WO2021136040A1 - Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice - Google Patents

Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice Download PDF

Info

Publication number
WO2021136040A1
WO2021136040A1 PCT/CN2020/138691 CN2020138691W WO2021136040A1 WO 2021136040 A1 WO2021136040 A1 WO 2021136040A1 CN 2020138691 W CN2020138691 W CN 2020138691W WO 2021136040 A1 WO2021136040 A1 WO 2021136040A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
cell
present
tumor
Prior art date
Application number
PCT/CN2020/138691
Other languages
English (en)
Chinese (zh)
Inventor
谭炳合
史秀娟
杜冰
刘明耀
席在喜
Original Assignee
华东师范大学
上海邦耀生物科技有限公司
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 华东师范大学, 上海邦耀生物科技有限公司 filed Critical 华东师范大学
Publication of WO2021136040A1 publication Critical patent/WO2021136040A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464436Cytokines
    • A61K39/464438Tumor necrosis factors [TNF], CD70
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464493Prostate associated antigens e.g. Prostate stem cell antigen [PSCA]; Prostate carcinoma tumor antigen [PCTA]; Prostatic acid phosphatase [PAP]; Prostate-specific G-protein-coupled receptor [PSGR]
    • A61K39/464495Prostate specific membrane antigen [PSMA]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • 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
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • 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
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/58Prostate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • 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
    • C12N2510/00Genetically modified cells
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Definitions

  • the present invention relates to the field of biomedicine, in particular to the preparation and application of a chimeric antigen receptor T cell co-expressing immunoregulatory molecules.
  • Chimeric antigen receptor (CAR) T cell therapy is a new cellular immunotherapy technology that has developed very rapidly in recent years. This technology combines the high affinity of antigen and antibody with the killing effect of T lymphocytes. Combine to construct a specific chimeric antigen receptor, insert the gene encoding the chimeric antigen receptor into T lymphocytes through a certain way, make the T lymphocytes express this chimeric antigen receptor, and then expand and purify the chimeric antigen receptor in vitro The genetically modified T cells are imported into the body. CAR-T cells specifically recognize the target antigen in the body and undergo a series of immune responses. The T cells activate and expand and secrete cytokines. The capacitive complex specifically kills target cells in a restrictive manner.
  • CAR Chimeric antigen receptor
  • tumors construct a tumor-associated antigen chimeric antigen receptor, which can be transferred to T cells to express the chimeric antigen receptor, which can specifically recognize the antigen on the surface of tumor cells, thereby activating T cells to exert cellular immunity and eliminate tumors.
  • T cells to achieve the purpose of anti-tumor.
  • CAR-T technology has achieved gratifying breakthroughs in the treatment of hematomas, its efficacy in the treatment of solid tumors, which account for the vast majority of malignant tumors, has not been satisfactory.
  • CAR-T therapy is not effective in the treatment of solid tumors. It is related to solid tumor tissue barriers, tumor cells are highly heterogeneous, lacks good tumor-specific antigens or tumor-related antigens, highly suppressed tumor immune microenvironment, and CAR-T The cell's own activation signal is closely related. In the process of treating solid tumors, after CAR-T cells break through the physical barrier to recognize tumor cells, they must play an effective tumor cell killing effect.
  • cytokine storm Systemically increasing cytokine expression will undoubtedly increase the risk of cytokine storm related to CAR-T treatment.
  • Integrating and expressing different costimulatory molecules in the intracellular activation domain of CAR molecules can provide costimulatory signals required for CAR-T cell activation, but the activation process depends on the expression of tumor-related or tumor-specific antigens and cannot By interacting with other immune cells in the tumor microenvironment to reshape and improve the tumor microenvironment, the therapeutic effect on solid tumors is very limited.
  • the purpose of the present invention is to provide a new type of chimeric antigen receptor molecule, which can effectively resist the inhibition of tumor immune microenvironment after being expressed on T cells, and maintain or improve the effector function and expansion of CAR-T cells Ability and sustainability, in the treatment of solid tumors, it can show a therapeutic effect that is significantly better than the existing CAR structure.
  • the first aspect of the present invention provides a chimeric antigen receptor CAR, the chimeric antigen receptor CAR comprising: an antigen binding domain, a transmembrane domain and an intracellular domain, wherein the antigen binding domain is specific Binding to tumor cell surface antigen;
  • the chimeric antigen receptor CAR also includes: an immunomodulatory molecule that is connected to the intracellular domain and can be co-expressed.
  • the tumor cell antigens include cell surface antigens of various solid tumors and non-solid tumors.
  • the tumor cell surface antigen is selected from the group consisting of CD19, BCMA, CD38, PSMA, HER2, GPC3, Mesothelin, Claudin 18.2, EGFR, EGFRVIII, CEA, GD2, IL13R, FAP, CD171, Or a combination.
  • the tumor cell surface antigen includes CD19 and/or PSMA.
  • the immunomodulatory molecule is selected from the group consisting of GITRL, 4-1BBL, CD40, LIGHT, B7.1, B7.2, OX40L, CD70, or a combination thereof.
  • the immunomodulatory molecule includes GITRL.
  • the antigen-binding domain is an antibody or an antigen-binding fragment.
  • the antigen-binding fragment is Fab or scFv or single domain antibody sdFv.
  • Each "-" is independently a connecting peptide or a peptide bond
  • Z1 is no or signal peptide sequence
  • T is an antibody single-chain variable region sequence targeting tumor cell surface antigen
  • H is no or hinge area
  • TM is the transmembrane domain
  • C is a costimulatory signal molecule
  • Z2 is the cytoplasmic signal transduction sequence derived from CD3 ⁇
  • Z3 is a self-cleaving protein
  • P is an immunomodulatory molecule
  • n 1, 2, 3, or 4.
  • the structure of the single-chain variable region sequence of the antibody targeting the tumor cell surface antigen is as shown in formula A1 or A2:
  • V L1 and V L2 are the light chain variable regions of antibodies against tumor cell surface antigens;
  • V H1 and V H2 are the heavy chain variable regions of antibodies against tumor cell surface antigens;
  • "-" is the connecting peptide (or Flexible linker) or peptide bond.
  • V L1 and V H1 are connected by a flexible joint.
  • the flexible linker is 1-5 (preferably, 2-4) consecutive sequences shown in SEQ ID NO.: 4 (GGGGS).
  • amino acid sequence of V L1 is as shown in SEQ ID NO.: 2 (CAR of PSMA) at positions 22-128, and the amino acid sequence of V H1 is as shown in SEQ ID NO.: 2 at positions 144-128. 258 shows.
  • amino acid sequence of V L2 is as shown in SEQ ID NO.: 3 (CAR of CD19) at positions 22-128, and the amino acid sequence of V H2 is as shown in SEQ ID NO.: 3 at positions 144-128. 263 shows.
  • the Z1 is a signal peptide of a protein selected from the group consisting of CD8a, CSF1R, or a combination thereof.
  • the H is the hinge region of a protein selected from the group consisting of CD8a, IgG, CD28, or a combination thereof.
  • the TM is a transmembrane region of a protein selected from the group consisting of CD8a, CD4, CD28, or a combination thereof.
  • the C is a costimulatory signal molecule of a protein selected from the group consisting of: 4-1BB (CD137), OX40, CD28, CD30, CD40, CD70, CD134, PD1, Dap10, CDS, ICAM- 1. HVEM, GITR, or a combination thereof.
  • the C includes a costimulatory signal molecule derived from 4-1BB.
  • the self-cleaving protein of Z3 is selected from the group consisting of T2A, P2A, E2A, F2A, or a combination thereof.
  • the self-cleaving protein of Z3 includes T2A.
  • amino acid sequence of the self-cleaving protein of Z3 is shown in SEQ ID NO.:7.
  • the immunomodulatory molecules include wild-type immunomodulatory molecules and mutant immunomodulatory molecules, or active fragments thereof.
  • the immunomodulatory molecule is selected from the group consisting of GITRL, 4-1BBL, CD40, LIGHT, B7.1, B7.2, OX40L, CD70, or a combination thereof.
  • the GITRL has an amino acid sequence as shown in SEQ ID NO.:1.
  • amino acid sequence of the GITRL is shown in SEQ ID NO.:1.
  • amino acid sequence of the CAR is shown in SEQ ID No.: 2 or 3.
  • the second aspect of the present invention provides a nucleic acid molecule that encodes the chimeric antigen receptor (CAR) of the first aspect of the present invention.
  • CAR chimeric antigen receptor
  • nucleic acid molecule encoding the chimeric antigen receptor (CAR) of claim 1 is shown in SEQ ID NO.: 5 or 6.
  • the third aspect of the present invention provides a vector containing the nucleic acid molecule according to the second aspect of the present invention.
  • the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, or a combination thereof.
  • the vector is a lentiviral vector.
  • the fourth aspect of the present invention provides a host cell that contains the vector of the third aspect of the present invention or the nucleic acid molecule of the second aspect of the present invention integrated into the chromosome.
  • the cell is an isolated cell, and/or the cell is a genetically engineered cell.
  • the cell is a mammalian cell, preferably a human cell.
  • the host cells include engineered immune cells.
  • the immune cells also express exogenous immune regulatory molecular proteins.
  • the exogenous immunomodulatory molecule protein is independently expressed and/or co-expressed with the CAR targeting tumor cell surface antigen.
  • the co-expression with the CAR targeting the tumor cell surface antigen includes the tandem expression of an immunomodulatory molecule protein and the CAR targeting the tumor cell surface antigen.
  • the engineered immune cells include T cells, NK cells or macrophages.
  • the cell is a T cell.
  • the engineered immune cells are selected from the following group:
  • CAR-NK cells Chimeric antigen receptor NK cells
  • TCR Exogenous T cell receptor (TCR) T cells
  • the immune cells are autologous.
  • the immune cells are allogeneic.
  • the cell is a CAR-T cell, and the CAR-T cell expresses the chimeric antigen receptor CAR of claim 1.
  • the fifth aspect of the present invention provides a method for preparing engineered immune cells that express the CAR according to the first aspect of the present invention, wherein the method includes the steps of: The nucleic acid molecule or the vector described in the third aspect of the present invention is transferred into immune cells to obtain the engineered immune cells.
  • the introduction includes simultaneous, sequential, or sequential introduction.
  • the immune cells are T cells or NK cells.
  • the method further includes the step of performing function and effectiveness testing on the obtained engineered immune cells.
  • the sixth aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the CAR according to the first aspect of the present invention, the nucleic acid molecule according to the second aspect of the present invention, the carrier according to the third aspect of the present invention, Or the host cell described in the fourth aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition is a liquid preparation.
  • the dosage form of the pharmaceutical composition is an injection.
  • the host cell includes engineered immune cells.
  • the engineered immune cells are (i) chimeric antigen receptor T cells (CAR-T cells); or (ii) chimeric antigen receptor NK cells (CAR-NK cells).
  • the concentration of the cells is 1 ⁇ 10 3 -1 ⁇ 10 8 cells/mL, preferably 1 ⁇ 10 6 -1 ⁇ 10 7 cells/mL .
  • the pharmaceutical composition further includes an immunomodulatory molecule agonist.
  • the immunomodulatory molecule agonist is selected from the group consisting of antibodies, small molecule compounds, synthetic or recombinant polypeptides, or combinations thereof.
  • the pharmaceutical composition also contains other drugs that kill tumor cells (such as antibody drugs, chemotherapeutic drugs or other CAR-T drugs).
  • the seventh aspect of the present invention provides a CAR according to the first aspect of the present invention, a nucleic acid molecule according to the second aspect of the present invention, a vector according to the third aspect of the present invention, and a host cell according to the fourth aspect of the present invention Or the use of the pharmaceutical composition of the sixth aspect of the present invention to prepare drugs or preparations for killing tumor cells.
  • the tumor cells include CD19-positive tumor cells.
  • the tumor cells include PSMA-positive tumor cells.
  • the tumor cells are derived from solid tumors.
  • the solid tumor is selected from the group consisting of breast cancer, pancreatic cancer, colon cancer, gastric cancer, lung cancer, renal cell carcinoma, liver cancer, ovarian cancer, esophageal adenocarcinoma, prostate cancer, cervical cancer, multiple Osteosarcoma, melanoma, nasopharyngeal carcinoma, or a combination thereof.
  • the eighth aspect of the present invention provides a kit for killing tumor cells, the kit containing a container, and the CAR according to the first aspect of the present invention and the nucleic acid molecule according to the second aspect of the present invention in the container , The vector according to the third aspect of the present invention, or the host cell according to the fourth aspect of the present invention.
  • the kit further contains a label or instructions for use.
  • the ninth aspect of the present invention provides a method for killing tumor cells, including:
  • the subject includes humans or non-human mammals.
  • the non-human mammals include rodents (such as mice, rats, rabbits) and primates (such as monkeys).
  • the method is non-therapeutic and non-diagnostic.
  • the tenth aspect of the present invention provides a method for treating cancer or tumor, including:
  • the tumor cells include CD19-positive tumor cells.
  • the tumor cells include PSMA-positive tumor cells.
  • the tumor includes a solid tumor.
  • the solid tumor is selected from the group consisting of breast cancer, pancreatic cancer, colon cancer, gastric cancer, lung cancer, renal cell carcinoma, liver cancer, ovarian cancer, esophageal adenocarcinoma, prostate cancer, cervical cancer, multiple Osteosarcoma, melanoma, nasopharyngeal carcinoma, or a combination thereof.
  • Figure 1 shows the GITRL-CAR expression framework.
  • FIG. 2 shows that GITRL-CAR is highly expressed in human primary T cells.
  • FIG. 3 shows that the anti-tumor effect and proliferation ability of GITRL-CART is significantly better than the existing second-generation CART.
  • FIG. 4 shows that GITRL promotes the differentiation of Th9 subsets in CAR-T cells.
  • Figure 5 shows that the depletion capacity of GITRL-CART is reduced and the continuity is significantly enhanced.
  • Figure 6 shows that GITRL-CART has a stronger anti-tumor effect in vivo.
  • CAR chimeric antigen receptor
  • the present invention takes CAR-T cells as an example, and representatively describes the engineered immune cells of the present invention in detail.
  • the engineered immune cells of the present invention are not limited to the CAR-T cells described in the context, and the engineered immune cells of the present invention have the same or similar technical features and beneficial effects as the CAR-T cells described in the context.
  • immune cells express chimeric antigen receptor CAR
  • NK cells are equivalent to T cells (or T cells can replace NK cells)
  • TCR is equivalent to CAR (or CAR can be replaced by TCR ).
  • chimeric antigen receptor is a fusion protein comprising an extracellular domain capable of binding antigen, a transmembrane domain derived from a different polypeptide from the extracellular domain, and at least one cell Inner domain.
  • Chimeric antigen receptor is also called “chimeric receptor", “T-body” or “chimeric immune receptor (CIR)”.
  • the "extracellular domain capable of binding to an antigen” refers to any oligopeptide or polypeptide capable of binding to a certain antigen.
  • Extracellular domain refers to any oligopeptide or polypeptide known as a domain that transmits signals to activate or inhibit biological processes in a cell.
  • domain refers to a region in a polypeptide that is independent of other regions and folds into a specific structure.
  • tumor antigen refers to a biological molecule with antigenicity, the expression of which leads to cancer.
  • administering refers to the application of exogenous drugs, therapeutic agents, diagnostic agents or compositions to animals, humans, subjects, cells, tissues, organs, or biological fluids.
  • administering can refer to treatment, pharmacokinetics, diagnosis, research, and experimental methods.
  • the treatment of cells includes contact between reagents and cells, contact between reagents and fluids, and contact between fluids and cells.
  • administering also mean treatment by reagents, diagnostics, binding compositions, or by another cell in vitro and ex vivo.
  • Treatment when applied to humans, animals or research subjects, refers to treatment, preventive or preventive measures, research and diagnosis; including anti-human LAG-3 antibodies and humans or animals, subjects, cells, tissues , Physiological compartment or physiological fluid contact.
  • treatment refers to the administration of an internal or external therapeutic agent, including any one CAR of the present invention and a composition thereof, to a patient who has one or more disease symptoms, and the therapeutic agent is known to These symptoms have a therapeutic effect.
  • the patient is administered in an amount (therapeutically effective amount) of a therapeutic agent effective to alleviate one or more disease symptoms.
  • the term “optional” or “optionally” means that the event or situation described later can occur but does not have to occur.
  • “optionally comprising 1-3 antibody heavy chain variable regions” means that the antibody heavy chain variable regions of a specific sequence can have but not necessarily have, and can be 1, 2, or 3.
  • sequence identity in the present invention refers to the degree of identity between two nucleic acid or two amino acid sequences when optimally aligned and compared with appropriate mutations such as substitutions, insertions, or deletions.
  • sequence identity between the sequence described in the present invention and its identical sequence may be at least 85%, 90% or 95%, preferably at least 95%. Non-limiting examples include 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% ,100%.
  • Immunomodulatory molecules are substances produced by immune cells or other cells that regulate the immune response, including antibodies, lymphokines, polysaccharides, polypeptides, lysozyme and even some small molecule compounds.
  • the immunomodulatory molecules of the present invention include but are not limited to: GITRL, 4-1BBL, CD40, LIGHT, B7.1, B7.2, OX40L, CD70.
  • the immunomodulatory molecule of the present invention includes GITRL.
  • amino acid sequence of the GITRL of the present invention is shown in SEQ ID NO.:1.
  • the tumor cell surface antigens of the present invention include but are not limited to CD19, BCMA, CD38, PSMA, HER2, GPC3, Mesothelin, Claudin 18.2, EGFR, EGFRVIII, CEA, GD2, IL13R, FAP, CD171.
  • CD19 is one of the important membrane antigens involved in the activation and proliferation of B cells. It is a surface marker shared by all B cells. B cells do not disappear after activation. It is the most important B cell marker factor. At the same time, CD19 is also a signal complex on the surface of B cells. The constituent parts of the body. CAR-T targeting CD19 is mainly used in the treatment of B cell malignant tumors. CD19 can be widely expressed on the surface of a variety of B-cell malignant tumor cells, but it is not expressed in other tissues and blood cells, and the presence of CD19 soluble protein has not been detected in the blood. Therefore, it is considered as an ideal target for CAR-T treatment of B-cell tumors. Clinical trial results show that the cure rate of CD19 CAR-T for acute B-lymphocytic leukemia (B-ALL) has reached 90%.
  • B-ALL acute B-lymphocytic leukemia
  • PSMA is a prostate-specific membrane surface antigen, which is highly expressed in most prostate cancer tissues. It is a tumor-associated antigen and a new diagnostic marker for prostate cancer.
  • the antigen binding domain of the chimeric antigen receptor CAR specifically binds to tumor cell surface antigens.
  • the antigen binding domain of the chimeric antigen receptor CAR of the present invention targets CD19 and/or PSMA.
  • the CAR can be designed to include a transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain that is naturally associated with one of the domains in the CAR is used.
  • transmembrane domains can be selected or modified by amino acid substitutions to avoid binding such domains to the transmembrane domains of the same or different surface membrane proteins, thereby minimizing the interaction with the receptor complex. Interaction of other members.
  • the transmembrane domain can be derived from natural sources or synthetic sources. In natural sources, the domain can be derived from any membrane-bound or transmembrane protein.
  • the hinge region in the CAR of the present invention is the hinge region of CD8a
  • the transmembrane region of the present invention is the transmembrane region of CD8a.
  • the intracellular domain or another intracellular signaling domain of the CAR of the present invention is responsible for the activation of at least one normal effector function of the immune cell in which the CAR has been placed.
  • effector function refers to the exclusive function of the cell.
  • the effector function of T cells may be cytolytic activity or auxiliary activity including cytokine secretion. Therefore, the term “intracellular signal transduction domain” refers to the part of the protein that transduces effector function signals and directs the cell to perform specific functions.
  • the entire intracellular signaling domain can generally be used, in many cases, the entire chain need not be used.
  • intracellular signaling domain In terms of using truncated portions of intracellular signaling domains, such truncated portions can be used to replace the complete chain as long as it transduces effector function signals.
  • the term intracellular signaling domain therefore refers to any truncated portion of the intracellular signaling domain that is sufficient to transduce effector function signals.
  • intracellular signal transduction domain used in the CAR of the present invention include the cytoplasmic sequence of T cell receptor (TCR) and co-receptors that act in concert to initiate signal transduction after antigen receptor binding, and these sequences Any derivative or variant of and any synthetic sequence with the same functional capabilities.
  • TCR T cell receptor
  • the cytoplasmic domain of the CAR can be designed to include the CD3- ⁇ signaling domain itself, or can be combined with any other desired cytoplasmic domain (a Or multiple) joint.
  • the cytoplasmic domain of CAR may include a CD3 ⁇ chain portion and a costimulatory signal transduction region.
  • the costimulatory signal transduction region refers to a part of CAR that includes the intracellular domain of costimulatory molecules.
  • Co-stimulatory molecules are cell surface molecules required for effective response of lymphocytes to antigens, not antigen receptors or their ligands. Preferably, it includes 4-1BB (CD137) and the like.
  • the cytoplasmic signal transduction sequences in the cytoplasmic signal transduction portion of the CAR of the present invention can be connected to each other randomly or in a prescribed order.
  • short oligopeptide or polypeptide linkers preferably between 2 and 10 amino acids in length, can form the link.
  • the glycine-serine doublet provides a particularly suitable linker.
  • the cytoplasmic domain in the CAR of the present invention is designed to include the signaling domain of 4-1BB (costimulatory molecule) and the signaling domain of CD3 ⁇ .
  • Chimeric antigen receptors are composed of extracellular antigen recognition regions, usually scFv (single-chain variable fragment), transmembrane regions and intracellular co-stimulatory signal regions.
  • the design of CARs has gone through the following process:
  • the first-generation CAR has only one intracellular signal component CD3 ⁇ or Fc ⁇ RI molecule. Since there is only one activation domain in the cell, it can only cause transient T cell proliferation and less cytokine secretion. , And cannot provide long-term T cell proliferation signals and sustained anti-tumor effects in vivo, so it has not achieved good clinical effects.
  • the second-generation CARs introduce a costimulatory molecule based on the original structure, such as CD28, 4-1BB, OX40, and ICOS. Compared with the first-generation CARs, the function has been greatly improved, which further strengthens the persistence of CAR-T cells and the effect on tumor cells. The lethality. On the basis of the second-generation CARs, some new immunostimulatory molecules such as CD27 and CD134 are connected in series to develop into the third-generation and fourth-generation CARs.
  • the extracellular segment of CARs can recognize a specific antigen, and then transduce the signal through the intracellular domain to cause cell activation, proliferation, cytolytic toxicity, and cytokine secretion, thereby eliminating target cells.
  • autologous cells or heterologous donors
  • CAR immune cells or heterologous donors
  • the probability of graft-versus-host disease is extremely low, and the antigen is recognized by immune cells in a non-MHC-restricted manner.
  • CAR-immune cell therapy has achieved a very high clinical response rate in the treatment of hematological malignancies. Such a high response rate could not be achieved by any previous treatment method. It has triggered an upsurge of clinical research in the world.
  • the chimeric antigen receptor (CAR) of the present invention includes an extracellular domain, a transmembrane domain, and an intracellular domain.
  • the extracellular domain includes target-specific binding elements (also called antigen binding domains).
  • the intracellular domain includes a costimulatory signal transduction region and/or a zeta chain portion.
  • the costimulatory signal transduction region refers to a part of the intracellular domain including costimulatory molecules.
  • Co-stimulatory molecules are cell surface molecules required for effective response of lymphocytes to antigens, rather than antigen receptors or their ligands.
  • a linker can be incorporated between the extracellular domain and the transmembrane domain of the CAR, or between the cytoplasmic domain and the transmembrane domain of the CAR.
  • the term "linker” generally refers to any oligopeptide or polypeptide that functions to connect the transmembrane domain to the extracellular or cytoplasmic domain of a polypeptide chain.
  • the linker may comprise 0-300 amino acids, preferably 2 to 100 amino acids and most preferably 3 to 50 amino acids.
  • the CAR of the present invention When the CAR of the present invention is expressed in T cells, it can perform antigen recognition based on the antigen binding specificity. When it binds to its associated antigen, it affects tumor cells, resulting in tumor cells not growing, being promoted to die or being affected in other ways, and causing the patient's tumor burden to shrink or eliminate.
  • the antigen binding domain is preferably fused with an intracellular domain from one or more of the costimulatory molecule and/or zeta chain.
  • the antigen binding domain is fused with the intracellular domain combined with the 4-1BB signaling domain and/or the CD3 ⁇ signaling domain.
  • antigen-binding domain and “single-chain antibody fragment” all refer to Fab fragments, Fab" fragments, F(ab")2 fragments, or single Fv fragments that have antigen-binding activity.
  • the Fv antibody contains the variable region of the heavy chain and the variable region of the light chain, but does not have the constant region, and has the smallest antibody fragment with all the antigen binding sites.
  • an Fv antibody also contains a polypeptide linker between the VH and VL domains, and can form the structure required for antigen binding.
  • the antigen binding domain is usually scFv (single-chain variable fragment). The size of scFv is generally 1/6 that of a complete antibody.
  • the single-chain antibody is preferably an amino acid chain sequence encoded by a nucleotide chain.
  • the scFv includes an antibody that specifically recognizes the tumor highly expressed antigens CD19 and PSMA, preferably a single-chain antibody.
  • the scFv of the present invention also includes its conservative variants, which means that compared with the amino acid sequence of the scFv of the present invention, there are at most 10, preferably at most 8, more preferably at most 5, and most preferably Up to 3 amino acids are replaced by amino acids with similar or similar properties to form a polypeptide.
  • the number of added, deleted, modified and/or substituted amino acids is preferably no more than 40% of the total number of amino acids in the initial amino acid sequence, more preferably no more than 35%, more preferably 1-33%, It is more preferably 5-30%, more preferably 10-25%, and more preferably 15-20%.
  • the number of added, deleted, modified and/or substituted amino acids is usually 1, 2, 3, 4 or 5, preferably 1-3, more preferably 1-2, The best is one.
  • the CAR can be designed to include a transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain that is naturally associated with one of the domains in the CAR is used.
  • transmembrane domains can be selected or modified by amino acid substitutions to avoid binding such domains to the transmembrane domains of the same or different surface membrane proteins, thereby minimizing the interaction with the receptor complex. Interaction of other members.
  • the extracellular domain of the CAR of the present invention includes an antibody single-chain variable region sequence targeting a tumor cell surface antigen, preferably an antibody single-chain variable region sequence targeting a tumor cell surface antigen with a specific sequence.
  • the intracellular domain in the CAR of the present invention includes the transmembrane region of CD8a, the costimulatory factor of 4-1BB, and the signal transduction domain of CD3 ⁇ .
  • amino acid sequence of the CAR (the amino acid sequence of the CAR containing an immunomodulatory molecule (such as GITRL)) is shown in SEQ ID NO.: 2 or 3:
  • CD19 CAR containing immunomodulatory molecules such as GITRL
  • nucleotide sequence of the CAR (the nucleotide sequence of the CAR containing an immunomodulatory molecule (such as GITRL)) is shown in SEQ ID NO.: 5 or 6:
  • CD19 CAR containing immunomodulatory molecules such as GITRL
  • CAR-T cell As used herein, the terms “CAR-T cell”, “CAR-T” and “CAR-T cell of the present invention” all refer to the CAR-T cell of the present invention.
  • the CAR-T cell of the present invention can target tumor surface antigens. (Such as CD19, PSMA), used to treat tumors with high expression or positive tumor cell surface antigens (such as CD19, PSMA), especially solid tumors.
  • CAR-T cells have the following advantages over other T cell-based therapies: (1) The action process of CAR-T cells is not restricted by MHC; (2) In view of the fact that many tumor cells express the same tumor antigen, they are targeted at a certain type of tumor. Once the CAR gene construction of the antigen is completed, it can be widely used; (3) CAR can use both tumor protein antigens and glycolipid non-protein antigens, expanding the target range of tumor antigens; (4) using the patient's own body Cells reduce the risk of rejection; (5) CAR-T cells have immune memory function and can survive in the body for a long time.
  • the CAR of the present invention comprises (i) an extracellular domain, which comprises an antibody single-chain variable region sequence targeting a tumor cell surface antigen; (ii) a transmembrane domain; (iii) a costimulatory factor; and (iv) the signal transduction domain of CD3 ⁇ ; and; (v) self-cleaving protein; (vi) immunomodulatory molecules (such as GITRL, 4-1BBL, CD40, LIGHT, B7.1, B7.2, OX40L, CD70) .
  • immunomodulatory molecules such as GITRL, 4-1BBL, CD40, LIGHT, B7.1, B7.2, OX40L, CD70.
  • CAR-NK cells Chimeric antigen receptor NK cells
  • CAR-NK cell As used herein, the terms “CAR-NK cell”, “CAR-NK”, and “CAR-NK cell of the present invention” all refer to the CAR-NK cell of the present invention.
  • the CAR-NK cells of the present invention can target tumor surface antigens (such as CD19, PSMA) and are used to treat tumors with high expression or positive tumor cell surface antigens (such as CD19, PSMA), especially solid tumors.
  • Natural killer (NK) cells are a major type of immune effector cells that protect the body from virus infection and tumor cell invasion through non-antigen-specific ways.
  • the engineered (gene modified) NK cells may acquire new functions, including the ability to specifically recognize tumor antigens and enhanced anti-tumor cytotoxicity.
  • CAR-NK cells Compared with autologous CAR-T cells, CAR-NK cells also have the following advantages, for example: (1) They directly kill tumor cells by releasing perforin and granzyme, but have no killing effect on normal cells in the body; (2) They release A small amount of cytokines reduces the risk of cytokine storm; (3) It is easy to expand and develop into "off-the-shelf" products in vitro. Otherwise, it is similar to CAR-T cell therapy.
  • T cell receptor T cell receptor
  • retroviruses are vectors that are transferred exogenously into TCR in T cells.
  • T cells modified by exogenous TCR can specifically recognize and kill tumor cells, and by optimizing the affinity of TCR and tumor-specific antigens, the affinity of T cells and tumors can be improved, and the anti-tumor effect can be improved.
  • the nucleic acid sequence encoding the desired molecule can be obtained using recombinant methods known in the art, such as, for example, by screening a library from cells expressing the gene, by obtaining the gene from a vector known to include the gene, or by using standard Technology to separate directly from the cells and tissues that contain the gene.
  • the gene of interest can be produced synthetically.
  • the present invention also provides a vector into which the expression cassette of the present invention is inserted.
  • Vectors derived from retroviruses such as lentiviruses are suitable tools to achieve long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in daughter cells.
  • Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses such as murine leukemia virus because they can transduce non-proliferating cells, such as hepatocytes. They also have the advantage of low immunogenicity.
  • the expression cassette or nucleic acid sequence of the present invention is usually operably linked to a promoter and incorporated into an expression vector.
  • the vector is suitable for replication and integration of eukaryotic cells.
  • a typical cloning vector contains transcription and translation terminators, initial sequences, and promoters that can be used to regulate the expression of the desired nucleic acid sequence.
  • the expression construct of the present invention can also use standard gene delivery protocols for nucleic acid immunization and gene therapy. Methods of gene delivery are known in the art. See, for example, U.S. Patent Nos. 5,399,346, 5,580,859, 5,589,466, which are hereby incorporated by reference in their entirety.
  • the present invention provides a gene therapy vector.
  • the nucleic acid can be cloned into many types of vectors.
  • the nucleic acid can be cloned into such vectors, which include, but are not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids.
  • Specific vectors of interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.
  • the expression vector can be provided to the cell in the form of a viral vector.
  • Viral vector technology is well known in the art and is described in, for example, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York) and other virology and molecular biology manuals.
  • Viruses that can be used as vectors include, but are not limited to, retrovirus, adenovirus, adeno-associated virus, herpes virus, and lentivirus.
  • a suitable vector contains an origin of replication that functions in at least one organism, a promoter sequence, a convenient restriction enzyme site, and one or more selectable markers (e.g., WO01/96584; WO01/29058; and U.S. Patent No. 6,326,193).
  • retroviruses provide a convenient platform for gene delivery systems.
  • the selected gene can be inserted into a vector and packaged into retroviral particles using techniques known in the art.
  • the recombinant virus can then be isolated and delivered to target cells in vivo or in vitro.
  • Many retroviral systems are known in the art.
  • adenovirus vectors are used.
  • Many adenovirus vectors are known in the art.
  • a lentiviral vector is used.
  • promoter elements can regulate the frequency of transcription initiation. Generally, these are located in the 30-110 bp region upstream of the start site, although it has recently been shown that many promoters also contain functional elements downstream of the start site.
  • the spacing between promoter elements is often flexible in order to maintain promoter function when the elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased by 50 bp before the activity begins to decrease.
  • tk thymidine kinase
  • a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence.
  • the promoter sequence is a strong constitutive promoter sequence capable of driving high-level expression of any polynucleotide sequence operably linked to it.
  • Another example of a suitable promoter is elongation growth factor-1 ⁇ (EF-1 ⁇ ).
  • constitutive promoter sequences can also be used, including but not limited to the simian virus 40 (SV40) early promoter, mouse breast cancer virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, avian leukemia virus promoter, Epstein-Barr virus immediate early promoter, Ruth sarcoma virus promoter, and human gene promoters, such as but not limited to actin promoter , Myosin promoter, heme promoter and creatine kinase promoter.
  • the present invention should not be limited to the application of constitutive promoters. Inducible promoters are also considered part of the invention.
  • an inducible promoter provides a molecular switch that can turn on expression of a polynucleotide sequence operably linked to an inducible promoter when such expression is desired, or turn off expression when expression is undesirable.
  • inducible promoters include, but are not limited to, metallothionein promoter, glucocorticoid promoter, progesterone promoter and tetracycline promoter.
  • the expression vector introduced into the cell may also contain either or both of the selectable marker gene or the reporter gene, so as to facilitate the search for the cell population to be transfected or infected by the viral vector.
  • the selectable marker can be carried on a single piece of DNA and used in the co-transfection procedure. Both the selectable marker and the reporter gene can be flanked by appropriate regulatory sequences so that they can be expressed in the host cell.
  • Useful selectable markers include, for example, antibiotic resistance genes such as neo and the like.
  • Reporter genes are used to identify potentially transfected cells and to evaluate the functionality of regulatory sequences.
  • a reporter gene is a gene that does not exist in or is expressed by a recipient organism or tissue, and it encodes a polypeptide whose expression is clearly indicated by some easily detectable properties such as enzyme activity. After the DNA has been introduced into the recipient cell, the expression of the reporter gene is measured at an appropriate time.
  • Suitable reporter genes may include genes encoding luciferase, ⁇ -galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein (e.g., Ui-Tei et al., 2000 FEBS Letters 479:79 -82).
  • Suitable expression systems are well known and can be prepared using known techniques or obtained commercially. Generally, a construct with a minimum of 5 flanking regions that shows the highest level of reporter gene expression is identified as a promoter. Such a promoter region can be linked to a reporter gene and used to evaluate the ability of the reagent to regulate the promoter-driven transcription.
  • the vector can be easily introduced into a host cell by any method in the art, for example, a mammalian, bacterial, yeast, or insect cell.
  • the expression vector can be transferred into the host cell by physical, chemical or biological means.
  • Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and so on. Methods of producing cells including vectors and/or exogenous nucleic acids are well known in the art. See, for example, Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York). The preferred method for introducing polynucleotides into host cells is calcium phosphate transfection.
  • Biological methods for introducing polynucleotides of interest into host cells include the use of DNA and RNA vectors.
  • Viral vectors especially retroviral vectors, have become the most widely used method of inserting genes into mammalian, e.g., human cells.
  • Other viral vectors can be derived from lentivirus, poxvirus, herpes simplex virus I, adenovirus, adeno-associated virus, and so on. See, for example, U.S. Patent Nos. 5,350,674 and 5,585,362.
  • colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, and beads
  • lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and lipids Plastid.
  • Exemplary colloidal systems used as delivery vehicles in vitro and in vivo are liposomes (e.g., artificial membrane vesicles).
  • an exemplary delivery vehicle is a liposome.
  • lipid formulations to introduce nucleic acids into host cells (in vitro, ex vivo, or in vivo).
  • the nucleic acid can be associated with lipids.
  • Lipid-associated nucleic acids can be encapsulated in the aqueous interior of liposomes, dispersed in the lipid bilayer of liposomes, and attached via linking molecules associated with both liposomes and oligonucleotides
  • the lipid, lipid/DNA or lipid/expression vector associated with the composition is not limited to any specific structure in the solution.
  • Lipids are fatty substances, which can be naturally occurring or synthetic lipids.
  • lipids include fat droplets, which occur naturally in the cytoplasm and in such compounds containing long-chain aliphatic hydrocarbons and their derivatives such as fatty acids, alcohols, amines, amino alcohols, and aldehydes.
  • the vector is a lentiviral vector.
  • the present invention provides a CAR according to the first aspect of the present invention, a nucleic acid molecule according to the second aspect of the present invention, a vector according to the third aspect of the present invention, or a host cell according to the fourth aspect of the present invention, and A pharmaceutically acceptable carrier, diluent or excipient.
  • the formulation is a liquid formulation.
  • the preparation is an injection.
  • the concentration of the CAR-T cells in the preparation is 1 ⁇ 10 3 -1 ⁇ 10 8 cells/Kg body weight, more preferably 1 ⁇ 10 6 -1 ⁇ 10 7 cells/Kg body weight.
  • the formulation may include buffers such as neutral buffered saline, sulfate buffered saline, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; protein; polypeptides or amino acids such as glycine ; Antioxidant; Chelating agent such as EDTA or glutathione; Adjuvant (for example, aluminum hydroxide); and Preservative.
  • buffers such as neutral buffered saline, sulfate buffered saline, etc.
  • carbohydrates such as glucose, mannose, sucrose or dextran, mannitol
  • protein polypeptides or amino acids such as glycine
  • Antioxidant such as EDTA or glutathione
  • Adjuvant for example, aluminum hydroxide
  • Preservative for example, aluminum hydroxide
  • the present invention includes therapeutic applications with cells (e.g., T cells) transduced with a lentiviral vector (LV) encoding the expression cassette of the present invention.
  • the transduced T cells can target tumor cell markers (such as CD19, and/or PSMA) proteins to synergistically activate T cells and cause cellular immune responses, thereby significantly improving their killing efficiency on tumor cells from malignant tumors.
  • tumor cell markers such as CD19, and/or PSMA
  • the present invention also provides a method for stimulating a T cell-mediated immune response to a target cell population or tissue of a mammal, which comprises the following steps: administering the CAR-T cell of the present invention to the mammal.
  • the present invention includes a type of cell therapy in which the patient's autologous T cells (or heterologous donors) are isolated, activated and genetically modified to produce CAR-T cells, and then injected into the same patient.
  • the probability of suffering from graft-versus-host disease is extremely low, and the antigen is recognized by T cells in a non-MHC-restricted manner.
  • one CAR-T can treat all cancers that express the antigen.
  • CAR-T cells can replicate in vivo, producing long-term persistence that can lead to sustained tumor control.
  • the CAR-T cells of the present invention can undergo stable T cell expansion in vivo and last for an extended amount of time.
  • the CAR-mediated immune response can be part of an adoptive immunotherapy step in which CAR-modified T cells induce an immune response specific to the antigen binding domain in the CAR.
  • CAR-T cells that are tumor cell markers such as CD19, and/or PSMA
  • cause a specific immune response against cells expressing tumor cell markers such as CD19, and/or PSMA).
  • Cancers that can be treated include tumors that have not been vascularized or have not been substantially vascularized, as well as vascularized tumors.
  • the cancer may include non-solid tumors (such as hematological tumors such as leukemia and lymphoma) or may include solid tumors.
  • the types of cancer treated with the CAR of the present invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignant tumors, such as sarcoma, carcinoma, and melanoma. It also includes adult tumors/cancers and childhood tumors/cancers.
  • Hematological cancer is cancer of the blood or bone marrow.
  • leukemias include leukemias, including acute leukemias (such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia and myeloblastic, promyelocytic, myelomonocytic type , Monocytic and erythroleukemia), chronic leukemia (such as chronic myeloid (granulocyte) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin’s disease, non- Hodgkin's lymphoma (painless and high-grade form), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia, and myelodysplasia.
  • acute leukemias such as acute lymphoblastic leukemia, acute myeloid leuk
  • a solid tumor is an abnormal mass of tissue that does not usually contain a cyst or fluid area.
  • Solid tumors can be benign or malignant. Different types of solid tumors are named after the cell type that formed them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcoma and cancer include fibrosarcoma, myxosarcoma, liposarcoma, mesothelioma, lymphoid malignancies, pancreatic cancer, and ovarian cancer.
  • the CAR-modified T cells of the present invention can also be used as a type of vaccine for ex vivo immunity and/or in vivo therapy of mammals.
  • the mammal is a human.
  • cells are isolated from mammals (preferably humans) and genetically modified (ie, transduced or transfected in vitro) with a vector expressing the CAR disclosed herein.
  • CAR-modified cells can be administered to mammalian recipients to provide therapeutic benefits.
  • the mammalian recipient can be a human, and the CAR-modified cell can be autologous relative to the recipient.
  • the cell may be allogeneic, syngeneic, or xenogeneic relative to the recipient.
  • the present invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient.
  • the present invention provides a method for treating tumors, which comprises administering to a subject in need thereof a therapeutically effective amount of the CAR-modified T cells of the present invention.
  • the CAR-modified T cells of the present invention can be administered alone or as a pharmaceutical composition in combination with a diluent and/or with other components or other cytokines or cell populations.
  • the pharmaceutical composition of the present invention may include the target cell population as described herein in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients.
  • compositions may include buffers such as neutral buffered saline, sulfate buffered saline, etc.; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelate Mixtures such as EDTA or glutathione; adjuvants (for example, aluminum hydroxide); and preservatives.
  • buffers such as neutral buffered saline, sulfate buffered saline, etc.
  • carbohydrates such as glucose, mannose, sucrose or dextran, mannitol
  • proteins polypeptides or amino acids
  • antioxidants such as glycine
  • chelate Mixtures such as EDTA or glutathione
  • adjuvants for example, aluminum hydroxide
  • preservatives for example, aluminum hydroxide
  • the pharmaceutical composition of the present invention can be administered in a manner suitable for the disease to be treated (or prevented).
  • the number and frequency of administration will be determined by factors such as the patient's condition, and the type and severity of the patient's disease-although the appropriate dosage can be determined by clinical trials.
  • the precise amount of the composition of the present invention to be administered can be determined by the physician, who considers the patient (subject ) Individual differences in age, weight, tumor size, degree of infection or metastasis, and disease. May generally indicated: including those described herein, the pharmaceutical compositions of T cells may be 104 to 109 doses cells / kg body weight, preferably 105 to 106 cells / kg body weight doses (including all integers within that range Value) application. The T cell composition can also be administered multiple times at these doses.
  • the cells can be administered by using injection techniques well known in immunotherapy (see, for example, Rosenberg et al., New Eng. J. of Med. 319:1676, 1988).
  • the optimal dosage and treatment regimen for a specific patient can be easily determined by those skilled in the medical field by monitoring the patient's signs of disease and adjusting the treatment accordingly.
  • the administration of the subject composition can be carried out in any convenient manner, including by spraying, injection, swallowing, infusion, implantation, or transplantation.
  • the compositions described herein can be administered to patients subcutaneously, intracutaneously, intratumorally, intranodal, intraspinal, intramuscular, by intravenous (i.v.) injection, or intraperitoneally.
  • the T cell composition of the present invention is administered to the patient by intradermal or subcutaneous injection.
  • the T cell composition of the present invention is preferably administered by i.v. injection.
  • the composition of T cells can be injected directly into tumors, lymph nodes or sites of infection.
  • the treatment modality includes, but is not limited to, treatment with the following agents: the agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known It is ARA-C) or natalizumab treatment for MS patients or erfaizumab treatment for psoriasis patients or other treatments for PML patients.
  • the agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known It is ARA-C) or natalizumab treatment for MS patients or erfaizumab treatment for psoriasis patients or other treatments for PML patients.
  • the T cells of the present invention can be used in combination with chemotherapy, radiation, immunosuppressants, such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil, and FK506, antibodies Or other immunotherapeutics.
  • the cell composition of the present invention is administered to bone marrow transplantation, using chemotherapeutic agents such as fludarabine, external beam radiotherapy (XRT), cyclophosphamide (for example, before, simultaneously, or after). patient.
  • chemotherapeutic agents such as fludarabine, external beam radiotherapy (XRT), cyclophosphamide (for example, before, simultaneously, or after).
  • the subject may undergo the standard treatment of high-dose chemotherapy followed by peripheral blood stem cell transplantation.
  • the subject receives an infusion of the expanded immune cells of the invention.
  • the expanded cells are administered before or after surgery.
  • the dosage of the above treatment administered to the patient will vary with the precise nature of the condition being treated and the recipient of the treatment.
  • the dosage ratio for human administration can be implemented according to the practice accepted in the art.
  • 1 ⁇ 10 6 to 1 ⁇ 10 10 modified T cells of the present invention can be injected into each treatment or course of treatment by, for example, intravenous infusion, Apply to the patient.
  • the engineered immune cells of the present invention can specifically target the antibody single-chain variable region sequence of tumor cell surface antigens (such as CD19, PSMA), thereby efficiently killing tumors (especially solid tumors).
  • tumor cell surface antigens such as CD19, PSMA
  • CARs expressing immunomodulatory molecules can more specifically kill tumor cells, especially tumor cells with high expression or positive CD19 and/or PSMA.
  • the present invention found for the first time that in CAR-modified T cells or NK cells, expression of exogenous immunomodulatory molecules (such as GITRL) together with CAR can significantly improve tumor suppressor activity and has a synergistic effect.
  • exogenous immunomodulatory molecules such as GITRL
  • the present invention developed a new chimeric antigen receptor molecule for the first time. After the CAR molecule is expressed on T cells, it can effectively resist the inhibitory effect of the tumor immune microenvironment, and maintain or improve the effector function of CAR-T cells. Expansion capacity and continuity capacity, in the treatment of solid tumors represented by prostate cancer, show a significantly better therapeutic effect than the existing CAR structure.
  • the present invention finds for the first time that the expression sequence of tumor necrosis factor receptor (GITR) ligand GITRL induced by glucocorticoids is expressed in CAR molecules and expressed in a fusion manner in a 2A connection. Since activated T cells highly express GITR, the co-expressed GITRL enhances their respective effector functions through the mutual cooperation between CAR-T cells.
  • GITR tumor necrosis factor receptor
  • the present invention found for the first time that the expression of GITRL can significantly promote the differentiation of Th9 subgroups in CAR-T cells, while inhibiting the formation of regulatory T cells Treg, significantly improve the subgroup composition of CAR-T cells, and show stronger Anti-tumor effect function.
  • the present invention introduces the important immunomodulatory molecule GITRL into the CAR molecule for the first time, and it is co-expressed with CAR through 2A connection.
  • This molecule promotes the activation of CAR-T cells and enhances the differentiation of CD4+ T cells into Th9, and at the same time inhibits The differentiation and formation of Treg T cells play an important role in overcoming the immunosuppressive microenvironment of solid tumors. It is significantly better than the existing CAR-T technology in the treatment of prostate cancer CAR-T targeting PSMA.
  • the immunotherapy of solid tumors has huge application potential.
  • the CAR-T cells co-expressing GITRL provided by the present invention have the function of regulating the function of other immune cells expressing its receptor GITR.
  • the immune cells include but are not limited to B cells, macrophages, and natural immune cells. Killer cells, granulocytes, and mast cells can enhance the anti-tumor immune response of patients by comprehensively and multi-directionally regulating the tumor immune microenvironment.
  • the GITRL-CAR was constructed in the following order ( Figure 1): antigen recognition area ⁇ junction/transmembrane area ⁇ 4-1BB costimulation ⁇ CD3 ⁇ ⁇ P2A ⁇ GITRL coding sequence, and then integrated into the 5 and 3 end LTR of the pELPS lentiviral vector Between the sequences, the CAR expression master plasmid is constructed, followed by lentivirus packaging.
  • the plasmid mixture is evenly added dropwise to the 293T cells used for transfection, and cultured in an incubator at 37°C and 5% CO2. After culturing for 6-8 hours, the cells were replaced with fresh complete medium DMEM (denoted as transfection 0h). Continue to incubate at 37°C and 5% CO2.
  • the cell supernatants were collected into clean 50ml centrifuge tubes, labeled, and 10mL complete medium DMEM was added to the petri dish, and the culture was continued at 37°C and 5% CO2. After 72 hours of transfection, the cell culture supernatant was collected again, and the collected virus stock solution was placed in a 250 ml centrifuge tube. In the process of continuing the culture, regularly observe the cell growth state to ensure the cell toxin production efficiency.
  • HEK-293T cells Take HEK-293T cells in good growth condition, count them after digestion, spread them evenly into a 24-well plate according to 2*10 ⁇ 5 wells, culture for about 8 hours until the cells adhere to the wall, press 3 times into the cell culture supernatant of the 24-well plate
  • the dilution gradient was added with different volumes of virus concentrates, a total of 8 gradients were set, and the infection was carried out in the incubator for 48 hours. After 48 hours of infection, the cells were digested and collected.
  • Titer number of cells at the time of plating*percentage of positive cells*1000/volume of virus solution added ( ⁇ L)TU/mL.
  • X-VIVO complete medium X-VIVO basic medium+10%FBS+1%P/S+10 ⁇ g/mLIL-2
  • the positively infected T cells were the target CAR-T cells.
  • Example 2 GITRL-CART cell luciferase killing detection and amplification detection
  • the co-culture experiment can refer to the following table.
  • target cell efficiency target ratio
  • the co-cultivation experiment has a target ratio of 2:1 as shown in Table 1.
  • Killing rate calculation formula (target cell fluorescence value-effector cell fluorescence value-target cell and effector cell co-incubation fluorescence value) / (target cell fluorescence value-effector cell fluorescence value) * 100%.
  • three identical 96-well plates one is an ultra-low adsorption 96-well plate, and the other two are normal plates).
  • the wall 96-well plates are numbered as plate one, plate two, plate three)
  • the cells in the plate 6 were killed according to the cell luciferase killing assay method to calculate the killing rate to the target cells, which is the three-round killing rate.
  • control CART and GITRL-CART cells obtained by culturing for two days after infection with the corresponding virus and the T cells obtained from the same batch were cultured strictly in accordance with the same culture conditions (x-vivo medium + 10% FBS + 1% P /S+10 ⁇ g/mL IL-2).
  • GITRL-CART cells are incubated with target cells and then expanded
  • the numbers of target cells and effector cells are 1*10 ⁇ 6 and 2*10 ⁇ , respectively. 6 pieces, add X-VIVO complete medium and put them in a six-well plate for culture
  • Example 3 Detection of IL-9 cytokine expression after GITRL-CART cells and target cells are co-cultured
  • Reading the plate within 10 minutes after termination, use the detection wavelength of 450nm to read the value.
  • the evaluation tool mice used in this example are 6-8 week old NSG mice, raised in an SPF-class laminar flow room, standard pellets, litter and other items related to mice are sterilized. Carry out the in vivo function evaluation of GITRL-CART according to the following steps.
  • mice Select 6-8 weeks old NSG mice, each mouse is intraperitoneally injected with 400 ⁇ L of sodium pentobarbital to anesthetize the mice, and remove the hair on the back of the mice with a razor.
  • mice Fifteen days after the target cell injection, use the in vivo imager to perform in vivo imaging of the mice to observe the tumor growth, and eliminate the unsuccessful mouse models based on the imaging results. The remaining mice are randomly grouped according to the experimental arrangement and marked .
  • mice Weigh the substrate D-Luciferin potassium salt, dissolve it in PBS, store it in the dark, and enter the SPF-level system through the delivery window.
  • the substrate was injected intraperitoneally according to the mouse body weight of 3m/each, and the mice were subjected to isoflurane anesthesia after 3 minutes. After the mice are anesthetized, they are put into the imager for imaging and taking pictures, the pictures are counted and processed, and the fluorescence values read are recorded at the same time.
  • mice will be imaged in vivo again, and the mouse body weight will be included.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Mycology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Plant Pathology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Developmental Biology & Embryology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Hematology (AREA)
  • Reproductive Health (AREA)
  • Gynecology & Obstetrics (AREA)

Abstract

La présente invention concerne une préparation et des applications d'une cellule T de récepteur antigénique chimérique (CAR) coexprimant une molécule immunomodulatrice. La présente invention concerne plus particulièrement un CAR et la cellule CAR-T de celui-ci ciblant plus précisément un antigène de surface de cellule tumorale, le récepteur antigénique chimérique (CAR) comprenant un domaine structurel de liaison à l'antigène et une molécule immunomodulatrice. La cellule CAR-T de la présente invention présente d'excellents effets d'élimination des tumeurs.
PCT/CN2020/138691 2019-12-31 2020-12-23 Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice WO2021136040A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911422121.4 2019-12-31
CN201911422121.4A CN113087805A (zh) 2019-12-31 2019-12-31 一种共表达免疫调节分子的嵌合抗原受体t细胞的制备及其应用

Publications (1)

Publication Number Publication Date
WO2021136040A1 true WO2021136040A1 (fr) 2021-07-08

Family

ID=76663184

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/138691 WO2021136040A1 (fr) 2019-12-31 2020-12-23 Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice

Country Status (2)

Country Link
CN (1) CN113087805A (fr)
WO (1) WO2021136040A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113651893A (zh) * 2021-08-12 2021-11-16 上海生物制品研究所有限责任公司 联合her2和meso双靶点car-t载体及其构建方法和在癌症中的应用
EP4008404A4 (fr) * 2019-08-01 2023-03-01 Mie University Récepteur d'antigène
US20230251262A1 (en) * 2022-02-09 2023-08-10 Universitaesklinikum Hamburg-Eppendorf Enrichment, detection and characterization of circulating tumor cells with susd2 and enpp1

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106467906A (zh) * 2015-08-20 2017-03-01 北京马力喏生物科技有限公司 构建体、转基因淋巴细胞及其制备方法和用途

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013051718A1 (fr) * 2011-10-07 2013-04-11 国立大学法人三重大学 Récepteur d'antigène chimérique
JP6450690B2 (ja) * 2013-02-15 2019-01-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア キメラ抗原受容体及びその使用方法
CN104177499B (zh) * 2013-05-27 2019-01-08 上海雅科生物科技有限公司 一种嵌合抗原受体、编码基因、表达载体及其应用
US20190194617A1 (en) * 2017-12-22 2019-06-27 Cell Design Labs, Inc. Single- and multi-chain chimeric antigen receptors
EP3790629A1 (fr) * 2018-05-11 2021-03-17 CRISPR Therapeutics AG Procédés et compositions pour le traitement du cancer
US11866494B2 (en) * 2018-08-31 2024-01-09 Innovative Cellular Therapeutics Holdings, Ltd. CAR T therapy through uses of co-stimulation
CN110564695A (zh) * 2019-09-23 2019-12-13 华东师范大学 靶向前列腺癌的增强型car-t细胞及其制备方法和药物

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106467906A (zh) * 2015-08-20 2017-03-01 北京马力喏生物科技有限公司 构建体、转基因淋巴细胞及其制备方法和用途

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4008404A4 (fr) * 2019-08-01 2023-03-01 Mie University Récepteur d'antigène
CN113651893A (zh) * 2021-08-12 2021-11-16 上海生物制品研究所有限责任公司 联合her2和meso双靶点car-t载体及其构建方法和在癌症中的应用
CN113651893B (zh) * 2021-08-12 2023-08-01 上海生物制品研究所有限责任公司 联合her2和meso双靶点car-t载体及其构建方法和在癌症中的应用
US20230251262A1 (en) * 2022-02-09 2023-08-10 Universitaesklinikum Hamburg-Eppendorf Enrichment, detection and characterization of circulating tumor cells with susd2 and enpp1

Also Published As

Publication number Publication date
CN113087805A (zh) 2021-07-09

Similar Documents

Publication Publication Date Title
US11142581B2 (en) BCMA-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
CN109306016B (zh) 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
WO2019062817A1 (fr) Cellule immunitaire modifiée capable d'induire la sécrétion d'anticorps anti-cd47
WO2018145649A1 (fr) Construction d'un récepteur d'antigène chimère ciblant un antigène cd20 et identification d'activité de lymphocytes t génétiquement modifiées de celui-ci
WO2021136040A1 (fr) Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice
US20230159636A1 (en) Cd7 chimeric antigen receptor-modified nk-92mi cell and use thereof
US20240082401A1 (en) Bispecific cs1-bcma car-t cell and application thereof
WO2023046110A1 (fr) Cellules immunitaires artificielles co-exprimant ccr2b, leur preparation et leur application
CN113087806B (zh) 靶向多种肿瘤的新型car-t细胞及其制备和方法
WO2018145648A1 (fr) Construction de car ciblant cd20 et identification d'activité de lymphocytes t génétiquement modifiées de celui-ci
CN113755448B (zh) 联合表达CCR2b和CD40L的工程化免疫细胞及其制备和应用
CN111378625A (zh) 一种cxcl13趋化型car-t细胞的制备和应用
WO2018068766A1 (fr) Récepteur antigénique chimérique ciblant le cd19, son procédé de préparation et son application
WO2021239020A1 (fr) Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci
WO2020151752A1 (fr) Cellules immunitaires modifiées ciblant une combinaison cd20
CN109897114B (zh) 具有自杀基因开关的靶向cd47的工程化免疫细胞
US20230144447A1 (en) Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
CN115806626A (zh) 一种基于csf1的嵌合抗原受体免疫细胞制备及其应用
CN115819614A (zh) 一种基于il34的嵌合抗原受体免疫细胞制备及其应用
CN114685683A (zh) 靶向gd2的car-t细胞及其制备和应用
WO2022151959A1 (fr) Cellule car t ciblant b7-h3 et son utilisation dans le traitement de la leucémie aiguë myéloïde
CN114716564B (zh) 基于sectm1构建的嵌合抗原受体免疫细胞制备及其应用
CN116496397B (zh) 靶向cd19 car-t细胞人源化抗体
WO2023246908A1 (fr) Préparation et utilisation d'une cellule immunitaire à récepteur antigénique chimérique ciblant csf1r
EP4357369A1 (fr) Préparation et utilisation d'une cellule immunitaire de récepteur antigénique chimérique construite sur la base de granzyme b

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: 20910347

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20910347

Country of ref document: EP

Kind code of ref document: A1