WO2021239020A1 - Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci - Google Patents

Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci Download PDF

Info

Publication number
WO2021239020A1
WO2021239020A1 PCT/CN2021/096191 CN2021096191W WO2021239020A1 WO 2021239020 A1 WO2021239020 A1 WO 2021239020A1 CN 2021096191 W CN2021096191 W CN 2021096191W WO 2021239020 A1 WO2021239020 A1 WO 2021239020A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
psma
cell
tumor
Prior art date
Application number
PCT/CN2021/096191
Other languages
English (en)
Chinese (zh)
Inventor
张娜
刘小红
杜冰
张楫钦
刘明耀
席在喜
Original Assignee
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 上海邦耀生物科技有限公司 filed Critical 上海邦耀生物科技有限公司
Publication of WO2021239020A1 publication Critical patent/WO2021239020A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • 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
    • A61K39/464441Interferons [IFN]
    • 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
    • A61K39/464474Proteoglycans, e.g. glypican, brevican or CSPG4
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • 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
    • 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
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • 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
    • C07K16/3007Carcino-embryonic Antigens
    • 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
    • C07K16/303Liver or Pancreas
    • 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
    • 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
    • C07K16/3076Immunoglobulins [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 against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [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 against structure-related tumour-associated moieties against tumour-associated gangliosides
    • 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
    • C07K16/3076Immunoglobulins [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 against structure-related tumour-associated moieties
    • C07K16/3092Immunoglobulins [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 against structure-related tumour-associated moieties against tumour-associated mucins
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal 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
    • 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
    • A61K2239/54Pancreas
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • 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
    • 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

Definitions

  • the invention belongs to the field of biotechnology. Specifically, the present invention relates to an immunotherapy method combining chimeric antigen receptor and type I interferon and its application.
  • Malignant tumors have become one of the main public health problems that seriously threaten people's health, and increasingly become the main risk factor for death of residents.
  • the incidence of tumors in my country has continued to rise for many years. Because of its high mortality rate, more than one-fifth of deaths are caused by malignant tumors, and it has become the leading cause of death.
  • tumor immunotherapy mainly uses immune cells as the research object. It enhances the body's anti-tumor immune response by regulating the tumor microenvironment, innate immunity, and adaptive immunity, and has the potential for long-term benefits.
  • PD-1/programmed cell death-ligand1 (PDL1) inhibitors in immune checkpoint inhibitors have achieved success in tumor treatment
  • a huge success it can block tumor escape, activate its own immune system, and improve the body's anti-tumor immunity, thereby achieving the purpose of inhibiting and killing tumor cells, providing a new direction for cancer immunotherapy, but it may be due to tumor infiltration Low infiltration of sexual lymphocytes, difficulty in immune activation, and relative lack of PD-L1 expression in cancer tissues.
  • the effect in clinical trials is not optimistic.
  • Objective response (OR) rate Low Low. Therefore, it is still an unmet need to develop or upgrade new immunotherapies to enhance the therapeutic effect of solid tumors.
  • CAR-T manually replenishes "ammunition", although it has shown great relief in hematoma, it still faces exhaustion, which is manifested by proliferation and continuous decline.
  • the abnormal blood vessels promoted by tumors make it difficult for immune cells to infiltrate into tumor tissues, which may affect the formation of anti-tumor "base"-TLS, so it is impossible to obtain signals from innate immune cells such as DC and B cells.
  • the immunosuppressive tumor microenvironment Second, the immunosuppressive tumor microenvironment.
  • the purpose of the present invention is to provide a new type of chimeric antigen receptor T cells, which can directly act on tumor cells to inhibit and kill tumor cells, and can also regulate the innate immune response, thereby promoting antigen presentation and killing cell functions.
  • the first aspect of the present invention provides an engineered immune cell that expresses a chimeric antigen receptor CAR targeting tumor cell markers and type I interferon, the tumor cell marker Selected from the following group: PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof.
  • the tumor cell marker selected from the following group: PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof.
  • the immune cells are NK cells, macrophages or T cells, preferably T cells.
  • the chimeric antigen receptor CAR is located on the cell membrane of the immune cell.
  • the chimeric antigen receptor CAR contains an antigen binding domain that targets tumor cell markers.
  • 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.
  • the "-" is a connecting peptide or a peptide bond
  • L is no or signal peptide sequence
  • S is an antigen binding domain targeting tumor cell markers, which are selected from the following group: PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof;
  • H is no or hinge area
  • TM is the transmembrane domain
  • C is a costimulatory signal molecule
  • CD3 ⁇ is a cytoplasmic signal transduction sequence derived from CD3 ⁇ .
  • the antigen binding domain of the tumor cell marker includes a single-chain variable region sequence of an antibody targeting the tumor cell marker.
  • the structure of the single-chain variable region sequence of the antibody targeting tumor cell marker is as shown in formula A1 or A2:
  • V L1 is the light chain variable region of the anti-tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-tumor cell marker antibody
  • "-" 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 by GGGGS.
  • amino acid sequence of the flexible linker is shown in SEQ ID NO.: 15.
  • amino acid sequence of V L1 is shown in SEQ ID NO.:1, positions 22-128, and the amino acid sequence of V H1 is shown in SEQ ID NO.:1, positions 144-258.
  • amino acid sequence of V L1 is shown in SEQ ID NO.: 49, positions 22-134
  • amino acid sequence of V H1 is shown in SEQ ID NO.: 49, positions 150-264.
  • amino acid sequence of V L1 is shown in SEQ ID NO.: 56 at positions 22 to 131
  • amino acid sequence of V H1 is shown in SEQ ID NO.: 56 at positions 146 to 272.
  • amino acid sequence of V L1 is as shown in SEQ ID NO.: 2 or 51 or 58.
  • amino acid sequence of V H1 is as shown in SEQ ID NO.: 3 or 53 or 60.
  • sequence of the single-chain variable region of the antibody targeting tumor cell markers is murine, human, human and murine chimeric, or fully humanized single-chain antibody variable. District fragments.
  • the L is a signal peptide of a protein selected from the group consisting of CD8a, CD8, CD28, GM-CSF, CD4, CD137, or a combination thereof.
  • the L is a signal peptide derived from CD8a.
  • nucleotide sequence of L is shown in SEQ ID NO.: 12.
  • amino acid sequence of L is shown in SEQ ID NO.:4.
  • the H is a hinge region of a protein selected from the group consisting of CD8, Ig (immunoglobulin) hinge, or a combination thereof.
  • the H is a hinge region derived from CD8.
  • amino acid sequence of H is shown in SEQ ID NO.:5.
  • the TM is a transmembrane region of a protein selected from the group consisting of CD8a, CD8, CD28, CD33, CD37, CD8 ⁇ , CD5, CD16, ICOS, CD9, CD22, CD134, CD137, CD154, CD19, CD45, CD4, CD3 ⁇ , or a combination thereof.
  • the TM is the transmembrane region derived from CD8a.
  • amino acid sequence of the TM is shown in SEQ ID NO.:6.
  • the C is a costimulatory signal molecule of a protein selected from the group consisting of: OX40, CD28, CD30, CD40, CD70, CD134, 4-1BB (CD137), LIGHT, DAP10, CDS, ICAM- 1. Or a combination thereof.
  • the C is a costimulatory signal molecule derived from 4-1BB.
  • amino acid sequence of C is shown in SEQ ID NO.:7.
  • amino acid sequence of the CD3 ⁇ is shown in SEQ ID NO.: 8.
  • amino acid sequence of the CAR is shown in SEQ ID NO.: 1 or 49 or 56.
  • the type I interferon is selected from the following group: IFN ⁇ 1, IFN ⁇ 2 (including IFN ⁇ 2a, IFN ⁇ 2b, IFN ⁇ 2c), IFN ⁇ 4, IFN ⁇ 5, IFN ⁇ 6, IFN ⁇ 7, IFN ⁇ 8, IFN ⁇ 10, IFN ⁇ 13, IFN ⁇ 14, IFN ⁇ 17, IFN ⁇ 21, IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , or a combination thereof, preferably IFN ⁇ 2b.
  • amino acid sequence of the type I interferon is shown in any one of SEQ ID NO.: 9, 36-48.
  • the second aspect of the present invention provides a method for preparing the engineered immune cells according to the first aspect of the present invention, including the following steps:
  • the immune cells are modified so that the immune cells express the chimeric antigen receptor CAR and type I interferon targeted to tumor cell markers, thereby obtaining the engineering described in the first aspect of the present invention Of immune cells.
  • step (A) it further includes isolating and/or activating the immune cells to be modified.
  • step (B) in step (B), it includes (B1) introducing the first expression cassette expressing the CAR targeting tumor cell marker into the immune cell; and (B2) expressing type I interference
  • the second expression cassette of the protein is introduced into the immune cells; wherein the step (B1) can be performed before, after, at the same time, or alternately after the step (B2).
  • step (B) the first expression cassette and/or the second expression cassette are introduced into the nucleus of the immune cell.
  • step (B1) when the immune cells to be modified in step (A) already express the CAR, step (B1) can be omitted.
  • the immune cells are NK cells, macrophages or T cells.
  • the first expression cassette contains a nucleic acid sequence encoding the chimeric antigen receptor CAR.
  • the second expression cassette contains a nucleic acid sequence encoding type I interferon.
  • first expression cassette and the second expression cassette are located on the same or different vectors.
  • first expression cassette and the second expression cassette are located in the same vector.
  • the vector is a viral vector or a transposon.
  • the vector is selected from the group consisting of DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, other gene transfer systems, or a combination thereof.
  • the vector is a lentiviral vector or a transposon.
  • the method further includes the step of performing function and effectiveness testing on the obtained engineered immune cells.
  • the third aspect of the present invention provides a preparation containing the engineered immune cells described in the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the formulation is a liquid formulation.
  • the dosage form of the preparation includes an injection.
  • the concentration of the engineered immune cells in the preparation is 1 ⁇ 10 3 -1 ⁇ 10 8 cells/ml, preferably 1 ⁇ 10 4 -1 ⁇ 10 7 cells/ml ml.
  • the preparation also contains other drugs for treating cancer or tumors (such as emerging antibody drugs, other CAR-T drugs or chemotherapeutic drugs).
  • other drugs for treating cancer or tumors such as emerging antibody drugs, other CAR-T drugs or chemotherapeutic drugs.
  • the fourth aspect of the present invention provides a use of the engineered immune cells as described in the first aspect of the present invention to prepare drugs or preparations for selectively killing tumors.
  • the tumor includes highly expressing tumor cell markers (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D Body, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138) tumors.
  • tumor cell markers such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D Body, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138
  • the tumor is selected from the group consisting of hematological tumors, solid tumors, or a combination thereof.
  • the tumor is a solid tumor.
  • the hematological tumor is selected from the group consisting of acute myeloid leukemia (AML), multiple myeloma (MM), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), diffuse large B cell lymphoma (DLBCL), or a combination thereof.
  • AML acute myeloid leukemia
  • MM multiple myeloma
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • DLBCL diffuse large B cell lymphoma
  • the tumor includes a solid tumor.
  • the solid tumor is selected from the group consisting of prostate cancer, liver cancer, head and neck cancer, melanoma, non-Hodgkin’s lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, chondrosarcoma , Thyroid Cancer, Kidney Cancer, Mesothelioma, Osteosarcoma, Cholangiocarcinoma, Ovarian Cancer, Gastric Cancer, Bladder Cancer, Meningioma, Pancreatic Cancer, Multiple Squamous Cell Tumor, Esophageal Cancer, Lung Small Cell Carcinoma, Colorectal Cancer, Breast cancer, medulloblastoma, breast cancer, or a combination thereof.
  • the fifth aspect of the present invention provides a kit for selectively killing tumors, the kit containing a container, and in the container:
  • a first nucleic acid sequence said first nucleic acid sequence containing a first expression cassette for expressing a chimeric antigen receptor CAR targeting tumor cell markers, the tumor cell markers being selected from the following group: PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof ;with
  • first and second nucleic acid sequences are independent or connected.
  • first and second nucleic acid sequences are located in the same or different containers.
  • the first and second nucleic acid sequences are located on the same or different vectors.
  • first and second nucleic acid sequences are located in the same vector.
  • the sixth aspect of the present invention provides a method for selectively killing tumors, including:
  • a safe and effective amount of the engineered immune cell according to the first aspect of the present invention or the preparation according to the third aspect of the present invention is administered to a subject in need of treatment.
  • 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 seventh aspect of the present invention provides a method for treating diseases, comprising administering a safe and effective amount of the engineered immune cells according to the first aspect of the present invention or the preparation according to the third aspect of the present invention to a subject in need of treatment.
  • the method further includes administering other drugs for treating cancer or tumor to the subject in need of treatment.
  • the other drugs include CAR-T drugs.
  • the disease is cancer or tumor.
  • the tumor includes highly expressing tumor cell markers (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D Body, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138) tumors.
  • tumor cell markers such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D Body, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138
  • the tumor is selected from the group consisting of hematological tumors, solid tumors, or a combination thereof.
  • the tumor is a solid tumor.
  • the hematological tumor is selected from the group consisting of acute myeloid leukemia (AML), multiple myeloma (MM), chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), diffuse large B cell lymphoma (DLBCL), or a combination thereof.
  • AML acute myeloid leukemia
  • MM multiple myeloma
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • DLBCL diffuse large B cell lymphoma
  • the tumor includes a solid tumor.
  • the solid tumor is selected from the group consisting of prostate cancer, liver cancer, head and neck cancer, melanoma, non-Hodgkin’s lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, chondrosarcoma , Thyroid Cancer, Kidney Cancer, Mesothelioma, Osteosarcoma, Cholangiocarcinoma, Ovarian Cancer, Gastric Cancer, Bladder Cancer, Meningioma, Pancreatic Cancer, Multiple Squamous Cell Tumor, Esophageal Cancer, Lung Small Cell Carcinoma, Colorectal Cancer, Breast cancer, medulloblastoma, breast cancer, or a combination thereof.
  • the eighth aspect of the present invention provides a fusion protein comprising a chimeric antigen receptor CAR targeting tumor cell markers and type I interferon, wherein the tumor cell marker is selected from the group consisting of PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof .
  • the tumor cell marker is selected from the group consisting of PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof .
  • the CAR and the type I interferon are connected by a connecting peptide.
  • the connecting peptide includes a self-cleaving protein.
  • the self-cleaving protein is selected from the group consisting of T2A, P2A, E2A, F2A, or a combination thereof.
  • the self-cleaving protein includes P2A.
  • the structure of the fusion protein is shown in the following formula III:
  • Each "-" is independently a connecting peptide or a peptide bond
  • L is no or signal peptide sequence
  • S is an antigen binding domain targeting tumor cell markers, which are selected from the following group: PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138, or a combination thereof;
  • H is no or hinge area
  • TM is the transmembrane domain
  • C is a costimulatory signal molecule
  • CD3 ⁇ is a cytoplasmic signal transduction sequence derived from CD3 ⁇ ;
  • Z3 is a connecting peptide
  • P is type I interferon
  • n 1, 2, 3, or 4.
  • amino acid sequence of the fusion protein is shown in SEQ ID NO.: 10 or 54 or 61.
  • the ninth aspect of the present invention provides a polynucleotide encoding the fusion protein of the eighth aspect of the present invention.
  • polynucleotide is selected from the following group:
  • polynucleotide sequence is as shown in SEQ ID NO.: 11 or 55 or 62.
  • the tenth aspect of the present invention provides a vector comprising the polynucleotide according to the ninth aspect of the present invention.
  • the vector includes DNA and RNA.
  • the vector is selected from the following group: plasmid, viral vector, transposon, or a combination thereof.
  • the vector includes DNA virus and retroviral vector.
  • the vector is selected from the group consisting of a lentiviral vector, an adenovirus vector, an adeno-associated virus vector, a transposon, or a combination thereof.
  • the vector is a lentiviral vector or a transposon.
  • the vector includes one or more promoters, which are operably linked to the nucleic acid sequence, enhancer, intron, transcription termination signal, polyadenylation sequence, and origin of replication. , Selectable markers, nucleic acid restriction sites, and/or homologous recombination sites.
  • the vector is a vector containing or inserted the polynucleotide of the ninth aspect of the present invention.
  • the vector is used to express the fusion protein according to the eighth aspect of the present invention.
  • Figure 1 shows the structural schematic diagram of the expression elements contained in the first nucleic acid sequence and the second nucleic acid sequence in Example 1.
  • A is the structural schematic diagram of the first nucleic acid sequence targeting the PSMA target
  • B is the structural schematic diagram of the first nucleic acid sequence targeting the PSMA target.
  • a schematic diagram of the structure of the connection between the first nucleic acid and the second nucleic acid of the target is a schematic diagram of the structure of the first nucleic acid sequence targeting the GPC3 target, and D is the schematic diagram of the structure of the connection between the first nucleic acid and the second nucleic acid targeting the GPC3 target ;
  • E is a schematic structural diagram of the first nucleic acid sequence targeting the BCMA target, and
  • F is a schematic structural diagram of the first nucleic acid and the second nucleic acid targeting the BCMA target.
  • Figure 2 shows the flow chart of the virus titer detection of the control viruses PSMA-CAR and IFN ⁇ 2b-CAR.
  • Figure 3 shows the flow chart of the positive detection of PSMA-CART and IFN ⁇ 2b-CART CAR prepared with lentivirus.
  • Figure 4A is a flow diagram of positive detection of PB-PSMA-CART and PB-IFN ⁇ 2b-PSMA-CART CAR prepared by transposon electroporation
  • Fig. 4B is PB-GPC3-CART and PB-IFN ⁇ 2b- prepared by transposon electroporation
  • Figure 4C is the flow chart of the positive detection of PB-BCMA-CART and PB-IFN ⁇ 2b-BCMA-CART CAR prepared by transposon electroporation
  • Figure 4D is the flow diagram of positive detection of PB-BCMA-CART and PB-IFN ⁇ 2b-BCMA-CART CAR prepared by transposon electroporation
  • Figure 5A shows the results of killing PC3-PSMA tumor cells by PB-PSMA-CART and PB-IFN ⁇ 2b-PSMA-CART prepared by transposon electroporation. IFN ⁇ 2b-GPC3-CART kills huh7 tumor cells.
  • Figure 5C is the result of PB-BCMA-CART and PB-IFN ⁇ 2b-BCMA-CART prepared by transposon electroporation to kill mm1s tumor cells.
  • Figure 6 is a graph showing the comparison of proliferation of PB-PSMA-CART and PB-IFN ⁇ 2b-PSMA-CART prepared by transposon electrotransformation.
  • Figure 7 shows the flow cytometric comparison between PB-PSMA-CART and PB-IFN ⁇ 2b-PSMA-CART prepared by transposon electroporation.
  • Figure 8A PB-PSMA-CART and PB-IFN ⁇ 2b-PSMA-CART cell supernatant prepared by transposon electrotransduction to stimulate human PBMC QPCR results
  • Figure 8B PB-PSMA-CART and PB-IFN ⁇ 2b prepared by transposon electrotransduction -Flow cytometric results of TRAIL expression after stimulation of human PBMC by PSMA-CART cell supernatant.
  • Figure 8C is a graph of the killing effect of traditional CART and IFN ⁇ 2b-CART cells prepared by electrotransduction with transposon incubation with PBMC and huh7 tumor cells.
  • FIG. 9 The QPCR results (left) and the results of the migration experiment (right) after stimulating the tumor cells DU145 with the supernatant of PSMA-CART and IFN ⁇ 2b-CART cells prepared by transposon electroporation.
  • tumor cell markers such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1) ), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138
  • chimeric antigen receptor CAR and type I interferon engineered immune cells can mobilize endogenous immunity to exert a stronger anti-tumor effect Function, more effective and selective killing of tumor cells, such as tumor cells with high expression of PSMA.
  • the term “about” means that the value can vary from the recited value by no more than 1%.
  • the expression “about 100” includes all values between 99 and 101 (eg, 99.1, 99.2, 99.3, 99.4, etc.).
  • the term “comprising” or “including (including)” can be open, semi-closed, and closed. In other words, the term also includes “substantially composed of” or “consisting of”.
  • 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.
  • 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 that is 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 may but not necessarily have, and may 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%.
  • PSMA proteostrate specific membrane antigen, prostate specific membrane antigen
  • PSMA prostate specific membrane antigen
  • CAR-T cells targeting PSMA are extremely specific in tumor immunotherapy.
  • Junghans R et al. (2016) and Zuccolotto G (2014) have demonstrated the effectiveness and safety of CART cells in preclinical studies and clinical trials, but their application is still There are limitations, so further research and exploration are needed.
  • Glypican 3 is highly expressed in hepatocellular carcinoma, gastric cancer and other cancers. It is a meaningful diagnostic, therapeutic and prognostic biomarker for liver cancer, and it has been used in the second generation /Research report on the treatment of hepatocellular carcinoma with CAR-T cells targeted by GPC3 of the third generation.
  • Gangliosides such as GD2 are not only highly expressed in neuroblastoma, but also in a variety of solid tumors, including osteosarcoma, retinoblastoma, some soft tissue sarcomas, brain tumors and other tumors.
  • a clinical trial (NCT02765243) was carried out to evaluate the safety and effectiveness of GD2CART.
  • human epidermal growth factor receptor HER2 occurs in approximately 15-30% of breast cancers and 10-30% of gastric/gastroesophageal cancers, and serves as a prognostic and predictive biomarker. HER2 overexpression is also seen in other cancers, such as ovarian cancer, endometrial cancer, bladder cancer, lung cancer, colon cancer, and head and neck cancer. HER2 is a fully validated target for breast cancer and sarcoma. Targeting HER2 with CAR T cells is very effective in the treatment of brain metastatic breast cancer and sarcoma in animal models, and relevant clinical trials have been carried out (NCT03696030, NCT00902044) .
  • MSLN Mesothelin
  • Carcinoembryonic antigen is widely expressed in cancers such as gastric cancer, lung cancer, pancreatic cancer, breast cancer and colorectal cancer. It has been developed and confirmed that anti-CEA CAR modified T cells play a role in solid tumors (NCT02349724).
  • Epidermal growth factor receptor EGFRvIII is the most common mutant of EGFR, and it is highly expressed in tumors such as malignant glioma, glioblastoma, brain cancer, and glioma. Its expression promotes tumorigenesis and is associated with poor prognosis. It is not expressed in normal tissues and is an attractive target for immunotherapy. In in vitro experiments, CART against EGFRvIII has a good tumor-killing ability, and there have been related clinical experimental studies (NCT01454596).
  • CLDN18.2 The gastric-specific membrane protein Claudin 18.2 (CLDN18.2) is considered to be a potential therapeutic target for gastric cancer and other cancer types.
  • CLDN18.2-specific CAR T cells may be the most promising for gastric cancer and other potential CLDN18.2-positive tumors Treatment strategy (NCT03874897).
  • MUC-1 is a glycosylated transmembrane protein. In normal cells, it is expressed on the apical surface of epithelial cells. Cancer cells express 100 times more MUC1 protein than normal cells. Studies have shown that MUC1-related antibody production and cellular immune responses have a positive impact on the prognosis of cancer patients. There have been clinical trials on the safety and effectiveness of MUC-1CART in the treatment of intrahepatic cholangiocarcinoma (NCT03633773).
  • NKG2D ligand is not only expressed in most human tumor cells, including solid tumors (ovarian cancer, bladder cancer, breast cancer, lung cancer, liver cancer, colon cancer, kidney cancer, prostate cancer, melanoma, Ewing sarcoma, glioma) And neuroblastoma), and various leukemias (AML, CML, CLL), lymphoma and multiple myeloma. It is also expressed on immunosuppressive cells in the tumor microenvironment. Therefore, it provides an attractive target for cancer treatment.
  • CD19 is the most widely used hematological malignant tumor target in CAR-T cell therapy. It is used in acute B-cell lymphocytic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL) and Clinical trials of diffuse large B-cell lymphoma (DLBCL) and other hematological malignancies have achieved good therapeutic effects.
  • B-ALL B-cell lymphocytic leukemia
  • CLL chronic lymphocytic leukemia
  • FL follicular lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • BCMA, CD20, CD22, CD30, IL3RA, CD38, CD138 and other molecules are also research targets for hematological malignancies, especially CD19-negative hematological tumors.
  • BCMA B cell maturation antigen
  • TNFRSF17 TNF receptor superfamily member 17, TNF ligand superfamily member 17
  • BCMA is the most selectively expressed receptor on multiple myeloma cell lines, and its expression gradually increases with the differentiation of B cells, and it also gradually increases in the disease process of multiple myeloma.
  • Immunotherapy targeting BCMA has significant effects in preclinical and clinical studies, especially CAR-T technology, which can specifically recognize tumor antigens in a non-major histocompatibility complex-dependent manner and exert a powerful anti-tumor immune effect .
  • Interferon is a glycoprotein, which is mainly produced by a variety of cells derived from hematopoietic and matrix. According to its structural characteristics, receptors, cell source and biological activity, it can be divided into three types, type I, II and III. Viruses, inhibiting cell proliferation, regulating immunity and anti-tumor effects all have important regulatory effects.
  • type I interferon has the potential to regulate the immune microenvironment.
  • IFN ⁇ can enhance the activation of TH1 cells by inhibiting the proliferation and activity of Treg cells.
  • Type I interferons include seven types: IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ and IFN ⁇ .
  • IFN ⁇ has multiple subtypes, and these subtypes have homology. , But the function is different.
  • Its receptor is a heterodimeric transmembrane IFN ⁇ receptor (IFNAR).
  • IFNAR immunofluorescence-activated protein kinas
  • All type I IFNs can activate receptor-related JAK1 and TYK2 kinases and downstream signal transducers and activators of transcription (STAT ) Transcription factors.
  • STAT signal transducers and activators of transcription
  • IFN stimulating gene factor 3 (ISGF3) complex composed of STAT1, STAT2 and IFN regulatory factor 9 (IRF9) is formed.
  • ISGF3 IFN stimulating gene factor 3
  • ISGF3 complex IFN stimulated response element
  • ISG IFN stimulating gene
  • Type I interferon includes, but is not limited to: IFN ⁇ 1, IFN ⁇ 2 (including IFN ⁇ 2a, IFN ⁇ 2b, IFN ⁇ 2c), IFN ⁇ 4, IFN ⁇ 5, IFN ⁇ 6, IFN ⁇ 7, IFN ⁇ 8, IFN ⁇ 10, IFN ⁇ 13, IFN ⁇ 14, IFN ⁇ 17, IFN ⁇ 21 IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , IFN ⁇ , or a combination thereof, preferably IFN ⁇ 2b.
  • the present invention finds for the first time that in the presence of type I interferons, on the one hand, they can directly act on tumor cells to inhibit and kill tumor cells; on the other hand, they can regulate the innate immune response, thereby promoting antigen presentation and killing cell functions; one On the one hand, they activate the adaptive immune system, thereby promoting the development of high-affinity antigen-specific T and B cell responses and immune memory; on the one hand, they not only inhibit tumor angiogenesis but also promote immune cell migration and infiltration. On the one hand, they can act on Treg and MDSC to regulate the immunosuppressive tumor microenvironment.
  • the amino acid sequence of Type I interferon is shown in any one of SEQ ID NO.: 9, 36-48.
  • the antigen binding domain of the chimeric antigen receptor CAR specifically binds to tumor cell markers (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138).
  • tumor cell markers such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138.
  • 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 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 CD8, and 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 include 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 a specific function.
  • the entire intracellular signaling domain can generally be used, in many cases, the entire chain need not be used.
  • intracellular signaling domain generally refers to any truncated portion of an 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 may be designed to include the CD3 ⁇ signaling domain itself, or may be combined with any other desired cytoplasmic domains (one or more) useful in the content of the CAR of the present invention.
  • 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 the 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 of 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- 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 and proliferation, cytolytic toxicity, and secretion of cytokines, 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 various countries around 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 part.
  • the costimulatory signal transduction region refers to a part of the intracellular domain that includes 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 of the antibody 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 of that of a complete antibody.
  • the single-chain antibody is preferably an amino acid chain sequence encoded by a nucleotide chain.
  • the scFv includes markers that specifically recognize tumors with high expression tumor cells (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1) ), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138) antibodies, preferably single-chain antibodies.
  • the antigen-binding portion of the CAR of the present invention targets tumor cell markers.
  • the antigen binding portion of the CAR of the present invention is a scFV targeting PSMA.
  • the structure of the scFv is as shown in formula A1 or A2:
  • V L1 is the light chain variable region of the anti-tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or peptide bond.
  • amino acid sequence of V L1 is shown at positions 22-128 of SEQ ID NO.:1
  • amino acid sequence of V H1 is shown at positions 144-258 of SEQ ID NO.:1.
  • amino acid sequence of V L1 is shown in SEQ ID NO.:2.
  • amino acid sequence of V H1 is shown in SEQ ID NO.:3.
  • the antigen binding portion of the CAR of the present invention is a scFV targeting GPC3.
  • the structure of the scFv is as shown in formula A1 or A2:
  • V L1 is the light chain variable region of the anti-tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or peptide bond.
  • amino acid sequence of V L1 is shown in SEQ ID NO.: 49, positions 22-134
  • amino acid sequence of V H1 is shown in SEQ ID NO.: 49, positions 150-264.
  • amino acid sequence of V L1 is shown in SEQ ID NO.:51.
  • amino acid sequence of V H1 is shown in SEQ ID NO.:53.
  • the antigen binding portion of the CAR of the present invention is a scFV targeting BCMA.
  • the structure of the scFv is as shown in formula A1 or A2:
  • V L1 is the light chain variable region of the anti-tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or peptide bond.
  • amino acid sequence of V L1 is shown in SEQ ID NO.: 56 at positions 22 to 131
  • amino acid sequence of V H1 is shown in SEQ ID NO.: 56 at positions 146 to 272.
  • amino acid sequence of V L1 is shown in SEQ ID NO.:58.
  • amino acid sequence of V H1 is shown in SEQ ID NO.:60.
  • the scFV comprises a variant form which has ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 95%, ⁇ 98% or ⁇ 99% homology with its wild-type scFV sequence sex.
  • 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 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 is shown in any one of SEQ ID NO.: 1, 10, 49, 54, 56 or 61.
  • nucleotide sequence of the CAR is shown in any one of SEQ ID NO.: 11, 55 or 62.
  • positions 1-21 are signal peptides; positions 22-258 are antigen-binding domains targeting tumor cell markers (such as PSMA-targeted antibody single-chain variable region sequences) ; 259-303 is the hinge region; 304-327 are transmembrane regions (such as the transmembrane region of CD8a); 328-370 are costimulatory components (such as 4-1BB); 371-481 are CD3 ⁇ , Positions 482-500 are connecting peptides (such as self-cleaving proteins), and positions 501-689 are type I interferons (such as IFN ⁇ 2b).
  • positions 1-21 are signal peptides; positions 22-264 are antigen-binding domains targeting tumor cell markers (such as the single-chain variable region sequence of an antibody targeting GPC3); 265-309 are the hinge region; 310-333 are transmembrane regions (such as the transmembrane region of CD8a); 334-375 are costimulatory components (such as 4-1BB); 376-487 are CD3 ⁇ , and 488 Position -506 is for connecting peptide (such as self-cleaving protein), position 507-694 is for type I interferon (such as IFN ⁇ 2b).
  • 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 PSMA), used to treat tumors with high or positive PSMA expression, especially solid tumors.
  • PSMA tumor surface antigens.
  • 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 includes (i) an extracellular domain, which includes an antigen targeting tumor cell surface antigen; (ii) a transmembrane domain; (iii) a costimulatory factor; and (iv) a signal of CD3 ⁇ Conduction domain; and; (v) connecting peptide (such as self-cleaving protein); (vi) type I interferon.
  • CAR-M cells As used herein, the terms “CAR-M cells”, “CAR-M”, and “CAR-M cells of the present invention” all refer to the CAR-M cells of the present invention, and the CAR-M cells of the present invention can target tumor surface antigens.
  • PSMA public tumor antigens
  • PSMA positive tumor antigens
  • Macrophages are the main effectors and regulators of the innate immune system. They have the ability to swallow, secrete pro-inflammatory factors, and present antigens to T cells to activate the immune system.
  • CAR-M itself can directly kill antigen-specific tumor cells in vitro, inhibit tumor growth in vivo, reshape the tumor microenvironment, and has good anti-tumor activity.
  • CAR-M also has the ability to present antigen , Presenting tumor cell antigens and activating endogenous T cells.
  • 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 PSMA) for the treatment of tumors with high or positive PSMA expression, especially solid tumors.
  • PSMA tumor surface antigens
  • 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 (genetically 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 thus 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.
  • the foreign T cell antigen receptor refers to the cloning of the ⁇ chain and ⁇ chain of TCR from tumor-reactive T cells through gene transfer technology, and the use of lentivirus or Retroviruses are vectors, which 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, directly isolated from the cells and tissues containing 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 chronic viruses 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 this vector, which includes, but is 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, Rous 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, for example, a mammalian, bacterial, yeast, or insect cell, by any method in the art.
  • 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, such as 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 and a transposon.
  • the transposon system Compared with the traditional retroviral system with limited loading capacity, complicated preparation process, and random insertion risk, the transposon system has a relatively simple preparation process, integrates foreign genes into the genome through transposase, and has the advantages of high loading capacity.
  • the earliest used mammalian transposon system is the "Sleeping-Beauty” transposon, but the “Sleeping-Beauty” transposon has defects such as excessive inhibitory effect and small carrying fragments (about 5kb), making it difficult to The application of genetic modification is restricted.
  • the piggyBac (PB) transposon derived from lepidopteran insects is currently the most active transposon in mammals.
  • the host range is extremely wide, from single-celled organisms to mammals, capable of carrying large foreign DNA fragments. When the transposable fragment is within 14kb, the transposition efficiency will not decrease significantly.
  • PB transposon mainly adopts the "cut-paste" mechanism for transposition.
  • PB transposase has high plasticity. By fusing with other functional proteins or changing the functional region of the transposase, it can not only change the activity and mode of action of the transposase, but also improve the targeting of foreign gene transposition. In recent years, the integration efficiency of PB in mammalian cells has been further improved through codon optimization, site-directed mutations of amino acids at specific sites, and the introduction of corresponding nuclear localization tags, making it widely used in cell therapy and gene therapy.
  • the lentiviral system has an upper limit on the sequence length when expressing long-segment sequences and cannot complete the packaging of large-segment expression vectors.
  • the transposon system can insert long fragments within 14k at most, and the risk of random insertion is less than that of lentivirus, which has a wider range of applications. Scope and safety of use.
  • the present invention provides an engineered immune cell according to the first 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 4 -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 lentiviral vectors (LV) encoding the expression cassettes of the present invention.
  • the transduced T cells can target tumor cell markers (such as PSMA, etc.) proteins to coordinately activate T cells and cause cellular immune responses, thereby selectively killing tumor cells, such as tumor cells with high expression of PSMA.
  • tumor cell markers such as PSMA, etc.
  • 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 graft-versus-host disease is extremely low, and antigens are 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 PSMA, etc.
  • cause a specific immune response against cells expressing tumor cell markers such as PSMA, etc.
  • lentiviruses including antigen-binding domains, hinges and transmembrane regions that target tumor cell surface antigens, and 4-1BB and CD3 ⁇ signaling domains, P2A, type I interferons (such as IFN ⁇ 2b) Vector, but the present invention should be interpreted as including any number of changes in each part of the construct.
  • Treatable cancers include tumors that have not been vascularized or have not been substantially vascularized, as well as vascularized tumors.
  • Cancers may include non-solid tumors (such as hematological tumors such as leukemia and lymphoma) or 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 malignancies 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 prostate cancer, liver cancer, fibrosarcoma, myxosarcoma, liposarcoma, mesothelioma, lymphoid malignancies, pancreatic cancer, and ovarian cancer.
  • the CAR-modified T cell 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 can 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 cell 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 particular 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.
  • cells activated and expanded using the methods described herein or other methods known in the art to expand T cells to therapeutic levels are combined with any number of relevant treatment modalities (e.g., previous , At the same time or after) administration to the patient, the treatment modality includes but 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, at the same time, or after). patient.
  • chemotherapeutic agents such as fludarabine, external beam radiotherapy (XRT), cyclophosphamide (for example, before, at the same time, 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.
  • fusion protein As used herein, the terms “fusion protein”, “fusion protein of the present invention”, and “polypeptide of the present invention” have the same meaning, and all have the structure described in the eighth aspect of the present invention.
  • amino acid sequence of the fusion protein is shown in SEQ ID NO.: 10 or 54 or 61.
  • the term "fusion protein” also includes variant forms of the sequence of SEQ ID NO.: 10 or 54 or 61 having the above-mentioned activity. These variant forms include (but are not limited to): 1-3 (usually 1-2, more preferably 1) amino acid deletion, insertion and/or substitution, and addition or addition at the C-terminus and/or N-terminus One or several (usually within 3, preferably within 2, more preferably within 1) amino acid is missing.
  • amino acids with similar or similar properties are substituted, the function of the protein is usually not changed.
  • adding or deleting one or several amino acids at the C-terminus and/or N-terminus usually does not change the structure and function of the protein.
  • the term also includes the polypeptides of the present invention in monomeric and multimeric forms. The term also includes linear and non-linear polypeptides (such as cyclic peptides).
  • the present invention also includes active fragments, derivatives and analogs of the above-mentioned fusion protein.
  • fragment refers to a polypeptide that substantially retains the function or activity of the fusion protein of the present invention.
  • polypeptide fragments, derivatives or analogues of the present invention can be (i) one or several conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted, or (ii) in one or more A polypeptide with substitution groups in three amino acid residues, or (iii) a polypeptide formed by fusion of a polypeptide with another compound (such as a compound that prolongs the half-life of the polypeptide, such as polyethylene glycol), or (iv) an additional amino acid sequence fusion A polypeptide formed from this polypeptide sequence (a fusion protein formed by fusion with a leader sequence, a secretory sequence, or a tag sequence such as 6His). According to the teachings herein, these fragments, derivatives and analogs belong to the scope well known to those skilled in the art.
  • a preferred type of active derivative means that compared with the amino acid sequence of the present invention, there are at most 3, preferably at most 2, and more preferably at most 1 amino acid replaced by an amino acid with similar or similar properties to form a polypeptide. These conservative variant polypeptides are best produced according to Table 1 through amino acid substitutions.
  • substitutions Ala(A) Val; Leu; Ile Val Arg(R) Lys; Gln; Asn Lys Asn(N) Gln; His; Lys; Arg Gln Asp(D) Glu Glu Cys(C) Ser Ser Gln(Q) Asn Asn Glu(E) Asp Asp Gly(G) Pro; Ala Ala His(H) Asn; Gln; Lys; Arg Arg Ile(I) Leu; Val; Met; Ala; Phe Leu
  • the present invention also provides analogs of the fusion protein of the present invention.
  • the difference between these analogs and the polypeptide shown in SEQ ID NO.: 10 or 54 or 61 may be the difference in the amino acid sequence, the difference in the modification form that does not affect the sequence, or both.
  • Analogs also include analogs having residues different from natural L-amino acids (such as D-amino acids), and analogs having non-naturally occurring or synthetic amino acids (such as ⁇ , ⁇ -amino acids). It should be understood that the polypeptide of the present invention is not limited to the representative polypeptides exemplified above.
  • Modified forms include: chemically derived forms of polypeptides in vivo or in vitro, such as acetylation or carboxylation. Modifications also include glycosylation, such as those polypeptides produced by glycosylation modifications during the synthesis and processing of the polypeptide or during further processing steps. This modification can be accomplished by exposing the polypeptide to an enzyme that performs glycosylation (such as a mammalian glycosylase or deglycosylase). Modified forms also include sequences with phosphorylated amino acid residues (such as phosphotyrosine, phosphoserine, and phosphothreonine). It also includes polypeptides that have been modified to improve their resistance to proteolysis or to optimize their solubility.
  • glycosylation such as those polypeptides produced by glycosylation modifications during the synthesis and processing of the polypeptide or during further processing steps. This modification can be accomplished by exposing the polypeptide to an enzyme that performs glycosylation (such as a mammalian
  • the amino acid sequence of the fusion protein is as shown in SEQ ID NO.: 10 or 54 or 61.
  • the invention also relates to polynucleotides encoding the fusion protein according to the invention.
  • the polynucleotide of the present invention may be in the form of DNA or RNA.
  • DNA can be a coding strand or a non-coding strand.
  • the sequence of the coding region encoding the mature polypeptide may be the same as the sequence encoding the polypeptide shown in SEQ ID NO.: 10 or 54 or 61 or a degenerate variant.
  • degenerate variant in the present invention refers to a nucleic acid sequence that encodes a polypeptide shown in SEQ ID NO.: 10 or 54 or 61, but differs in the sequence of the corresponding coding region.
  • the full-length nucleotide sequence of the present invention or its fragments can usually be obtained by PCR amplification method, recombination method or artificial synthesis method.
  • the DNA sequence encoding the polypeptide (or fragment or derivative thereof) of the present invention can be obtained completely through chemical synthesis.
  • the DNA sequence can then be introduced into various existing DNA molecules (or such as vectors) and cells known in the art.
  • the present invention also relates to a vector containing the polynucleotide of the present invention, and a host cell produced by genetic engineering using the vector or polypeptide coding sequence of the present invention.
  • the aforementioned polynucleotides, vectors or host cells may be isolated.
  • isolated refers to the separation of a substance from its original environment (if it is a natural substance, the original environment is the natural environment).
  • the polynucleotides and polypeptides in the natural state in living cells are not separated and purified, but the same polynucleotides or polypeptides are separated and purified if they are separated from other substances that coexist in the natural state.
  • the polynucleotide of the present invention may be in the form of DNA or RNA.
  • the form of DNA includes cDNA, genomic DNA or synthetic DNA.
  • DNA can be single-stranded or double-stranded.
  • DNA can be a coding strand or a non-coding strand.
  • the present invention also relates to variants of the aforementioned polynucleotides, which encode protein fragments, analogs and derivatives having the same amino acid sequence as the present invention.
  • the variants of this polynucleotide can be naturally occurring allelic variants or non-naturally occurring variants. These nucleotide variants include substitution variants, deletion variants and insertion variants.
  • an allelic variant is an alternative form of a polynucleotide. It may be a substitution, deletion or insertion of one or more nucleotides, but it will not substantially change its encoding of the fusion protein of the present invention. Function.
  • the full-length nucleotide sequence or fragments thereof encoding the fusion protein of the present invention can usually be obtained by PCR amplification method, recombination method or artificial synthesis method.
  • primers can be designed according to the published relevant nucleotide sequence, especially the open reading frame sequence, and a commercially available cDNA library or a cDNA library prepared by a conventional method known to those skilled in the art can be used as Template, amplified and get related sequence.
  • a commercially available cDNA library or a cDNA library prepared by a conventional method known to those skilled in the art can be used as Template, amplified and get related sequence.
  • the sequence is long, it is often necessary to perform two or more PCR amplifications, and then splice the amplified fragments together in the correct order.
  • the polynucleotide sequence encoding the fusion protein is as shown in SEQ ID NO.: 11 or 55 or 62.
  • the recombination method can be used to obtain the relevant sequence in large quantities. This is usually done by cloning it into a vector, then transferring it into a cell, and then isolating the relevant sequence from the proliferated host cell by conventional methods.
  • artificial synthesis methods can also be used to synthesize related sequences, especially when the fragment length is short. Usually, by first synthesizing multiple small fragments, and then ligating to obtain fragments with very long sequences.
  • the method of using PCR technology to amplify DNA/RNA is preferably used to obtain the gene of the present invention.
  • the primers used for PCR can be appropriately selected according to the sequence information of the present invention disclosed herein, and can be synthesized by conventional methods.
  • the amplified DNA/RNA fragments can be separated and purified by conventional methods such as gel electrophoresis.
  • the present invention also relates to a vector containing the polynucleotide of the present invention, a host cell produced by genetic engineering using the vector or protein coding sequence of the present invention, and a method for expressing the fusion protein of the present invention on the NK cell by recombinant technology.
  • the polynucleotide sequence of the present invention can be used to obtain NK cells expressing the fusion protein of the present invention. Generally, it includes the steps of: transducing the first expression cassette and/or the second expression cassette of the present invention into NK cells, so as to obtain the NK cells.
  • an expression vector containing the DNA sequence encoding the fusion protein of the present invention and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA technology, DNA synthesis technology, and in vivo recombination technology.
  • the DNA sequence can be effectively linked to an appropriate promoter in the expression vector to guide mRNA synthesis.
  • the expression vector also includes a ribosome binding site for translation initiation and a transcription terminator.
  • the expression vector preferably contains one or more selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green Fluorescent protein (GFP), or tetracycline or ampicillin resistance for E. coli.
  • selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase for eukaryotic cell culture, neomycin resistance, and green Fluorescent protein (GFP), or tetracycline or ampicillin resistance for E. coli.
  • a vector containing the above-mentioned appropriate DNA sequence and an appropriate promoter or control sequence can be used to transform an appropriate host cell so that it can express the protein.
  • the host cell can be a prokaryotic cell, such as a bacterial cell; or a lower eukaryotic cell, such as a yeast cell; or a higher eukaryotic cell, such as a mammalian cell.
  • a prokaryotic cell such as a bacterial cell
  • a lower eukaryotic cell such as a yeast cell
  • a higher eukaryotic cell such as a mammalian cell.
  • Representative examples are: Escherichia coli, Bacillus subtilis, Streptomyces bacterial cells; fungal cells such as Pichia pastoris, Saccharomyces cerevisiae cells; plant cells; Drosophila S2 or Sf9 insect cells; CHO, NS0, COS7, or 293 Cells of animal cells and so on.
  • NK cells are selected as host cells.
  • Transformation of host cells with recombinant DNA can be carried out by conventional techniques well known to those skilled in the art.
  • the host is a prokaryotic organism such as Escherichia coli
  • competent cells that can absorb DNA can be harvested after the exponential growth phase and treated with the CaCl 2 method.
  • the steps used are well known in the art.
  • Another method is to use MgCl 2 .
  • the transformation can also be carried out by electroporation.
  • the host is a eukaryote, the following DNA transfection methods can be used: calcium phosphate co-precipitation method, conventional mechanical methods such as microinjection, electroporation, liposome packaging, etc.
  • the obtained transformants can be cultured by conventional methods to express the protein encoded by the gene of the present invention.
  • the medium used in the culture can be selected from various conventional mediums.
  • the culture is carried out under conditions suitable for the growth of the host cell. After the host cells have grown to an appropriate cell density, the selected promoter is induced by an appropriate method (such as temperature conversion or chemical induction), and the cells are cultured for a period of time.
  • the protein in the above method can be expressed in the cell or on the cell membrane, or secreted out of the cell. If necessary, the physical, chemical, and other properties can be used to separate and purify the protein through various separation methods. These methods are well known to those skilled in the art. Examples of these methods include, but are not limited to: conventional renaturation treatment, treatment with protein precipitation agent (salting out method), centrifugation, osmotic sterilization, ultra-treatment, ultra-centrifugation, molecular sieve chromatography (gel filtration), adsorption layer Analysis, ion exchange chromatography, high performance liquid chromatography (HPLC) and various other liquid chromatography techniques and combinations of these methods.
  • the present invention finds for the first time that it contains targeted tumor cell markers (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138) chimeric antigen receptor CAR and type I interferon engineered immune cells can mobilize endogenous immunity to exert a stronger anti-tumor effect, and more effectively and selectively kill tumors Cells, such as tumor cells with high expression of PSMA, tumor cells with high expression of GPC3, and tumor cells with high expression of BCMA.
  • targeted tumor cell markers such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin 18.2, Mucin 1 (MUC1), NKG2D ligand, CD19, CD20, BCMA, CD22
  • type I interferon can directly act on tumor cells, inhibiting and killing tumor cells
  • type I interferon can regulate the innate immune response, thereby promoting antigen presentation and the killing function of CAR engineered immune cells;
  • the present invention finds for the first time that the adaptive immune system is activated, thereby promoting the development of high-affinity antigen-specific T and B cell responses and immune memory, and improving the persistence of CAR engineered immune cells;
  • the present invention finds for the first time that type I interferon not only inhibits tumor angiogenesis but also promotes the migration and infiltration of CAR engineered immune cells.
  • the present invention finds for the first time that type I interferon can act on Treg and MDSC, thereby releasing the tumor microenvironment that inhibits CAR engineered immune cells.
  • the present invention utilizes the transposon system to mediate the efficient integration of the chimeric antigen receptor and type I interferon into the host cell genome, and can obtain positive rates of stable expression of IFN ⁇ 2b-CART at different targets, and the IFN ⁇ 2b-CART can be known by ELISA verification.
  • CART can express up to about 30 times IFN ⁇ 2b than traditional CART.
  • PSMA, GPC3, and BCMA were used as targets to construct plasmid vectors containing two expression cassettes expressing chimeric antigen receptor and IFN ⁇ 2b.
  • Figure 1 for the structure and positional relationship of each element on the expression cassette.
  • the first expression cassette (PSMA-CAR) includes: CD8 ⁇ signal peptide , PSMA single-chain antibody heavy chain variable region, Linker1, PSMA single-chain antibody light chain, CD8 hinge region, CD8 ⁇ transmembrane domain, intracellular costimulatory element of 4-1BB and intracellular domain of CD3 ⁇ ( Figure 1, A) Connect the above sequences in sequence, and introduce the Kozak sequence and the corresponding restriction site at the forefront.
  • the first expression cassette was transferred to the lentiviral shuttle plasmid (obtained from Shanghai Bangyao Biotechnology Co., Ltd.), and the chimeric antigen receptor expression vector pELPS-PSMA-BBz transfer vector (as Control plasmid).
  • pELPS-PSMA-BBz transfer vector plasmid as the initial plasmid, a second expression cassette expressing IFN ⁇ 2b was added.
  • the first expression cassette and the second expression cassette were connected by P2A, and the second expression cassette (IFN ⁇ 2b) was IFN ⁇ 2b ( Figure 1, B).
  • the resulting plasmid was named pELPS-PSMA-BBz-2A-IFN ⁇ 2b transfer vector plasmid.
  • the first expression cassette expressing chimeric antigen receptor targeting hepatocyte-specific membrane antigen is synthesized by GenScript, and the first expression cassette (GPC3-CAR) includes: CD8 ⁇ signal Peptides, GPC3 single-chain antibody heavy chain variable region, Linker1, GPC3 single-chain antibody light chain, CD8 hinge region, CD8 ⁇ transmembrane domain, intracellular costimulatory element of 4-1BB and intracellular domain of CD3 ⁇ ( Figure 1 , C), connect the above sequences in sequence, and introduce the Kozak sequence and the corresponding restriction site at the forefront.
  • the first expression cassette was transferred to the lentiviral shuttle plasmid (obtained from Shanghai Bangyao Biotechnology Co., Ltd.), and the chimeric antigen receptor expression vector pELPS-PSMA-BBz transfer vector (as Control plasmid).
  • pELPS-GPC3-BBz transfer vector plasmid as the initial plasmid, a second expression cassette expressing IFN ⁇ 2b was added, the first expression cassette and the second expression cassette were connected by P2A, and the second expression cassette (IFN ⁇ 2b) was IFN ⁇ 2b ( Figure 1, D).
  • the resulting plasmid was named pELPS-GPC3-BBz-2A-IFN ⁇ 2b transfer vector plasmid.
  • the first expression cassette that expresses the chimeric antigen receptor targeting the specific membrane antigen of multiple myeloma is synthesized by GenScript.
  • the first expression cassette (BCMA-CAR) includes: CD8 ⁇ signal peptide, BCMA single-chain antibody heavy chain variable region, Linker1, BCMA single-chain antibody light chain, CD8 hinge region, CD8 ⁇ transmembrane domain, intracellular costimulatory element of 4-1BB and intracellular domain of CD3 ⁇ ( Figure 1, E), connect the above sequences in sequence, and introduce the Kozak sequence and the corresponding restriction site at the forefront.
  • the first expression cassette was transferred to the lentiviral shuttle plasmid (obtained from Shanghai Bangyao Biotechnology Co., Ltd.), and the chimeric antigen receptor expression vector pELPS-BCMA-BBz transfer vector (as Control plasmid).
  • pELPS-BCMA-BBz transfer vector plasmid as the initial plasmid, a second expression cassette expressing IFN ⁇ 2b was added, the first expression cassette and the second expression cassette were connected by P2A, and the second expression cassette (IFN ⁇ 2b) was IFN ⁇ 2b ( Figure 1, F).
  • the resulting plasmid was named pELPS-BCMA-BBz-2A-IFN ⁇ 2b transfer vector plasmid.
  • the sequence of each element of the above-mentioned first expression cassette is as follows:
  • CD8 ⁇ Leader The base sequence of CD8 ⁇ signal peptide (CD8 ⁇ Leader) is shown in SEQ ID NO.: 12:
  • CD8 ⁇ signal peptide CD8 ⁇ Leader
  • SEQ ID NO.: 4 The amino acid sequence of CD8 ⁇ signal peptide (CD8 ⁇ Leader) is shown in SEQ ID NO.: 4: MALPVTALLLPLALLLHAARP;
  • amino acid sequence of PSMA-CAR without type I interferon is shown in SEQ ID NO.:1:
  • PSMA-ScFv VL The base sequence of the PSMA single-chain antibody light chain variable region (PSMA-ScFv VL) is shown in SEQ ID NO.: 13:
  • PSMA-ScFv VL The amino acid sequence of the PSMA single-chain antibody light chain variable region (PSMA-ScFv VL) is shown in SEQ ID NO.: 2:
  • PSMA-ScFv VH The base sequence of the PSMA single-chain antibody heavy chain variable region (PSMA-ScFv VH) is shown in SEQ ID NO.: 16:
  • PSMA-ScFv VH The amino acid sequence of the PSMA single-chain antibody heavy chain variable region (PSMA-ScFv VH) is shown in SEQ ID NO.: 3:
  • CD8 hinge region (CD8hinge) is shown in SEQ ID NO.: 17:
  • CD8hinge The amino acid sequence of the CD8 hinge region (CD8hinge) is shown in SEQ ID NO.: 5:
  • CD8 ⁇ transmembrane domain (CD8a-TM) is shown in SEQ ID NO.: 18:
  • CD8 ⁇ transmembrane domain CD8a-TM
  • the base sequence of the intracellular costimulatory element of 4-1BB is shown in SEQ ID NO.: 19:
  • amino acid sequence of the intracellular costimulatory element of 4-1BB is shown in SEQ ID NO.: 7:
  • the base sequence of the intracellular domain of CD3 ⁇ is shown in SEQ ID NO.: 20:
  • amino acid sequence of the intracellular domain of CD3 ⁇ is shown in SEQ ID NO.: 8:
  • the base sequence of P2A is shown in SEQ ID NO.: 21:
  • the base sequence of type I interferon is as follows:
  • amino acid sequence of type I interferon is shown below:
  • PSMA-CAR containing type I interferon IFN ⁇ 2b
  • SEQ ID NO. 10 The amino acid sequence of PSMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 10:
  • the nucleotide sequence of the PSMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 11:
  • the first expression cassette PSMA-ScFv VL-Linker-PSMA-ScFv VL ( Figure 1-A) expressing the chimeric antigen receptor targeting prostate-specific membrane antigen can be replaced with the following expressions targeting GPC3 and BCMA Corresponding components, other components can remain unchanged.
  • PB-GPC3-CAR plasmid ( Figure 1C) and PB-IFN ⁇ 2b-GPC3-CAR ( Figure 1D) plasmids were synthesized with reference to the method in Example 3 below.
  • amino acid sequence of GPC3-CAR without type I interferon is shown in SEQ ID NO.: 49:
  • GPC3-ScFv VL The base sequence of the GPC3 single-chain antibody light chain variable region (GPC3-ScFv VL) is shown in SEQ ID NO.: 50:
  • GPC3-ScFv VL The amino acid sequence of the GPC3 single-chain antibody light chain variable region (GPC3-ScFv VL) is shown in SEQ ID NO.: 51:
  • the Linker of GPC3-ScFv VL and GPC3-ScFv VH is the base sequence of SEQ ID NO.: 14 above.
  • the amino acid sequence of the Linker of GPC3-ScFv VL and GPC3-ScFv VH is shown in SEQ ID NO.: 15 above.
  • GPC3-ScFv VH The base sequence of the GPC3 single-chain antibody heavy chain variable region (GPC3-ScFv VH) is shown in SEQ ID NO.: 52:
  • GPC3-ScFv VH The amino acid sequence of the GPC3 single-chain antibody heavy chain variable region (GPC3-ScFv VH) is shown in SEQ ID NO.: 53:
  • GPC3-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 54:
  • GPC3-CAR containing type I interferon IFN ⁇ 2b
  • BCMA-ScFv VL BCMA single-chain antibody light chain variable region
  • BCMA-ScFv VL The amino acid sequence of the BCMA single-chain antibody light chain variable region (BCMA-ScFv VL) is shown in SEQ ID NO.: 58:
  • the Linker of BCMA-ScFv VL and BCMA-ScFv VH is the base sequence of SEQ ID NO.: 14 above.
  • the amino acid sequence of the Linker of BCMA-ScFv VLBCMA-ScFv VH is shown in SEQ ID NO.: 15 above.
  • BCMA-ScFv VH The base sequence of the BCMA single-chain antibody heavy chain variable region (BCMA-ScFv VH) is shown in SEQ ID NO.: 59:
  • BCMA-ScFv VH The amino acid sequence of the BCMA single-chain antibody heavy chain variable region (BCMA-ScFv VH) is shown in SEQ ID NO.: 60:
  • BCMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 61:
  • the nucleotide sequence of BCMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 62:
  • the method is as follows: Use Escherichia coli to amplify the pELPS-PSMA-BBz and pELPS-PSMA-BBz-2A-IFN ⁇ 2b plasmids and the lentivirus packaging auxiliary plasmids pMD2.G and psPAX2. After extracting the plasmids, perform agarose gel electrophoresis and sequencing for identification The correctness of the plasmid. The 293T with the highest generation number in good condition was selected as the lentivirus packaging cell, and the above three plasmids were transfected into the 293T cell with the transfection reagent PEI. The transfection was completed in a 10cm culture dish with a total system of 10mL.
  • DMEM fresh medium
  • the culture supernatant was harvested at 48h and 72h respectively, and the virus expressing chimeric antigen receptor and IFN ⁇ 2b was obtained after ultrafiltration and ultraisolation concentration.
  • the resulting virus was named IFN ⁇ 2b-PSMA-CAR virus.
  • Use the pELPS-PSMA-BBz transfer vector plasmid (compared to the pELPS-PSMA-BBz-2A-IFN ⁇ 2b plasmid, the PSMA-CAR plasmid lacks the second expression cassette expressing IFN ⁇ 2b) as a control, and refer to the above methods for transfection and treatment. Named PSMA-CAR virus.
  • titer (TU/mL) (2*10 ⁇ 5*CAR positive rate)/virus volume .
  • Titer detection follows the above-mentioned titer detection method. After the cells are attached to the plate, two volume gradients of 2 ⁇ L and 5 ⁇ L are set for the control viruses PSMA-CAR and IFN ⁇ 2b-CAR viruses, respectively. To avoid false positives caused by non-specific staining, it is necessary to set CTRL carries out the CAR-positive gate, and the CAR-positive cells fall into the APC-positive gate, and the ratio value shown is the CAR-positive rate.
  • 2 ⁇ L of PSMA-CAR concentrated virus infection can reach 60.4% positive rate of 200,000 293T, 5 ⁇ L corresponds to 81.3%; 2 ⁇ L of IFN ⁇ 2b-CAR virus infection can reach 27.4% of 200,000 293T
  • the positive rate, 5 ⁇ L corresponds to a positive rate of 43.9%.
  • a volume of 2 ⁇ L is used to calculate the titer.
  • the titer of the control virus PSMA-CAR can reach 6.04 ⁇ 10 ⁇ 7TU/ mL, and the IFN ⁇ 2b-PSMA-CAR virus titer is 1.8 ⁇ 10 ⁇ 7TU/mL.
  • lymphatic separation fluid to separate PBMC from human blood, and then use CD4 and CD8 magnetic bead sorting to separate T cells.
  • the two CART cells were named PSMA-CART cells and IFN ⁇ 2b-PSMA-CART cells, respectively.
  • the CAR expression levels of the above two CARTs were detected according to the similar method of titer detection 48h after infection. Among them, the positive rate of IFN ⁇ 2b-PSMA-CART was 49.3%, and the positive rate of PSMA-CART was 89.5%.
  • the lentiviral vector was replaced with a transposon system, and electroporation technology was used for delivery.
  • the IFN ⁇ 2b-CART with stable positive rate was successfully prepared, and the expression detection of IFN ⁇ 2b was completed:
  • the above-mentioned first expression cassette was synthesized by GenScript and cloned into the vector PB513B-1 (SBI) vector
  • the PiggyBac transposon vector containing PSMA CAR was constructed and named PB-PSMA-CAR vector.
  • the first expression cassette and the second expression cassette (the first expression cassette and the second expression cassette) in Example 1 were synthesized by GenScript Connected by P2A) and cloned into PB513B-1 (SBI) vector to synthesize PB-IFN ⁇ 2b-PSMA-CAR vector.
  • the PB-BCMA-CAR vector and the PB-IFN ⁇ 2b-BCMA-CAR vector, the PB-GPC3-CAR vector and the PB-IFN ⁇ 2b-GPC3-CAR vector were synthesized using the above-mentioned transposon system and method respectively, and the same method was used to prepare PB-BCMA -CART, PB-IFN ⁇ 2b-BCMA-CART, PB-GPC3-CART, PB-IFN ⁇ 2b-GPC3-CART, the positive rate of CAR was detected by the method of Example 2.
  • IFN ⁇ 2b-CAR For IFN ⁇ 2b-CAR, it is not only necessary to detect the expression of its CAR, but also to verify whether it has the ability to express IFN ⁇ 2b, so collect the above two CART cells under the same culture system conditions (the positive rate is adjusted to be the same, the density is 1M/mL) for 48h The supernatant of ELISA was used to verify the expression of IFN ⁇ 2b.
  • PB-IFN ⁇ 2b-PSMA-CART can express higher IFN ⁇ 2b than traditional PB-PSMA-CART, and the average is as high as about 30 times.
  • IFN ⁇ 2b-CART Both the IFN ⁇ 2b-CART and the traditional CART in the following examples (ie, Examples 4-10) are prepared using the transposon system of this example.
  • CART (PB-PSMA-CART, PB-GPC3-CART, PB-BCMA-CART) expressing only the first expression cassette mentioned above is collectively referred to as conventional CART
  • CART co-expressing the second expression cassette (PB-IFN ⁇ 2b -PSMA-CART, PB-IFN ⁇ 2b-GPC3-CART, PB-IFN ⁇ 2b-BCMA-CART) are collectively referred to as IFN ⁇ 2b-CART.
  • the transposon system in the present invention adopts the PiggyBac TM Transposon Vector System from SBI Company
  • the transposon PB513B-1 is purchased from SBI Company
  • the transposase PB220PA-1 is purchased from System Bioscience Company.
  • the base sequence of PB transposase (PB220PA-1) is shown in SEQ ID NO.: 63:
  • PB220PA-1 The amino acid sequence of PB transposase (PB220PA-1) is shown in SEQ ID NO.: 64:
  • IFN ⁇ 2b enhances the killing effect of CART on tumor cells
  • PB-IFN ⁇ 2b-PSMA-CART and PB-PSMA-CART which have the same adjustment of CAR positive rate, are used as effector cells, and PC3-PSMA (human prostate cancer cell line, using lentivirus to stably express PSMA and luciferase) as target cells, first Add the same amount of target cells (20,000) to the low-adsorption well plate, and add the corresponding number of CART effector cells according to the effective target ratio (effector cell: target cell) 1:1, 0.5:1, 0.25:1, and do different things at the same time
  • the gradient has only the wells of target cells (0.5, 1, 1.5, 2, 2.5, 3, 50,000) as the standard curve.
  • IFN ⁇ 2b did not affect the proliferation of CART
  • IFN ⁇ 2b In order to verify the ability of IFN ⁇ 2b to promote the proliferation of T cells, 200,000 IFN ⁇ 2b-CART and PSMA-CART with the same positive rate were cultured in the same culture system, and the absolute count was followed by a counter to track the two different CARTs within two weeks. Proliferation.
  • IFN ⁇ 2b-CART has a high proportion of Tn (memory T cell) phenotype
  • CART cells 72 hours after electroporation were used for staining with CD4, CD8, CD45RA, CCR7 flow cytometry antibodies, and flow cytometry software was used to analyze the expression of memory marker genes CD45RA and CCR7 in CD4 and CD8 T cells, respectively.
  • the results are shown in Figure 7.
  • CD4 T cells CCR7 + and CD45RA + double positive cells (Tn: The ratio of T cells) in PB-IFN ⁇ 2b-PSMA-CART is 29.5%, and the ratio in PB-PSMA-CART cells is 13.0%; in CD8 T cells, The proportion of T cells in PB-IFN ⁇ 2b-PSMA-CART is 37.9%, and the proportion in PB-PSMA-CART cells is 13.5%.
  • IFN ⁇ 2b-CART has more memory T cell subtypes than PSMA-CART.
  • IFN ⁇ 2b-CART activates PBMC cells and innate immunity
  • the supernatant from IFN ⁇ 2b-CART can effectively activate human PBMC to secrete chemokines (CXCL10, CXCL11, CCL2, CXCL9) that are beneficial to T cells and reduce IL10, CSF2, IL1 ⁇ cytokines, and at the same time enhance Cytokines (IL15, IL18) that promote the proliferation of T cells and other immune cells.
  • chemokines CXCL10, CXCL11, CCL2, CXCL9
  • IL15, IL18 enhance Cytokines
  • the IFN ⁇ 2b-CART supernatant stimulated the expression of NK cells and mononuclear cells (mono) in PBMC to up-regulate TRAI1, indicating that IFN ⁇ 2b-CART can more strongly activate innate immune cells to exert tumor-killing functions (Figure 8B).
  • type I interferon can enhance NK up-regulation of TRAIL by regulating innate immune cells, and mediating the killing effect of NK on tumor cells by binding to TRAIL receptors on the surface of tumor cells
  • the supernatant collected from the above-mentioned traditional CART and IFN ⁇ 2b-CART cells is combined with After homologous PBMC were incubated in a medium containing GM-CSF and IL 2 cytokines for 24 hours, the co-incubated PBMC and huh7 cells were co-incubated at a ratio of 1:1.
  • the four groups of experiments were set up as follows: A: huh7: Only huh7 cells; B: huh7+PBMC: huh7 and PBMC are cultured at a ratio of 1:1; C: huh7+PBMC (conventional CART supernatant stimulation 24h): huh7 and PBMC are cultured at a ratio of 1:1:1; D: huh7+ PBMC (IFN ⁇ 2b-CART supernatant stimulation for 24h): huh7 and PBMC were cultured at a ratio of 1:1; 24h later, the death of huh7 was observed under a microscope.
  • IFN ⁇ 2b-CART supernatant activates tumor cells to up-regulate the expression of chemokines and promote T cell migration.
  • Results Figure 9 shows that IFN ⁇ 2b-CART supernatant (DU145+IFN ⁇ 2b-PSMA-CART) can activate DU145 cells to up-regulate the expression of CCL8 (left). Therefore, a migration experiment was designed for verification: the lower chamber plating (24-well plate) 20 Ten thousand DU145 cells were stimulated with PB-IFN ⁇ 2b-PSMA-CART and PB-PSMA-CART supernatant for 24 hours, and 400,000 CFSE-labeled HT (human T cells) were added to the upper chamber. After 4 hours of migration, use counting magnetic beads for absolute Count the cells that migrated to the lower chamber. The results are shown in Figure 9 (right), IFN ⁇ 2b-CART can significantly promote the migration of HT to the lower ventricle.
  • IFN ⁇ 2b-CART inhibits tumor angiogenesis
  • IFN ⁇ 2b-CART mobilizes endogenous immunity to exert a stronger anti-tumor effect
  • IFN ⁇ 2b-CART promotes tumor regression in vivo
  • the PC3-PSMA cell line (labeled with Luciferase) was injected subcutaneously into the mouse, and the volume of the tumor was tracked using vernier calipers and mouse in vivo imaging technology. When it reached a certain size (200mm 3 ), grouping was set for treatment.
  • amino acid sequence of PSMA-CAR without type I interferon is shown in SEQ ID NO.:1:
  • PSMA-ScFv VL The amino acid sequence of the PSMA single-chain antibody light chain variable region (PSMA-ScFv VL) is shown in SEQ ID NO.: 2:
  • PSMA-ScFv VH The amino acid sequence of the PSMA single-chain antibody heavy chain variable region (PSMA-ScFv VH) is shown in SEQ ID NO.: 3:
  • CD8 ⁇ signal peptide CD8 ⁇ Leader
  • CD8hinge The amino acid sequence of the CD8 hinge region (CD8hinge) is shown in SEQ ID NO.: 5:
  • CD8 ⁇ transmembrane domain CD8a-TM
  • amino acid sequence of the intracellular costimulatory element of 4-1BB is shown in SEQ ID NO.: 7:
  • amino acid sequence of the intracellular domain of CD3 ⁇ is shown in SEQ ID NO.: 8:
  • PSMA-CAR containing type I interferon IFN ⁇ 2b
  • SEQ ID NO. 10 The amino acid sequence of PSMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 10:
  • the nucleotide sequence of the PSMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 11:
  • CD8 ⁇ Leader The base sequence of CD8 ⁇ signal peptide (CD8 ⁇ Leader) is shown in SEQ ID NO.: 12:
  • PSMA-ScFv VL The base sequence of the PSMA single-chain antibody light chain variable region (PSMA-ScFv VL) is shown in SEQ ID NO.: 13:
  • PSMA-ScFv VH The base sequence of the PSMA single-chain antibody heavy chain variable region (PSMA-ScFv VH) is shown in SEQ ID NO.: 16:
  • CD8 hinge region (CD8hinge) is shown in SEQ ID NO.: 17:
  • CD8 ⁇ transmembrane domain (CD8a-TM) is shown in SEQ ID NO.: 18:
  • the base sequence of the intracellular costimulatory element of 4-1BB is shown in SEQ ID NO.: 19:
  • the base sequence of the intracellular domain of CD3 ⁇ is shown in SEQ ID NO.: 20:
  • the base sequence of P2A is shown in SEQ ID NO.: 21:
  • the base sequence of type I interferon is as follows:
  • amino acid sequence of type I interferon is shown below:
  • amino acid sequence of GPC3-CAR without type I interferon is shown in SEQ ID NO.: 49:
  • GPC3-ScFv VL The base sequence of the GPC3 single-chain antibody light chain variable region (GPC3-ScFv VL) is shown in SEQ ID NO.: 50:
  • GPC3-ScFv VL The amino acid sequence of the GPC3 single-chain antibody light chain variable region (GPC3-ScFv VL) is shown in SEQ ID NO.: 51:
  • the Linker of GPC3-ScFv VL and GPC3-ScFv VH is the base sequence of SEQ ID NO.: 14 above.
  • the amino acid sequence of the Linker of GPC3-ScFv VL and GPC3-ScFv VH is shown in SEQ ID NO.: 15 above.
  • GPC3-ScFv VH The base sequence of the GPC3 single-chain antibody heavy chain variable region (GPC3-ScFv VH) is shown in SEQ ID NO.: 52:
  • GPC3-ScFv VH The amino acid sequence of the GPC3 single-chain antibody heavy chain variable region (GPC3-ScFv VH) is shown in SEQ ID NO.: 53:
  • GPC3-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 54:
  • GPC3-CAR containing type I interferon IFN ⁇ 2b
  • BCMA-ScFv VL BCMA single-chain antibody light chain variable region
  • BCMA-ScFv VL The amino acid sequence of the BCMA single-chain antibody light chain variable region (BCMA-ScFv VL) is shown in SEQ ID NO.: 58:
  • BCMA-ScFv VH The base sequence of the BCMA single-chain antibody heavy chain variable region (BCMA-ScFv VH) is shown in SEQ ID NO.: 59:
  • BCMA-ScFv VH The amino acid sequence of the BCMA single-chain antibody heavy chain variable region (BCMA-ScFv VH) is shown in SEQ ID NO.: 60:
  • BCMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 61:
  • the nucleotide sequence of BCMA-CAR containing type I interferon (IFN ⁇ 2b) is shown in SEQ ID NO.: 62:
  • PB220PA-1 The base sequence of PB transposase (PB220PA-1) is shown in SEQ ID NO.: 63:
  • PB220PA-1 The amino acid sequence of PB transposase (PB220PA-1) is shown in SEQ ID NO.: 64:

Abstract

L'invention concerne un procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type I et une application de celui-ci. L'invention concerne spécifiquement une cellule immunitaire modifiée exprimant un récepteur CAR d'antigène chimère et un interféron de type I ciblant des marqueurs de cellules tumorales. La cellule peut tuer sélectivement des cellules tumorales de manière significative.
PCT/CN2021/096191 2020-05-26 2021-05-26 Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci WO2021239020A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010456171.0 2020-05-26
CN202010456171 2020-05-26

Publications (1)

Publication Number Publication Date
WO2021239020A1 true WO2021239020A1 (fr) 2021-12-02

Family

ID=78672799

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/096191 WO2021239020A1 (fr) 2020-05-26 2021-05-26 Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci

Country Status (2)

Country Link
CN (1) CN113717942B (fr)
WO (1) WO2021239020A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024060424A1 (fr) * 2022-09-21 2024-03-28 深圳先进技术研究院 Procédé de préparation d'un bite sécrétant un car-t et son utilisation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114863993B (zh) * 2022-07-05 2022-09-27 臻和(北京)生物科技有限公司 用于结肠癌预后预测的标志物、模型构建方法和系统

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104203982A (zh) * 2011-10-28 2014-12-10 特瓦制药澳大利亚私人有限公司 多肽构建体及其用途
US20180028632A1 (en) * 2016-07-29 2018-02-01 Lan Bo Chen Method of treating tumors with nk and nkt cells expressing anti-ssea4 chimeric antigen receptors
CN109265559A (zh) * 2018-09-25 2019-01-25 山东兴瑞生物科技有限公司 嵌合抗原受体、其制备方法、利用其修饰的nk细胞及治疗hbv感染的应用
CN109306016A (zh) * 2018-08-15 2019-02-05 华东师范大学 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
CN109468278A (zh) * 2017-09-08 2019-03-15 科济生物医药(上海)有限公司 基因工程化的t细胞及应用
WO2020198654A1 (fr) * 2019-03-28 2020-10-01 Orionis Biosciences, Inc. Protéines d'interféron alpha 1 thérapeutiques
CN111850014A (zh) * 2019-06-25 2020-10-30 浙江康佰裕生物科技有限公司 一种细胞因子增效的嵌合抗原受体及其应用

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105949324B (zh) * 2016-06-30 2019-08-27 上海恒润达生生物科技有限公司 靶向gpc3的嵌合抗原受体及其用途
CN109554349B (zh) * 2017-09-27 2022-06-24 亘喜生物科技(上海)有限公司 Pd-1基因表达沉默的工程化免疫细胞
CN109554348A (zh) * 2017-09-27 2019-04-02 亘喜生物科技(上海)有限公司 可诱导分泌抗cd47抗体的工程化免疫细胞
CN109161532A (zh) * 2018-05-31 2019-01-08 华东师范大学 同时靶向psma和pd-l1的工程化免疫细胞
WO2020056228A1 (fr) * 2018-09-14 2020-03-19 Mayo Foundation For Medical Education And Research Matériels et méthodes de traitement au moyen de virus oncolytiques et de lymphocytes t car modifiés
CN109265550B (zh) * 2018-09-25 2020-09-15 华东师范大学 Bcma抗体、嵌合抗原受体和药物

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104203982A (zh) * 2011-10-28 2014-12-10 特瓦制药澳大利亚私人有限公司 多肽构建体及其用途
US20180028632A1 (en) * 2016-07-29 2018-02-01 Lan Bo Chen Method of treating tumors with nk and nkt cells expressing anti-ssea4 chimeric antigen receptors
CN109468278A (zh) * 2017-09-08 2019-03-15 科济生物医药(上海)有限公司 基因工程化的t细胞及应用
CN109306016A (zh) * 2018-08-15 2019-02-05 华东师范大学 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
CN109265559A (zh) * 2018-09-25 2019-01-25 山东兴瑞生物科技有限公司 嵌合抗原受体、其制备方法、利用其修饰的nk细胞及治疗hbv感染的应用
WO2020198654A1 (fr) * 2019-03-28 2020-10-01 Orionis Biosciences, Inc. Protéines d'interféron alpha 1 thérapeutiques
CN111850014A (zh) * 2019-06-25 2020-10-30 浙江康佰裕生物科技有限公司 一种细胞因子增效的嵌合抗原受体及其应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KRISTENM. HEGE;EMILYK. BERGSLAND;GEORGEA. FISHER;JOHNJ. NEMUNAITIS;ROBERTS. WARREN;JAMESG. MCARTHUR;ANDYA. LIN;JEFFREY SCHLOM;CARL: "Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer", JOURNAL FOR IMMUNOTHERAPY OF CANCER, BIOMED CENTRAL LTD, LONDON, UK, vol. 5, no. 1, 21 March 2017 (2017-03-21), London, UK , pages 1 - 14, XP021242925, DOI: 10.1186/s40425-017-0222-9 *
YOUNG PATRICIA A., YAMADA REIKO E., TRINH KHAM R., VASUTHASAWAT ALEX, DE OLIVEIRA SATIRO, YAMADA DOUGLAS H., MORRISON SHERIE L., T: "Activity of Anti-CD19 Chimeric Antigen Receptor T Cells Against B Cell Lymphoma Is Enhanced by Antibody-Targeted Interferon-Alpha", JOURNAL OF INTERFERON AND CYTOKINE RESEARCH., MARY ANN LIEBERT, NEW YORK, NY., US, vol. 38, no. 6, 1 June 2018 (2018-06-01), US , pages 239 - 254, XP055871604, ISSN: 1079-9907, DOI: 10.1089/jir.2018.0030 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024060424A1 (fr) * 2022-09-21 2024-03-28 深圳先进技术研究院 Procédé de préparation d'un bite sécrétant un car-t et son utilisation

Also Published As

Publication number Publication date
CN113717942A (zh) 2021-11-30
CN113717942B (zh) 2024-04-30

Similar Documents

Publication Publication Date Title
WO2021098882A1 (fr) Cellule cd7-car-t, sa préparation et son utilisation
WO2019196713A1 (fr) Récepteur antigénique chimérique ciblant bcma, son procédé de préparation et son utilisation
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
WO2010030002A1 (fr) Cellule capable d'exprimer un ligand gitr exogène
WO2017219936A1 (fr) Lymphocyte t exprimant le récepteur car capable d'exprimer avec efficacité et stabilité un anticorps activé, et ses utilisations
CN109306016B (zh) 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
CN109575143B (zh) 双特异性cd20-cd19-car及其应用
US20240082401A1 (en) Bispecific cs1-bcma car-t cell and application thereof
WO2018145648A1 (fr) Construction de car ciblant cd20 et identification d'activité de lymphocytes t génétiquement modifiées de celui-ci
CN111378625A (zh) 一种cxcl13趋化型car-t细胞的制备和应用
WO2021239020A1 (fr) Procédé d'immunothérapie pour combiner un récepteur d'antigène chimère et un interféron de type i et application de celui-ci
WO2023083192A1 (fr) Cellule immunitaire modifiée pour l'expression combinée de ccr2b et de cd40l, sa préparation et son application
WO2021136040A1 (fr) Préparation et applications de cellule t de récepteur antigénique chimérique coexprimant une molécule immunomodulatrice
CN113416260A (zh) 靶向Claudin18.2的特异性嵌合抗原受体细胞及其制备方法和应用
CN110054698B (zh) 抗cd19抗体的新型cd19-car载体的构建及应用
WO2023217192A1 (fr) Préparation d'une cellule immunitaire de récepteur antigénique chimérique construite sur la base d'une protéine précurseur de msln et son utilisation
WO2020151752A1 (fr) Cellules immunitaires modifiées ciblant une combinaison cd20
CN115806626B (zh) 一种基于csf1的嵌合抗原受体免疫细胞制备及其应用
CN109897114B (zh) 具有自杀基因开关的靶向cd47的工程化免疫细胞
WO2023161846A1 (fr) Lymphocyte t récepteur antigénique chimérique ciblant gpc3 et son utilisation
WO2021208750A1 (fr) Récepteur antigénique chimérique ciblant cd22, son procédé de préparation et son utilisation
CN115819614A (zh) 一种基于il34的嵌合抗原受体免疫细胞制备及其应用
US20210395362A1 (en) Car-t cells with humanized cd19 scfv with mutation in cdr 1 region
CN114907485A (zh) 一种以内源性蛋白质分子替代单结构域抗体的嵌合抗原受体
WO2022151959A1 (fr) Cellule car t ciblant b7-h3 et son utilisation dans le traitement de la leucémie aiguë myéloïde

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: 21813115

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: 21813115

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21813115

Country of ref document: EP

Kind code of ref document: A1