refractory and conventional therapies.
- CAR-T cells cells used for CAR therapy
- retroviruses have a high frequency of insertion mutations in proto-oncogenes (leukemia frequently occurs in gene therapy using hematopoietic stem cells), and safety is a problem.
- cell lines and fetal bovine serum are used for culturing in the conventional method, and long-term safety is a concern particularly in pediatric patients.
- 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).
- the use of the transposon method which is one of the gene modification techniques using non-viral vectors, is being studied.
- the transposon method enables permanent gene transfer in the same way as the virus vector method, the gene transfer efficiency is lower than that of the virus vector method, and it can be used for gene transfer operations (such as electroporation and its improved methods).
- gene transfer operations such as electroporation and its improved methods.
- An object of the present invention is to eliminate these problems and contribute to clinical application of CAR therapy and improvement of treatment results.
- the present inventors diligently studied to solve the above problems. As a result, gene transfer efficiency and survival rate / proliferation rate are improved and finally obtained by co-culturing T cells (gene-modified T cells) after gene transfer operation with separately prepared activated T cells. It was revealed that the number of CAR-T cells increased. On the other hand, efficient preparation of virus-specific CAR-T cells by the strategy of co-culturing T cells (gene-modified T cells) after gene transfer operation with activated T cells retaining viral peptides Successful. The following invention is mainly based on these results.
- a method for preparing a genetically modified T cell expressing a chimeric antigen receptor comprising the following steps (1) to (4): (1) A step of preparing a non-proliferative cell 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 the proliferation ability; (2) obtaining a genetically modified T cell into which a target antigen-specific chimeric antigen receptor gene has been introduced by a transposon method; (3) mixing the non-proliferating cells prepared in step (1) and the genetically modified T cells obtained in step (2) and co-culturing while stimulating with anti-CD3 antibody and anti-CD28 antibody; (4) A step of collecting the cultured cells.
- [2] The preparation method according to [1], wherein a step of culturing the cells after co-culture in the presence of a T cell growth factor is performed between step (3) and step (4).
- [3] The preparation method according to [1] or [2], wherein the co-culture period in step (3) is 1 day to 14 days.
- [4] The preparation method according to any one of [1] to [3], wherein step (3) is performed in the presence of a T cell growth factor.
- the preparation method according to [4], wherein the T cell growth factor is IL-15.
- the preparation method according to [4], wherein IL-15 and IL-7 are used in combination as T cell growth factors.
- a method for preparing a genetically modified T cell expressing a chimeric antigen receptor comprising the following steps (i) to (iv): (I) After stimulating a cell population containing T cells with an anti-CD3 antibody and an anti-CD28 antibody, the virus peptide antigen obtained by culturing in the presence of the virus peptide antigen and treatment for losing the proliferation ability was retained.
- non-proliferating cells Providing non-proliferating cells; (Ii) obtaining a genetically modified T cell into which a target antigen-specific chimeric antigen receptor gene has been introduced by a transposon method; (Iii) mixing the non-proliferating cells prepared in step (i) with the genetically modified T cells obtained in step (ii) and co-culturing; (Iv) A step of collecting the cultured cells.
- the preparation method according to [7] wherein a step of culturing the co-cultured cells in the presence of a T cell growth factor is performed between step (iii) and step (iv).
- the preparation method according to [7] or [8], wherein the co-culture period in step (iii) is 1 day to 14 days.
- step (iii) is performed in the presence of a T cell growth factor.
- PBMCs peripheral blood mononuclear cells
- [15] The preparation method according to any one of [1] to [14], wherein the transposon method is a PiggyBac transposon method.
- a genetically modified T cell that expresses a chimeric antigen receptor obtained by the preparation method according to any one of [1] to [17].
- a cell preparation comprising a therapeutically effective amount of the genetically modified T cell according to [18].
- a method for treating cancer comprising administering a therapeutically effective amount of the genetically modified T cell according to [18] to a cancer patient.
- Preparation and culture of CAR-T cells by conventional methods (culture method 1).
- Preparation and culture of CAR-T cells by a new method (culture method 2).
- Preparation and culture of CAR-T cells by a new method (culture method 3).
- Construction of pIRII-CAR.CD19.28z vector (SEQ ID NO: 1).
- the CD19CAR gene has a structure sandwiched between a 5 ′ inverted repeat (5′IR) and a 3 ′ inverted repeat (3′IR).
- CD19CAR consists of 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), It contains the transmembrane region and intracellular domain of CD28 (SEQ ID NO: 6) and CD3 ⁇ (SEQ ID NO: 7).
- Construction of pCMV-pigBac vector SEQ ID NO: 8).
- the piggyBac transposase gene is located under the control of the CMV early promoter (CMV immediate ear1y promoter). Number of CAR-T cells obtained by each culture method (mean ⁇ standard error).
- the Fc region (CH2, CH3) is deleted.
- the present invention relates to a method for preparing a genetically modified T cell (CAR-T cell) expressing a chimeric antigen receptor.
- CAR-T cells obtained by the preparation method of the present invention can be used for CAR therapy.
- the present invention is broadly classified into two types of preparation methods, that is, a method including co-culture with activated T cells (sometimes referred to as “first preparation method” for convenience of explanation) and a viral peptide.
- a method comprising co-culturing with activated T cells (sometimes referred to as “second preparation method” for convenience of explanation) is provided.
- various cells for example, T cells in this specification are human cells.
- Step (1) A step of preparing a non-proliferative cell 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.
- Target antigen by transposon method Step of obtaining genetically modified T cells into which a specific chimeric antigen receptor gene has been introduced
- the non-proliferative cells prepared in step (1) and the genetically modified T cells obtained in step (2) are mixed together to produce anti-CD3 Step of co-culture while stimulating with antibody and anti-CD28 antibody (4) Step of collecting cells after culture
- Step (1) is a step of obtaining non-proliferating cells used for protecting T cells after the gene transfer operation (gene-modified T cells used in step (2)). Stimulate with CD3 antibody and anti-CD28 antibody. This treatment yields activated T cells.
- PBMC peripheral blood mononuclear cells
- PBMCs purified and T cell content increased, or mononuclear cells collected from peripheral blood by pheresis can be used as the “cell population containing T cells”.
- a culture vessel eg, a culture dish coated with a culture surface with anti-CD3 antibody and anti-CD28 antibody.
- Stimulation with anti-CD3 and anti-CD28 antibodies can be applied to T cells in the cell population.
- Anti-CD3 antibodies for example, the trade name CD3pure antibody provided by Miltenyi Biotech can be used
- anti-CD28 antibodies for example, the trade name CD28pure antibody provided by Miltenyi Biotech can be used
- step (1) it is also possible to perform the stimulation of step (1) using magnetic beads coated with anti-CD3 antibody and anti-CD28 antibody (for example, Dynabeads T-Activator CD3 / CD28 provided by VERITAS).
- anti-CD3 antibody and anti-CD28 antibody for example, Dynabeads T-Activator CD3 / CD28 provided by VERITAS.
- the “OKT3” clone is preferably used as the anti-CD3 antibody.
- Cells that have been stimulated with anti-CD3 antibody and anti-CD28 antibody are subjected to a treatment for losing their proliferative ability, but before that, they may be cultured in the presence of T cell growth factor.
- This culture increases 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, 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, costimulatory molecules may be attenuated.
- the cultured cells may be once stored frozen. In this case, the cells are thawed at the time of use, and after being stimulated again with the anti-CD3 antibody and CD28 antibody (conditions are the same as described above), the cells may be subjected to “treatment for loss of proliferation ability”.
- Activating T cells that have lost their proliferative ability can be obtained by undergoing a “treatment to lose their proliferative ability”.
- the treatment for losing the proliferation ability is typically irradiation, but a drug may be used.
- An example of the irradiation conditions is a treatment for 15 to 30 minutes using gamma rays at an intensity of 25 Gy to 50 Gy.
- step (2) genetically modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced are obtained by the transposon method. That is, in the present invention, the CAR gene is introduced into T cells using the transposon method to obtain CAR-T cells.
- the transposon method is one of non-viral gene transfer methods.
- a transposon is a generic term for a short gene sequence that causes gene rearrangement, which has been conserved during evolution. Gene transposition is caused by a pair of gene enzyme (transposase) and its specific recognition sequence.
- the transposon method for example, the PiggyBac transposon method can be used.
- the PiggyBac transposon method uses a transposon isolated from an insect (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.), Enabling highly efficient integration into mammalian chromosomes.
- the PiggyBac transposon method is actually used for the introduction of CAR genes (eg 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.
- the introduction operation by the transposon method may be performed by a conventional method.
- a conventional method for example, in the 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, or Saha S, Nakazawa Y, Huee LE, Doherty JE, Galvan DL, Rooney CM, Wilson MH. J Vis Exp. 2012 Nov 5; (69): e4235).
- the PiggyBac transposon method is employed.
- a vector holding a gene encoding a PiggyBac transposase (transposase plasmid) and a vector having a structure in which a gene encoding a target protein (CAR gene) is sandwiched between inverted repeats of piggyBac (Transposon plasmid) is prepared, and these two vectors are introduced (transfected) into target cells.
- various techniques such as electroporation, nucleofection, lipofection, and calcium phosphate method can be used.
- Examples of cells (target 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, ⁇ -T cells, and ⁇ -T cells.
- PBMC peripheral blood mononuclear cells collected from peripheral blood is one of the preferred target cells. That is, in a preferred embodiment, a gene introduction operation is performed on PBMC.
- PBMC may be prepared by a conventional method. For the preparation method of PBMC, see, 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.
- T cell growth factor eg, IL-15 or IL-7. You may decide to culture in presence.
- the CAR gene encodes a chimeric antigen receptor (CAR) that recognizes a specific target antigen.
- CAR is a structure containing a target-specific extracellular domain, a transmembrane domain, and an intracellular signal domain for immune cell effector functions. Hereinafter, each domain will be described.
- Extracellular domain exhibits specific binding to the target.
- the extracellular domain includes an scFv fragment of an anti-target monoclonal antibody.
- the monoclonal antibody used here include rodent (mouse, rat, rabbit, etc.) antibodies, human antibodies, humanized antibodies, and the like.
- a humanized monoclonal antibody is an antibody in which the structure of a monoclonal antibody of another animal species (eg, 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.
- a chimeric antibody and a human-type CDR-grafted antibody (PTJohons et al., Nature 321,522 (1986) in which parts other than CDRs (complementarity determining regions) present in constant regions and variable regions are substituted with those of human antibodies. ))including.
- a method for selecting a human antibody framework (FR) highly homologous to a mouse antibody a method for producing a humanized antibody having a high homology, a mouse CDR for a human antibody
- Techniques for further substitution of amino acids in the FR region after transplantation have already been developed (US Patent No. 5585089, US Patent No. 5693761, US Patent No. 5693762, US Patent No. 6180370, European Patent No. 451216) , European Patent No. 682040, Patent No. 2828340, etc.) and can also be used for the production of humanized antibodies.
- An scFv fragment is a structure in which an immunoglobulin light chain variable region (VL) and heavy chain variable region (VH) 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 consisting of a peptide in which amino acids are linked in a straight chain.
- a typical example of a peptide linker is a linker composed of glycine and serine (GGS linker or GS linker).
- the GGS linker and glycine and serine, which are amino acids constituting the GS linker have a small size per se and are 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 length of the linker is preferably 8 to 25, more preferably 15 to 20.
- target antigens include CD19 antigen, CD20 antigen, GD2 antigen, CD22 antigen, CD30 antigen, CD33 antigen, CD44 variant7 / 8 antigen, CEA antigen, Her2 / neu antigen, MUC1 antigen, MUC4 antigen, MUC6 antigen, IL-13 receptor -alpha2, immunoglobulin light chain, anti-PSMA, VEGF receptor 2 etc.
- GM-CSF granulocyte monocyte colony stimulating factor receptor expressed in leukemic stem cells / progenitor cells
- GM-CSF which is a ligand of the GM-CSF receptor
- Myeloid tumor leukemia stem cells, leukemia progenitor cells, leukemia cells, etc. are targets of CAR-T cells, and myeloproliferative tumors, myelodysplastic / myeloproliferative tumors (CMML, JMML, CML, MDS / MPN-UC) ), Cells applicable to prevention and treatment of myelodysplastic syndrome, acute myeloid leukemia, etc. are prepared.
- CMML myelodysplastic / myeloproliferative tumors
- transmembrane domain The transmembrane domain is interposed 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. You may decide to use the transmembrane domain which consists of the polypeptide constructed artificially.
- the intracellular signal domain transmits a signal necessary for exerting an effector function of an immune cell. That is, an intracellular signal domain that can transmit a signal necessary for activation of immune cells when an extracellular domain binds to a target antigen is used.
- the intracellular signal domain includes a domain for transmitting a signal via the TCR complex (for convenience, referred to as “first domain”) and a domain for transmitting a costimulatory signal (for convenience, “second domain”). Called).
- an intracellular domain such as Fc ⁇ RI ⁇ can be used as the first domain.
- CD3 ⁇ is preferably used.
- an intracellular domain of a costimulatory molecule is used as the second domain.
- costimulatory molecules include CD28, 4-1BB (CD137), CD2, CD4, CD5, CD134, OX-40, or ICOS.
- the intracellular domain of CD28 or 4-1BB is employed.
- connection mode of the first domain and the second domain is not particularly limited, but preferably, it is known that costimulation was strongly transmitted when CD3 ⁇ was connected distally in the past case.
- the second domain is arranged on the side. A plurality of identical or different intracellular domains may be linked in tandem to form the first domain. The same applies to the second domain.
- the first domain and the second domain may be linked directly or a linker may be interposed between them.
- a linker for example, a peptide linker can be used.
- a peptide linker is a linker consisting of a peptide in which amino acids are linked in a straight chain. The structure, characteristics, etc. of the peptide linker are as described above. However, as a linker here, you may use what consists only of glycine.
- the length of the linker is not particularly limited. For example, a linker having 2 to 15 amino acid residues can be used.
- a leader sequence (signal peptide) is used to promote secretion of CAR.
- the leader sequence of GM-CSF receptor can be used.
- the extracellular domain and the transmembrane domain may be linked via a spacer domain.
- the spacer domain is used to promote binding between CAR and the target antigen.
- an Fc fragment of human IgG eg, human IgG1, human IgG4
- a part of the extracellular domain of CD28, a part of the extracellular domain of CD8 ⁇ , and the like can be used as the spacer domain.
- a spacer domain can also be provided between the transmembrane domain and the intracellular signal domain.
- a poly A addition signal sequence is placed downstream of the CAR gene. Transcription is terminated by the use of a poly A addition signal sequence.
- a poly A addition signal sequence SV40 poly A addition sequence, bovine-derived growth hormone gene poly A addition sequence, and the like can be used.
- the transposon plasmid may contain a detection gene (reporter gene, cell or tissue-specific gene, selection marker gene, etc.), enhancer sequence, WRPE sequence, and the like.
- the detection gene is used for determining the success or failure of the introduction of the expression cassette and determining the efficiency, detecting the CAR gene expression or determining the expression efficiency, selecting and sorting the cells in which the CAR gene is expressed, and the like.
- the use of an enhancer sequence can improve the expression efficiency.
- detection genes the neo gene conferring resistance to neomycin, the npt gene conferring resistance to kanamycin and the like (Herrera Estrella, EMBO J.
- nptII gene (Messing & Vierra. Gene 1) 9: 259-268 (1982)), hph gene conferring resistance to hygromycin (Blochinger & Diggl mann, Mol Cell Bio 4: 2929-2931), dhfr gene conferring resistance to metatrexate (Bourouis et al. , EMBO J.2 (7)) etc. (above, marker gene), luciferase gene (Giacomin, P1. Sci. 116 (1996), 59-72; Scikantha, J. Bact.
- GUS ⁇ -Glucuronidase
- fluorescent protein gene such as reporter gene
- intracellular domain such as GFP (Gerdes, FEBS Lett. 389 (1996), 44-47) and its variants (EGFP, d2EGFP, etc.)
- EGFR epidermal growth factor receptor
- the detection gene is linked to the CAR gene via, for example, a bicistronic regulatory sequence (for example, a ribosome internal recognition sequence (IRES)) or a sequence encoding a self-cleaving peptide.
- a bicistronic regulatory sequence for example, a ribosome internal recognition sequence (IRES)
- IRS ribosome internal recognition sequence
- a self-cleaving peptide is the 2A peptide (T2A) derived from Thosea asigna virus, but is not limited thereto.
- Known as self-cleaving peptides are 2A peptide (F2A) from horseshoe disease virus (FMDV), 2A peptide (E2A) from equine rhinitis A virus (ERAV), 2A peptide (P2A) from porcine teschovirus (PTV-1), etc. It has been.
- step (3) the non-proliferating cells prepared in step (1) and the genetically modified T cells obtained in step (2) are mixed and co-cultured while stimulating with anti-CD3 antibody and anti-CD28 antibody.
- stimulation through costimulatory molecules by non-proliferating cells and stimulation by anti-CD3 antibody and anti-CD28 antibody are added, and genetically modified T cells are activated and their survival and proliferation are promoted.
- the ratio of the number of non-proliferating cells and the number of genetically modified T cells used for co-culture is not particularly limited, but is, for example, 0.025 to 0.5.
- a culture solution to which T cell growth factor is added during co-culture IL-15 is preferred as the T cell growth factor.
- IL-15 is preferred as the T cell growth factor.
- 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.
- T cell growth factors such as IL-15 and IL-7 can be prepared according to conventional methods. Moreover, a commercial item can also be utilized.
- T cell growth factors of animal species other than humans are not excluded, usually, T cell growth factors are derived from humans (may be recombinant). Growth factors such as human IL-15 and human IL-7 can be obtained in advance (for example, provided by Miltenyi Biotech, R & D Systems, etc.).
- a medium supplemented with serum (human serum, fetal bovine serum, etc.) may be used, but by using a serum-free medium, the safety in clinical application is high and the culture efficiency due to the difference between serum lots is improved. It becomes possible to prepare cells having the advantage that differences are difficult to make.
- Specific examples of the serum-free medium for T cells are TexMACS TM (Miltenyi Biotech) and AIM V (registered trademark) (Thermo Fisher Scientific).
- serum autologous serum, that is, an individual derived from the genetically modified T cell obtained in step (2) (typically the chimeric antigen receptor genetically modified T cell obtained by the preparation method of the present invention). It is recommended to use serum collected from the patient.
- a medium suitable for T cell culture may be used, and specific examples include TexMACS TM and AIM V (registered disease trademark) described above.
- Other culture conditions may be any suitable culture conditions as long as they are suitable for the survival and proliferation of T cells. For example, it may be cultured in a CO 2 incubator set at 37 ° C. (CO 2 concentration 5%).
- CO 2 concentration 5% the stimulation by the anti-CD3 antibody and the anti-CD28 antibody is the same as that in the step (1), the description thereof is omitted.
- the period of co-culture in step (3) is, for example, 1 to 10 days, preferably 1 to 7 days, 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, there is a risk of cell activity (life force) being reduced.
- step (4) following step (3) the cultured cells are collected.
- the collection operation may be performed by a conventional method. For example, it collect
- a step of culturing the co-cultured cells in the presence of T cell growth factor is performed between step (3) and step (4). This step enables efficient expansion culture and 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.
- the culture period is, for example, 1 to 21 days, preferably 5 to 18 days, 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 cell activity (life force) decrease, cell exhaustion / fatigue, and the like. You may subculture during culture
- the medium is changed as necessary. For example, about 1/3 to 2/3 of the culture solution is replaced with a new medium once every three days.
- virus-specific CAR ⁇ virus-specific chimeric antigen receptor gene-modified T cell
- T cells virus-specific CAR ⁇
- Virus-specific CAR-T cells can be expected to improve persistence in the body by stimulation from viral T cell receptors when used for autologous transplantation, and further allogeneic immune response (GVHD) when used for allogeneic transplantation This reduction has an important advantage in clinical applications such as the possibility of preparing CAR-T from a transplant donor and the possibility of formulating CAR-T cells from a third-party donor.
- the preparation method of this embodiment includes the following steps (i) to (iv).
- matters that are not mentioned for example, a method for preparing a cell population containing T cells, a basic operation for stimulation with an anti-CD3 antibody and an anti-CD28 antibody, a method for treatment for losing proliferation ability, an operation for gene introduction by a transposon method, Since the basic operation of co-culture, the cell recovery method, and the like are the same as those in the first preparation method described above, the overlapping description is omitted and the corresponding description is used.
- Step of preparing non-proliferating cells Step of preparing non-proliferating cells (ii) Step of obtaining genetically modified T cells into which a target antigen-specific chimeric antigen receptor gene has been introduced by transposon method (iii) Non-proliferating cells prepared in step (i) Mixing and co-culturing the genetically modified T cells obtained in step (ii) (iv) recovering the cultured cells
- step (i) first, a cell population containing T cells is stimulated with anti-CD3 antibody and anti-CD28 antibody to obtain activated T cells. Then, the culture
- the order of the culture in the presence of the viral peptide antigen and the treatment for losing the growth ability is not particularly limited.
- the growth ability may be lost after culturing in the presence of the viral peptide antigen, or the culture may be performed in the presence of the viral peptide antigen after the growth ability is lost.
- the former order is employed because of the expectation that viral peptide antigen uptake will be better before loss of growth ability.
- a medium supplemented with a viral peptide antigen may be used.
- a viral peptide antigen may be added to the medium during culture. 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 that can induce cytotoxic T cells (CTL) specific for a specific virus or a long peptide containing an epitope.
- viral peptide antigens include, but are not limited to, adenoviral (AdV) antigenic peptides (see, for example, WO 2007015540 A1), cytomegalovirus (CMV) antigenic peptides (see, for example, JP2002 -255997, JP-A-2004-242599, JP-A-2012-87126), Epstein-Barr virus (EBV) antigen peptide (for example, WO2007049737A1, Japanese Patent Application No. 2011-177487, JP 2006-188513), etc. can be used.
- AdV adenoviral
- CMV cytomegalovirus
- EBV Epstein-Barr virus
- Viral peptide antigens can be prepared by conventional methods (eg, liquid phase synthesis method, solid phase synthesis method) based on sequence information. Some viral peptide antigens are commercially available (for example, provided by Medical Biology Laboratory, Takara Bio, Miltenyi Biotech, 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.
- two or more types of antigen peptides in combination a plurality of activated T cells with different targets (antigen peptides) can be obtained, and CAR-T cells obtained by the preparation method of the present invention are effective treatment subjects (patients) ) (Increased coverage).
- an antigen peptide mixture of EBV virus may be used alone or in combination with an antigen peptide mixture of another virus.
- AdV antigen peptide mixture 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 (provided by Miltenyi Biotech) (Registered trademark) 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.
- Step (ii) is the same as step (2) of the first preparation method of the present invention. This step yields genetically modified T cells (CAR-T cells).
- CAR-T cells genetically modified T cells
- step (iii) the non-proliferating cells (viral peptide-retaining non-proliferating cells) prepared in step (i) and the genetically modified T cells obtained in step (ii) are mixed and co-cultured.
- stimulation through costimulatory molecules and virus antigen peptides by non-proliferating cells is added, and virus antigen-specific genetically modified T cells are activated and their survival and proliferation are promoted.
- the ratio of the number of non-proliferating cells and the number of genetically modified T cells used for co-culture is not particularly limited, but is, for example, 0.025 to 0.5.
- This step is mainly for anti-CD3 antibody and anti-CD28 antibody for the purpose of selectively expanding virus-specific CAR-T cells, avoiding strong stimulation and preventing T cell exhaustion / fatigue, etc. Do not apply irritation.
- a culture solution to which T cell growth factor is added during co-culture IL-15 is preferred as the T cell growth factor.
- IL-7 is preferred as the T cell growth factor.
- 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.
- the conditions not mentioned are the same as in step (3) of the first preparation method of the present invention.
- Viral peptide-retaining non-proliferating cells may be added during step (iii). Alternatively, the cells after the co-culture may be collected and mixed with the virus peptide-retaining nonproliferative cells, and then co-culture may be performed again. These operations may be repeated twice or more. Thus, if stimulation is performed multiple times without activation using viral peptide-retaining nonproliferative cells, the induction rate of virus-specific CAR-T cells is improved, and the number of virus-specific CAR-T cells is increased. We can expect increase. In addition, what was prepared anew or what preserve
- the period of co-culture in step (iii) 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 cell activity (life force) decrease, cell exhaustion / fatigue, and the like.
- the genetically modified T cells obtained in step (ii) may be co-cultured with viral peptide-retaining non-proliferating PBMC (peripheral blood mononuclear cells).
- PBMC peripheral blood mononuclear cells
- a cell obtained by co-culturing the genetically modified T cell obtained in step (ii) and the non-proliferative PBMC retaining virus peptide, and the non-proliferating cell retaining virus peptide prepared in step (i) Are mixed and co-cultured.
- the viral peptide-retaining non-proliferative PBMC here can be prepared by subjecting PBMC to culture in the presence of a viral peptide antigen and a treatment to lose the proliferation ability. Specifically, for example, PBMCs isolated from peripheral blood are treated with radiation and then cultured in the presence of viral peptide antigens to obtain viral peptide-retaining nonproliferative PBMCs. In addition, while preparing a viral peptide-retaining nonproliferative PBMC using a part of PBMC isolated from peripheral blood obtained by one blood collection, and preparing a genetically modified T cell from the other part, The number of blood collections associated with the implementation of the present invention can be reduced, which is a great advantage in clinical application.
- step (i) is performed using the remaining PBMC to prepare non-proliferating cells that retain viral peptides (cells used for the second stage of co-culture)
- the necessary three types of cells A genetically modified T cell, a non-proliferative PBMC retaining viral peptide used for co-culture with the cell, and a non-proliferating cell retaining viral peptide used for the second stage co-culture can be prepared by a single blood collection. Therefore, the burden on the patient in the treatment using the CAR-T cells obtained in the present invention is greatly reduced.
- step (iv) following step (iii) the cultured cells are collected.
- a step (expansion culture) of culturing the co-cultured cells in the presence of T cell growth factor is performed between step (iii) and step (iv). It may be.
- Viral peptide-retaining non-proliferating cells may be added during expansion culture, or viral peptide-retaining non-proliferating cells may be added during expansion culture.
- a further aspect of the present invention provides a genetically modified T cell expressing a chimeric antigen receptor (hereinafter referred to as “the present invention”) obtained by the preparation method of the present invention. Called “Car-T cells”) and their uses.
- the CAR-T cells of the present invention can be used for the treatment, prevention or amelioration of various diseases for which CAR therapy is considered effective (hereinafter referred to as “target diseases”).
- a representative target disease is cancer, but is not limited thereto.
- target diseases include various B-cell lymphomas (such as follicular malignant lymphoma, diffuse malignant lymphoma, mantle cell lymphoma, MALT lymphoma, intravascular B-cell lymphoma, CD20 positive Hodgkin lymphoma), myeloproliferative tumors, bone marrow Dysplasia / myeloproliferative tumor (CMML, JMML, CML, MDS / MPN-UC), myelodysplastic syndrome, acute myeloblastic leukemia, neuroblastoma, brain tumor, Ewing sarcoma, osteosarcoma, retinoblastoma, small lung Cellular tumor, melanoma, ovarian cancer, rhabdomyosarcoma, kidney cancer, pancreatic cancer, malignant mesothelioma, prostate cancer and the like.
- B-cell lymphomas such as follicular malignant lymphoma, diffuse malignant lymphoma, mantle
- Treatment includes alleviation of symptoms or concomitant symptoms characteristic of the target disease (mildness), prevention or delay of deterioration of symptoms, and the like. “Prevention” refers to preventing or delaying the onset / onset of a disease (disorder) or its symptoms, or reducing the risk of onset / onset. On the other hand, “improvement” means that a disease (disorder) or a symptom thereof is alleviated (lightened), improved, ameliorated, or cured (including partial healing).
- the CAR-T cells of the present invention can also be provided in the form of cell preparations.
- the cell preparation of the present invention contains a therapeutically effective amount of the CAR-T cell of the present invention.
- a therapeutically effective amount of the CAR-T cell of the present invention for example, for a single administration, 10 4 to 10 10 cells are contained.
- Various components (vitamins) for the purpose of cell activation, proliferation or differentiation induction, such as dimethyl sulfoxide (DMSO) and serum albumin for the purpose of cell protection, such as antibiotics for the purpose of blocking bacterial contamination , Cytokines, growth factors, steroids, etc.) may be included in the cell preparation.
- the administration route of the CAR-T cell or cell preparation of the present invention is not particularly limited.
- administration is by intravenous injection, intraarterial injection, intraportal injection, intradermal injection, subcutaneous injection, intramuscular injection, or intraperitoneal injection.
- Local administration may be used instead of systemic administration. Examples of the local administration include direct injection into a target tissue / organ / organ.
- the administration schedule may be prepared in consideration of the subject's (patient) sex, age, weight, disease state, and the like. In addition to single administration, multiple administration may be performed continuously or periodically.
- CAR therapy using the transposon method is particularly advantageous in terms of safety compared to the case of using a viral vector.
- problems such as low gene transfer efficiency, easy cell damage due to the operation during gene transfer (for example, electroporation), and a small number of cells obtained. In order to overcome these problems, the following examination was conducted.
- 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, Add 0.5 ml / well to a 24-well uncoated plate. Leave the plate in a 37 ° C incubator for 2-4 hours. Aspirate antibody-diluted PBS and wash once with 1 ml PBS per well.
- Day 1 Stimulate with anti-CD3 / anti-CD28 antibody coated plate.
- Day 4 Cells are transferred to G-Rex10 and cultured in TexMACS TM supplemented with IL-7 10 ng / ml and IL15 5 ng / ml.
- Day 7 Half of the culture medium is replaced with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 10 Half of the culture is exchanged with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 14 End culture.
- Day 1 Stimulate with anti-CD3 / anti-CD28 antibody coated plate.
- Day 4 Transfer cells to G-Rex10 and culture with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 7 Half of the culture medium is replaced with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 10 Half of the culture is exchanged with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 14 End culture.
- Transfected cells and activated virus cells supplemented with irradiated virus peptide are mixed, floated on TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml, and cultured in a 24-well plate in a 37 ° C incubator To start. Day 2 to Day 5: If necessary, exchange half of the culture medium with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml. Day 7: Cells are collected and counted.
- Virus peptide-added activated T cells 2x10 6 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 solution, Start culture with G-Rex10.
- Day 10 Half of the culture is exchanged with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- Day 14 End culture.
- the number of CAR-T cells is 1.18 ⁇ 10 6 ( ⁇ 0.509 ⁇ 10 6 ) in culture method 1, 7.13 ⁇ 10 6 ( ⁇ 3.25 ⁇ 10 6 ) in culture method 2, and 1.09 ⁇ 10 7 ( ⁇ 2.98) in culture method 3. ⁇ 10 6 ) (FIG. 6).
- the gene transfer efficiency was 3.22% ( ⁇ 1.42) for culture method 1, 10.3% ( ⁇ 4.21) for culture method 2, and 26.9% ( ⁇ 2.79) for culture method 3 (FIG. 7).
- the parentheses are standard errors.
- the number of CAR-T cells increased and the gene transfer efficiency also increased.
- the number of CAR-T cells increases due to cell stimulation by the expression of costimulatory molecules (protective effect of transgenic cells damaged during electroporation) and cytokine stimulation by the culture microenvironment. It is thought that an increase in gene transfer efficiency was brought about.
- culture method 3 the number of CAR-T cells and gene transfer efficiency are increased by cell stimulation by expression of costimulatory molecules, cytokine stimulation by culture microenvironment, and relatively gentle cell stimulation from virus-specific T cell receptors. It is thought that the rise of
- Method Day 0 Isolate mononuclear cells (PBMC) 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 50ng each) and incubate at 37 ° C for 30 minutes.
- pIRII-CAR.CD19_optimized vector FIG. 10
- pCMV-pigBac vector FIG.
- virus peptides (PepTivator CMV pp65, PepTivator AdV5 Hexon, PepTivator EBV EBNA-1 and PepTivator EBV BZLF1 50ng each) were added, 37 Incubate at 30 ° C for 30 minutes.
- the virus peptide-added activated T cells 2x10 6 thus obtained and the recovered cells were suspended in 30 ml of a culture solution supplemented with IL-15 (5 ng / ml) and IL-7 (10 ng / ml), and G -Start culture at Rex10.
- Day 10 Half of the culture is exchanged with TexMACS TM supplemented with IL-7 10ng / ml and IL15 5ng / ml.
- CAR-T cells prepared by the novel culture method were evaluated by the following methods.
- CAR-T cells (1 ⁇ 10 5 cells) and CD19 positive leukemia cell lines (5 ⁇ 10 5 cells) were co-cultured in RPMI1640 medium containing 10% fetal calf serum for 7 days (effector cells: target cells 1: 5 ).
- mice treated with expanded CAR-T cells in contrast to tumor cells grown in mice treated with non-transgenic activated T cells (control (NT)). No tumor cell growth was observed. That is, CAT cells strongly suppressed tumor growth in tumor-bearing mice.
- CAR gene introduction efficiency and cell proliferation rate when CAR-T cells are prepared using the transposon method can be increased. That is, the present invention enhances the practicality of a highly safe method of preparing CAR-T cells using the transposon method, thereby promoting the clinical application of CAR therapy and contributing to the increase in the therapeutic effect of CAR therapy.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Immunology (AREA)
- Cell Biology (AREA)
- General Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Epidemiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Hematology (AREA)
- Toxicology (AREA)
- Virology (AREA)
- Pharmacology & Pharmacy (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Developmental Biology & Embryology (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
[1]以下のステップ(1)~(4)を含む、キメラ抗原受容体を発現する遺伝子改変T細胞の調製方法:
(1)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を用意するステップ;
(2)トランスポゾン法によって、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T細胞を得るステップ;
(3)ステップ(1)で用意した非増殖性細胞とステップ(2)で得た遺伝子改変T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ステップ;
(4)培養後の細胞を回収するステップ。
[2]ステップ(3)とステップ(4)の間に、共培養後の細胞をT細胞増殖因子の存在下で培養するステップを行う、[1]に記載の調製方法。
[3]ステップ(3)の共培養の期間が1日~14日である、[1]又は[2]に記載の調製方法。
[4]ステップ(3)を、T細胞増殖因子の存在下で行う、[1]~[3]のいずれか一項に記載の調製方法。
[5]T細胞増殖因子がIL-15である、[4]に記載の調製方法。
[6]T細胞増殖因子としてIL-15とIL-7を併用する、[4]に記載の調製方法。
[7]以下のステップ(i)~(iv)を含む、キメラ抗原受容体を発現する遺伝子改変T細胞の調製方法:
(i)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を用意するステップ;
(ii)トランスポゾン法によって、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T細胞を得るステップ;
(iii)ステップ(i)で用意した非増殖性細胞とステップ(ii)で得た遺伝子改変T細胞を混合し、共培養するステップ;
(iv)培養後の細胞を回収するステップ。
[8]ステップ(iii)とステップ(iv)の間に、共培養後の細胞をT細胞増殖因子の存在下で培養するステップを行う、[7]に記載の調製方法。
[9]ステップ(iii)の共培養の期間が1日~14日である、[7]又は[8]に記載の調製方法。
[10]ステップ(iii)を、T細胞増殖因子の存在下で行う、[7]~[9]のいずれか一項に記載の調製方法。
[11]T細胞増殖因子がIL-15である、[10]に記載の調製方法。
[12]T細胞増殖因子としてIL-15とIL-7を併用する、[10]に記載の調製方法。
[13]T細胞を含む細胞集団が末梢血単核細胞(PBMCs)である、[1]~[12]のいずれか一項に記載の調製方法。
[14]増殖能を喪失させる処理が放射線照射である、[1]~[13]のいずれか一項に記載の調製方法。
[15]トランスポゾン法がPiggyBacトランスポゾン法である、[1]~[14]のいずれか一項に記載の調製方法。
[16]標的抗原がCD19、GD2、GMCSF受容体又はIGF受容体である、[1]~[15]のいずれか一項に記載の調製方法。
[17]非増殖性細胞と、遺伝子改変T細胞が同一の個体に由来する、[1]~[16]のいずれか一項に記載の調製方法。
[18][1]~[17]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T細胞。
[19][18]に記載の遺伝子改変T細胞を治療上有効量含む、細胞製剤。
[20][18]に記載の遺伝子改変T細胞を、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。
この調製方法(第1の調製方法)では、以下のステップ(1)~(4)を行う。
(1)T細胞含む細胞集団を抗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を用意するステップ
(2)トランスポゾン法によって、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T細胞を得るステップ
(3)ステップ(1)で用意した非増殖性細胞とステップ(2)で得た遺伝子改変T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ステップ
(4)培養後の細胞を回収するステップ
細胞外ドメインは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。
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。
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。
CARの分泌を促すために、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場合がある。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。
本発明の別の態様(第2の調製方法)は、ウイルス特異的なキメラ抗原受容体遺伝子改変T細胞(以下、「ウイルス特異的CAR-T細胞」と呼ぶ)を調製する方法に関する。ウイルス特異的CAR-T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-T作製が可能になり、しかも第3者ドナーからのCAR-T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-T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。
(i)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を用意するステップ
(ii)トランスポゾン法によって、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T細胞を得るステップ
(iii)ステップ(i)で用意した非増殖性細胞とステップ(ii)で得た遺伝子改変T細胞を混合し、共培養するステップ
(iv)培養後の細胞を回収するステップ
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T細胞(以下、「本発明のCAR-T細胞」と呼ぶ)及びその用途に関する。本発明のCAR-T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。
トランスポゾン法を利用したCAR療法は、ウイルスベクターを利用した場合に比べ、特に安全性の面で有利である。その一方で、遺伝子導入効率が低いこと、遺伝子導入時の操作(例えばエレクトロポレーション)で細胞がダメージを受けやすく、得られる細胞数が少ないことなどが問題となる。これらの問題点を克服すべく、以下の検討を行った。
(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)
(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)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。
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TexMACS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。
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。
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5μg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。
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5μg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。
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5μg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。
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。
培養終了後(14日目)、培養法毎に増殖細胞数、CAR-T細胞数及び遺伝子導入効率(CAR-T細胞/全生細胞)を求め、培養法1~3の間で比較した。尚、増殖細胞数の計測には血球計算盤を用い、CAR-T細胞数及び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た。
1.方法
0日目:末梢血から単核球(PBMC)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10)とpCMV-pigBacベクター(図5)を5μg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(1)の方法に従い、残りのPBMCから活性化T細胞を調製しておく。
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。
培養終了後(14日目)、CAR-T細胞数及び遺伝子導入効率(CAR-T細胞/全生細胞)を求めた。CAR-T細胞数と遺伝子導入効率は、1回目の実験が2.81×107個と55.1%、2回目の実験が1.03×107個と52.0%、3回目の実験が9.22×106個と42.3%であった。このように高い遺伝子導入効率を達成した。尚、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。
新規培養法で調製したCAR-T細胞の細胞障害活性及び抗腫瘍活性を以下の方法で評価した。
1.CD19陽性白血病細胞株との共培養実験
6人の健常人末梢血から培養法3を用いてCAR-T細胞を作製した。CAR-T細胞(1×105個)とCD19陽性白血病細胞株(5×105個)を10%ウシ胎仔血清含RPMI1640培地下で7日間共培養した(エフェクター細胞:標的細胞=1:5)。CAR-T細胞の代わりに、遺伝子非導入活性化T細胞(培養法3から遺伝子導入操作を省略して調製したT細胞)を用いたものをコントロールとした。7日間の共培養後に細胞を回収し、抗CD19抗体と抗CD3抗体で染色後、counting beadsとフローサイトメーターで細胞数をカウントした。以下の計算式で残存腫瘍細胞数を算出し、CAR-T細胞の細胞障害活性を評価した。尚、3種類のCD19陽性白血病細胞株(KOPN30bi、SK-9、TCC-Y/sr)を用いて以上の実験を行った。
標準化腫瘍細胞残存率(%)=100×(T細胞と共培養したウェルの残存腫瘍細胞数-T細胞と共培養していないウェルの腫瘍細胞数)/T細胞と共培養していないウェルの腫瘍細胞数
3日前(D-3)に尾静脈からCD19陽性細胞株(ルシフェラーゼ遺伝子導入Daudi、1×106個)を注射したNSGマウス(各試験区 5匹)に対し、培養法3で作製したCAR-T細胞(1×107個)を尾静脈から注射した(D0)。コントロール(NT)では、遺伝子非導入活性化T細胞(培養法3から遺伝子導入操作を省略して調製したT細胞)を注射した。適宜ルシフェリンを腹腔内投与し、生体内イメージングシステムを用いてマウスを撮像した。
以上の通り、新規培養法で作製したCAR-T細胞が優れた活性を示すことがin vitro及びin vivoの実験で確認された。即ち、治療効果の高いCAR-T細胞を効率的に作製する手段として、新規培養法が極めて有効であることが示された。
Claims (20)
- 以下のステップ(1)~(4)を含む、キメラ抗原受容体を発現する遺伝子改変T細胞の調製方法:
(1)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を用意するステップ;
(2)トランスポゾン法によって、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T細胞を得るステップ;
(3)ステップ(1)で用意した非増殖性細胞とステップ(2)で得た遺伝子改変T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ステップ;
(4)培養後の細胞を回収するステップ。 - ステップ(3)とステップ(4)の間に、共培養後の細胞を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に記載の調製方法。
- ステップ(3)の共培養の期間が1日~14日である、請求項1又は2に記載の調製方法。
- ステップ(3)を、T細胞増殖因子の存在下で行う、請求項1~3のいずれか一項に記載の調製方法。
- T細胞増殖因子がIL-15である、請求項4に記載の調製方法。
- T細胞増殖因子としてIL-15とIL-7を併用する、請求項4に記載の調製方法。
- 以下のステップ(i)~(iv)を含む、キメラ抗原受容体を発現する遺伝子改変T細胞の調製方法:
(i)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を用意するステップ;
(ii)トランスポゾン法によって、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T細胞を得るステップ;
(iii)ステップ(i)で用意した非増殖性細胞とステップ(ii)で得た遺伝子改変T細胞を混合し、共培養するステップ;
(iv)培養後の細胞を回収するステップ。 - ステップ(iii)とステップ(iv)の間に、共培養後の細胞を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7に記載の調製方法。
- ステップ(iii)の共培養の期間が1日~14日である、請求項7又は8に記載の調製方法。
- ステップ(iii)を、T細胞増殖因子の存在下で行う、請求項7~9のいずれか一項に記載の調製方法。
- T細胞増殖因子がIL-15である、請求項10に記載の調製方法。
- T細胞増殖因子としてIL-15とIL-7を併用する、請求項10に記載の調製方法。
- 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1~12のいずれか一項に記載の調製方法。
- 増殖能を喪失させる処理が放射線照射である、請求項1~13のいずれか一項に記載の調製方法。
- トランスポゾン法がPiggyBacトランスポゾン法である、請求項1~14のいずれか一項に記載の調製方法。
- 標的抗原がCD19、CD19、GD2、GMCSF受容体又はIGF受容体である、請求項1~15のいずれか一項に記載の調製方法。
- 非増殖性細胞と、遺伝子改変T細胞が同一の個体に由来する、請求項1~16のいずれか一項に記載の調製方法。
- 請求項1~17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T細胞。
- 請求項18に記載の遺伝子改変T細胞を治療上有効量含む、細胞製剤。
- 請求項18に記載の遺伝子改変T細胞を、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。
Priority Applications (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN201680058705.6A CN108138171B (zh) | 2015-10-08 | 2016-10-07 | 表达嵌合抗原受体的基因修饰t细胞的制备方法 |
| EP16853766.0A EP3360961B1 (en) | 2015-10-08 | 2016-10-07 | Method for preparing genetically-modified t cells which express chimeric antigen receptor |
| US15/766,380 US11517590B2 (en) | 2015-10-08 | 2016-10-07 | Method for preparing genetically-modified T cells which express chimeric antigen receptor |
| EP23202460.4A EP4289944A3 (en) | 2015-10-08 | 2016-10-07 | Method for preparing genetically-modified t cells which express chimeric antigen receptor |
| JP2017544248A JP6857360B2 (ja) | 2015-10-08 | 2016-10-07 | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 |
| US17/969,071 US20230040878A1 (en) | 2015-10-08 | 2022-10-19 | Method for preparing genetically-modified t cells which express chimeric antigen receptor |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2015-200458 | 2015-10-08 | ||
| JP2015200458 | 2015-10-08 |
Related Child Applications (2)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| US15/766,380 A-371-Of-International US11517590B2 (en) | 2015-10-08 | 2016-10-07 | Method for preparing genetically-modified T cells which express chimeric antigen receptor |
| US17/969,071 Division US20230040878A1 (en) | 2015-10-08 | 2022-10-19 | Method for preparing genetically-modified t cells which express chimeric antigen receptor |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2017061615A1 true WO2017061615A1 (ja) | 2017-04-13 |
Family
ID=58487875
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2016/079989 Ceased WO2017061615A1 (ja) | 2015-10-08 | 2016-10-07 | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 |
Country Status (5)
| Country | Link |
|---|---|
| US (2) | US11517590B2 (ja) |
| EP (2) | EP4289944A3 (ja) |
| JP (1) | JP6857360B2 (ja) |
| CN (1) | CN108138171B (ja) |
| WO (1) | WO2017061615A1 (ja) |
Cited By (17)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN107119021A (zh) * | 2017-07-04 | 2017-09-01 | 王小平 | Pd‑1敲除cd19car‑t细胞的制备 |
| WO2018052142A1 (ja) * | 2016-09-16 | 2018-03-22 | キッセイ薬品工業株式会社 | 遺伝子改変細胞及びその作製方法 |
| US10227574B2 (en) | 2016-12-16 | 2019-03-12 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| US20190119640A1 (en) * | 2016-09-30 | 2019-04-25 | Poseida Therapeutics, Inc. | Modified stem cell memory t cells, methods of making and methods of using same |
| US10329543B2 (en) | 2017-10-23 | 2019-06-25 | Poseida Therapeutics, Inc. | Modified stem cell memory T cells, methods of making and methods of using same |
| WO2019128999A1 (zh) * | 2017-12-28 | 2019-07-04 | 上海细胞治疗研究院 | 获得高阳性率car-t细胞的方法 |
| WO2020085480A1 (ja) | 2018-10-26 | 2020-04-30 | キッセイ薬品工業株式会社 | 高効率な遺伝子改変細胞の作製方法 |
| WO2021020526A1 (ja) | 2019-07-31 | 2021-02-04 | 国立大学法人信州大学 | Car発現免疫細胞を含む細胞集団の製造方法 |
| JP2021016371A (ja) * | 2019-07-23 | 2021-02-15 | 株式会社東芝 | Car−t細胞の製造方法、核酸導入キャリア及びキット |
| JPWO2021100585A1 (ja) * | 2019-11-20 | 2021-05-27 | ||
| US11278570B2 (en) | 2016-12-16 | 2022-03-22 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| JP2022521486A (ja) * | 2019-02-08 | 2022-04-08 | ディーエヌエイ ツーポイント インコーポレイテッド | トランスポゾンに基づく免疫細胞の改変 |
| JP2022171485A (ja) * | 2021-04-30 | 2022-11-11 | 国立大学法人東海国立大学機構 | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 |
| JP2023523619A (ja) * | 2020-04-27 | 2023-06-06 | ベイラー カレッジ オブ メディスン | キメラ抗原受容体を発現するウイルス特異的免疫細胞による癌の治療及び予防 |
| US11760983B2 (en) | 2018-06-21 | 2023-09-19 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| WO2024024551A1 (ja) * | 2022-07-26 | 2024-02-01 | 国立大学法人京都大学 | シェディング構造を有する人工受容体 |
| US12419913B2 (en) | 2019-02-08 | 2025-09-23 | Dna Twopointo, Inc. | Modification of CAR-T cells |
Families Citing this family (6)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN108034669B (zh) * | 2017-11-17 | 2021-07-30 | 山东兴瑞生物科技有限公司 | 一种抗vegf基因、及利用其修饰的t细胞、制备方法和应用 |
| CN108841792A (zh) * | 2018-06-14 | 2018-11-20 | 浙江大学 | 靶向cd19和ebna1基因修饰的t细胞及其制备方法与应用 |
| CN115461361B (zh) * | 2020-05-08 | 2025-03-14 | 玛希敦大学 | 嵌合抗原受体和携带该受体的经修饰的细胞 |
| CN112625901B (zh) * | 2020-12-01 | 2023-01-06 | 山东大学第二医院 | 一种微流控装置及其在嵌合抗原受体t细胞感染中的应用 |
| CN114457037B (zh) * | 2022-01-26 | 2023-12-22 | 徐州医科大学 | C57bl/6j小鼠来源cd19嵌合抗原受体t细胞的构建方法及应用 |
| CN115109774B (zh) * | 2022-06-24 | 2023-06-06 | 浙江康佰裕生物科技有限公司 | 一种新型同种异体car-t细胞的制备及应用 |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2014179759A1 (en) * | 2013-05-03 | 2014-11-06 | Ohio State Innovation Foundation | Cs1-specific chimeric antigen receptor engineered immune effector cells |
| WO2015028444A1 (en) * | 2013-08-26 | 2015-03-05 | Universität Zu Köln | Anti cd30 chimeric antigen receptor and its use |
| WO2015058018A1 (en) * | 2013-10-17 | 2015-04-23 | National University Of Singapore | Chimeric receptor that triggers antibody-dependent cell cytotoxicity against multiple tumors |
Family Cites Families (13)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5530101A (en) | 1988-12-28 | 1996-06-25 | Protein Design Labs, Inc. | Humanized immunoglobulins |
| IL162181A (en) | 1988-12-28 | 2006-04-10 | Pdl Biopharma Inc | A method of producing humanized immunoglubulin, and polynucleotides encoding the same |
| EP1612217A3 (en) | 2000-03-27 | 2006-01-18 | City of Hope | Immuno-reactive peptide CTL epitopes of human cytomegalovirus |
| JP2002255997A (ja) | 2001-06-07 | 2002-09-11 | Aichi Prefecture | Cd8+細胞傷害性tリンパ球エピトープペプチド及びその用途 |
| JP2004242599A (ja) | 2003-02-14 | 2004-09-02 | Aichi Prefecture | Cd8+細胞傷害性tリンパ球エピトープペプチド及びその用途 |
| JP4976294B2 (ja) | 2005-08-03 | 2012-07-18 | 株式会社医学生物学研究所 | 細胞傷害性t細胞エピトープペプチド及びその用途 |
| US20070036773A1 (en) * | 2005-08-09 | 2007-02-15 | City Of Hope | Generation and application of universal T cells for B-ALL |
| JP4824389B2 (ja) | 2005-10-28 | 2011-11-30 | 株式会社医学生物学研究所 | エプスタイン−バールウイルス感染細胞を特異的に攻撃する細胞傷害性t細胞エピトープペプチド及びその用途 |
| JP2006188513A (ja) | 2005-12-26 | 2006-07-20 | Aichi Prefecture | Cd8+細胞傷害性tリンパ球エピトープペプチド及びその用途 |
| JP2011177487A (ja) | 2010-02-04 | 2011-09-15 | Imasen Electric Ind Co Ltd | 電動車いす向け車輪格納装置 |
| PH12013501201A1 (en) * | 2010-12-09 | 2013-07-29 | Univ Pennsylvania | Use of chimeric antigen receptor-modified t cells to treat cancer |
| BR112014014591A8 (pt) | 2011-12-12 | 2017-07-04 | Baylor College Medicine | processo para a expansão in vitro de células t específicas de antígeno, produto de células t autólogas ou alogênicas expandidas, composição farmacêutica, e, método de tratamento de um paciente, e, usos de uma linhagem de células engenheirada, de células dendríticas pulsadas com misturas de antígeno-peptídeo, de t-apcs pulsadas com misturas de antígeno-peptídeo/peptídeo, e de misturas de antígeno-peptídeo com pbmcs |
| JP6275530B2 (ja) | 2014-04-08 | 2018-02-07 | ホシザキ株式会社 | 冷却貯蔵庫 |
-
2016
- 2016-10-07 EP EP23202460.4A patent/EP4289944A3/en active Pending
- 2016-10-07 JP JP2017544248A patent/JP6857360B2/ja active Active
- 2016-10-07 WO PCT/JP2016/079989 patent/WO2017061615A1/ja not_active Ceased
- 2016-10-07 CN CN201680058705.6A patent/CN108138171B/zh active Active
- 2016-10-07 EP EP16853766.0A patent/EP3360961B1/en active Active
- 2016-10-07 US US15/766,380 patent/US11517590B2/en active Active
-
2022
- 2022-10-19 US US17/969,071 patent/US20230040878A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2014179759A1 (en) * | 2013-05-03 | 2014-11-06 | Ohio State Innovation Foundation | Cs1-specific chimeric antigen receptor engineered immune effector cells |
| WO2015028444A1 (en) * | 2013-08-26 | 2015-03-05 | Universität Zu Köln | Anti cd30 chimeric antigen receptor and its use |
| WO2015058018A1 (en) * | 2013-10-17 | 2015-04-23 | National University Of Singapore | Chimeric receptor that triggers antibody-dependent cell cytotoxicity against multiple tumors |
Non-Patent Citations (5)
Cited By (40)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| JPWO2018052142A1 (ja) * | 2016-09-16 | 2019-06-24 | キッセイ薬品工業株式会社 | 遺伝子改変細胞及びその作製方法 |
| WO2018052142A1 (ja) * | 2016-09-16 | 2018-03-22 | キッセイ薬品工業株式会社 | 遺伝子改変細胞及びその作製方法 |
| US10683355B2 (en) | 2016-09-16 | 2020-06-16 | Kissei Pharmaceutical Co., Ltd. | Genetically-modified cells and method for producing same |
| US10513686B2 (en) | 2016-09-30 | 2019-12-24 | Poseida Therapeutics, Inc. | Modified stem cell memory T cells, methods of making and methods of using same |
| US20190119640A1 (en) * | 2016-09-30 | 2019-04-25 | Poseida Therapeutics, Inc. | Modified stem cell memory t cells, methods of making and methods of using same |
| US20190119637A1 (en) * | 2016-09-30 | 2019-04-25 | Poseida Therapeutics, Inc. | Modified stem cell memory t cells, methods of making and methods of using same |
| US10415016B2 (en) * | 2016-09-30 | 2019-09-17 | Poseida Therapeutics, Inc. | Modified stem cell memory T cells, methods of making and methods of using same |
| US11802269B2 (en) | 2016-09-30 | 2023-10-31 | Poseida Therapeutics, Inc. | Superpiggybac transposase compositions |
| US11278570B2 (en) | 2016-12-16 | 2022-03-22 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| US11111483B2 (en) | 2016-12-16 | 2021-09-07 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems and methods |
| US12097220B2 (en) | 2016-12-16 | 2024-09-24 | R&D Systems, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| US10227574B2 (en) | 2016-12-16 | 2019-03-12 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| US12037611B2 (en) | 2016-12-16 | 2024-07-16 | R&D Systems, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| US11162084B2 (en) | 2016-12-16 | 2021-11-02 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| CN107119021A (zh) * | 2017-07-04 | 2017-09-01 | 王小平 | Pd‑1敲除cd19car‑t细胞的制备 |
| US10329543B2 (en) | 2017-10-23 | 2019-06-25 | Poseida Therapeutics, Inc. | Modified stem cell memory T cells, methods of making and methods of using same |
| CN109971718A (zh) * | 2017-12-28 | 2019-07-05 | 上海细胞治疗研究院 | 获得高阳性率car-t细胞的方法 |
| CN109971718B (zh) * | 2017-12-28 | 2023-09-01 | 上海细胞治疗研究院 | 获得高阳性率car-t细胞的方法 |
| WO2019128999A1 (zh) * | 2017-12-28 | 2019-07-04 | 上海细胞治疗研究院 | 获得高阳性率car-t细胞的方法 |
| US11760983B2 (en) | 2018-06-21 | 2023-09-19 | B-Mogen Biotechnologies, Inc. | Enhanced hAT family transposon-mediated gene transfer and associated compositions, systems, and methods |
| JPWO2020085480A1 (ja) * | 2018-10-26 | 2021-09-16 | 国立大学法人信州大学 | 高効率な遺伝子改変細胞の作製方法 |
| JP7632868B2 (ja) | 2018-10-26 | 2025-02-19 | 国立大学法人信州大学 | 高効率な遺伝子改変細胞の作製方法 |
| WO2020085480A1 (ja) | 2018-10-26 | 2020-04-30 | キッセイ薬品工業株式会社 | 高効率な遺伝子改変細胞の作製方法 |
| US12419913B2 (en) | 2019-02-08 | 2025-09-23 | Dna Twopointo, Inc. | Modification of CAR-T cells |
| JP2022521486A (ja) * | 2019-02-08 | 2022-04-08 | ディーエヌエイ ツーポイント インコーポレイテッド | トランスポゾンに基づく免疫細胞の改変 |
| JP2023153231A (ja) * | 2019-07-23 | 2023-10-17 | 株式会社東芝 | Car-t細胞の製造方法、核酸導入キャリア及びキット |
| US12398190B2 (en) | 2019-07-23 | 2025-08-26 | Kabushiki Kaisha Toshiba | Method of producing CAR-T cells, nucleic acid-introducing carrier and kit |
| JP7653113B2 (ja) | 2019-07-23 | 2025-03-28 | 株式会社東芝 | Car-t細胞の製造方法、核酸導入キャリア及びキット |
| JP2021016371A (ja) * | 2019-07-23 | 2021-02-15 | 株式会社東芝 | Car−t細胞の製造方法、核酸導入キャリア及びキット |
| WO2021020526A1 (ja) | 2019-07-31 | 2021-02-04 | 国立大学法人信州大学 | Car発現免疫細胞を含む細胞集団の製造方法 |
| JPWO2021020526A1 (ja) * | 2019-07-31 | 2021-02-04 | ||
| WO2021019706A1 (ja) * | 2019-07-31 | 2021-02-04 | 国立大学法人信州大学 | Car発現免疫細胞を含む細胞集団の製造方法 |
| JP7576547B2 (ja) | 2019-07-31 | 2024-10-31 | 国立大学法人信州大学 | Car発現免疫細胞を含む細胞集団の製造方法 |
| WO2021100585A1 (ja) * | 2019-11-20 | 2021-05-27 | 国立大学法人東海国立大学機構 | キメラ抗原受容体遺伝子改変リンパ球の調製方法 |
| JPWO2021100585A1 (ja) * | 2019-11-20 | 2021-05-27 | ||
| JP2023523620A (ja) * | 2020-04-27 | 2023-06-06 | ベイラー カレッジ オブ メディスン | キメラ抗原受容体を発現するウイルス特異的免疫細胞 |
| JP2023523621A (ja) * | 2020-04-27 | 2023-06-06 | ベイラー カレッジ オブ メディスン | キメラ抗原受容体を発現するウイルス特異的免疫細胞によるアロ反応性の治療及び予防 |
| JP2023523619A (ja) * | 2020-04-27 | 2023-06-06 | ベイラー カレッジ オブ メディスン | キメラ抗原受容体を発現するウイルス特異的免疫細胞による癌の治療及び予防 |
| JP2022171485A (ja) * | 2021-04-30 | 2022-11-11 | 国立大学法人東海国立大学機構 | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 |
| WO2024024551A1 (ja) * | 2022-07-26 | 2024-02-01 | 国立大学法人京都大学 | シェディング構造を有する人工受容体 |
Also Published As
| Publication number | Publication date |
|---|---|
| EP3360961A4 (en) | 2019-04-17 |
| JP6857360B2 (ja) | 2021-04-14 |
| CN108138171A (zh) | 2018-06-08 |
| EP3360961B1 (en) | 2023-11-22 |
| CN108138171B (zh) | 2022-05-13 |
| EP3360961A1 (en) | 2018-08-15 |
| US11517590B2 (en) | 2022-12-06 |
| EP4289944A3 (en) | 2024-03-13 |
| US20230040878A1 (en) | 2023-02-09 |
| EP4289944A2 (en) | 2023-12-13 |
| US20180289742A1 (en) | 2018-10-11 |
| JPWO2017061615A1 (ja) | 2018-08-02 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP6857360B2 (ja) | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 | |
| EP3825404A1 (en) | Anti-gpc3 single-chain antibody-containing car | |
| WO2017133175A1 (en) | Engineered mammalian cells for cancer therapy | |
| JP6884697B2 (ja) | T細胞を刺激および拡大する組成物および方法 | |
| CN111247242A (zh) | 嵌合抗原受体(CARs)、组合物及其使用方法 | |
| JP6971986B2 (ja) | 免疫療法の抗腫瘍活性を高めるための間葉系幹細胞 | |
| JP6853514B2 (ja) | 炎症性サイトカインの産生が抑制されるキメラ抗原受容体遺伝子改変リンパ球 | |
| JP2018510652A (ja) | Her2/erbb2キメラ抗原受容体 | |
| KR20180021683A (ko) | 생체내 지속성 및 치료학적 활성 및 이의 증식을 위한 nkt-세포 서브세트 | |
| WO2024199197A1 (zh) | 功能增强型工程化免疫细胞及其制备和应用 | |
| JP7057975B2 (ja) | 殺細胞効果を有するキメラ抗原受容体遺伝子改変リンパ球 | |
| CN118613499A (zh) | 对b细胞受体特异的嵌合抗原受体的组合物和方法 | |
| WO2019210207A2 (en) | Chimeric antigen receptor t regulatory cells for the treatment of atherosclerosis | |
| US20180369282A1 (en) | Gene-modified lymphocytes expressing chimeric antigen receptor in which production of inflammatory cytokines is inhibited | |
| JP7576547B2 (ja) | Car発現免疫細胞を含む細胞集団の製造方法 | |
| WO2021100585A1 (ja) | キメラ抗原受容体遺伝子改変リンパ球の調製方法 | |
| US20230381312A1 (en) | Redirecting glucose metabolism to limit stress and improve adoptive cell therapy | |
| WO2023080178A1 (ja) | Car-t細胞の製造方法 | |
| HK40104781A (en) | Method for preparing genetically-modified t cells which express chimeric antigen receptor | |
| CN116254230A (zh) | 用于制备和扩增通用型人源化抗cd19 car-nk细胞的方法及其用途 | |
| JP2022171485A (ja) | キメラ抗原受容体を発現する遺伝子改変t細胞の調製方法 | |
| JP7075595B2 (ja) | チオプリン高感受性キメラ抗原受容体遺伝子改変リンパ球 | |
| US20230295568A1 (en) | Chimeric antigen receptor gene-modified lymphocyte having cytocidal effect | |
| Sheng et al. | PD-1 Restrains Interferon-Gamma Mediated Activation-Induced Cell Death that Limits T cell Engraftment and Efficacy | |
| WO2024041618A1 (zh) | 联合表达cd40l的工程化免疫细胞及其制备和应用 |
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: 16853766 Country of ref document: EP Kind code of ref document: A1 |
|
| ENP | Entry into the national phase |
Ref document number: 2017544248 Country of ref document: JP Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 15766380 Country of ref document: US |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2016853766 Country of ref document: EP |