WO2023125766A1 - Récepteur antigénique chimérique ciblant cs1, récepteur antigénique chimérique bispécifique ciblant bcma/cs1 et utilisation associée - Google Patents

Récepteur antigénique chimérique ciblant cs1, récepteur antigénique chimérique bispécifique ciblant bcma/cs1 et utilisation associée Download PDF

Info

Publication number
WO2023125766A1
WO2023125766A1 PCT/CN2022/143236 CN2022143236W WO2023125766A1 WO 2023125766 A1 WO2023125766 A1 WO 2023125766A1 CN 2022143236 W CN2022143236 W CN 2022143236W WO 2023125766 A1 WO2023125766 A1 WO 2023125766A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
cells
acid sequence
scfv
Prior art date
Application number
PCT/CN2022/143236
Other languages
English (en)
Chinese (zh)
Inventor
张超
丁伟
张其猛
吕璐璐
周立
王永增
张云龙
白大勇
路佳兴
Original Assignee
合源生物科技(天津)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 合源生物科技(天津)有限公司 filed Critical 合源生物科技(天津)有限公司
Publication of WO2023125766A1 publication Critical patent/WO2023125766A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or 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
    • C07K16/46Hybrid immunoglobulins
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian

Definitions

  • the present application relates to the field of biomedicine, in particular to a chimeric antigen receptor targeting CS1, a bispecific chimeric antigen receptor targeting BCMA/CS1 and applications thereof.
  • Multiple myeloma defined as the malignant proliferation of plasma cells in the bone marrow, is the second most common hematological malignancy, accounting for 1% of all cancers. Studies have shown that multiple myeloma has a high incidence in the elderly over 60 years old and the incidence has increased steadily in recent years. For most patients, multiple myeloma is incurable and will eventually develop into relapsed/refractory multiple myeloma. The survival period of patients with relapsed/refractory multiple myeloma who are ineffective with existing multiple myeloma treatments (such as immunomodulators, proteasome inhibitors, antibody drugs) is only about 13 months.
  • multiple myeloma treatments such as immunomodulators, proteasome inhibitors, antibody drugs
  • CS1 also known as SLAMF7, CRACC or CD319
  • SLAMF7 CRACC
  • CD319 is a glycoprotein expressed on the surface of myeloma cells, which is a strong marker between normal plasma cells and malignant plasma cells in multiple myeloma.
  • Chu et al. developed two second-generation CARs for the treatment of multiple myeloma, one target antigen is BCMA, and the other target antigen is CS1; in vitro experiments, although two CAR-Ts with different targets showed Similar killing activity, but in vivo experiments in mice, CS1 CAR-T cells have stronger anti-tumor activity.
  • a chimeric antigen receptor (Chimeric Antigen Receptor, CAR) is the core component of a CAR cell therapy drug, which may include a targeting moiety (for example, a part that binds a tumor-associated antigen (Tumor-Associated Antigen, TAA)), a hinge region, a spanning Membrane domains and intracellular domains.
  • CAR-T cell immunotherapy is considered to be one of the most promising means to overcome tumors.
  • CAR-T cells use genetic modification to enable T cells to express CAR proteins. This CAR protein has the ability to recognize the intact protein on the membrane surface without relying on antigen presentation, thereby causing the activation and functional effects of T cells.
  • the present application provides a chimeric antigen receptor targeting CS1, a bispecific chimeric antigen receptor targeting BCMA/CS1 and applications thereof.
  • the inventors used multiple CS1-targeting scFv antibodies to construct chimeric antigen receptor expression vectors and prepare CS1-targeting CAR-T cells, and also verified that CS1 CAR-T cells have good tumor-suppressing functions and Determination of the optimal chimeric antigen receptor targeting CS1.
  • a chimeric antigen receptor targeting CS1 comprising an extracellular antigen recognition domain, a hinge region, a transmembrane region and an intracellular domain; wherein: the extracellular antigen recognition domain comprises an anti-CS1 scFv antibody,
  • the amino acid sequences of the VH complementarity-determining regions CDR1, CDR2, and CDR3 of the scFv antibody include the amino acid sequences shown in SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the VL complementarity-determining regions of the scFv antibody
  • the amino acid sequences of regions CDR1, CDR2, and CDR3 include the amino acid sequences shown in SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6, respectively.
  • the VH sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO: 7
  • the VL sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO: 8. amino acid sequence.
  • the scFv antibody is a humanized antibody.
  • the VH sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO: 9
  • the VL sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO: 10. amino acid sequence.
  • the scFv antibody is a rabbit-derived antibody.
  • connecting region between VH and VL in the scFv antibody, and the connecting region is selected from one or more of the following: SEQ ID NOs: 59-61.
  • the sequence of the scFv antibody is shown in SEQ ID NO: 11 or SEQ ID NO: 12.
  • the hinge region is derived from one or more of IgG1, IgG4, CD4, CD7, CD28, CD84, and CD8 ⁇ .
  • the above-mentioned chimeric antigen receptor, the transmembrane region is derived from one of CD3, CD4, CD7, CD8 ⁇ , CD28, CD80, CD86, CD88, 4-1BB, CD152, OX40, Fc70 or Various.
  • the intracellular domain includes an intracellular signal transduction region; optionally, it also includes a co-stimulatory signal transduction region.
  • the above-mentioned chimeric antigen receptor wherein the intracellular signaling region is derived from CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, FcR ⁇ , FcR ⁇ , CD66d, DAP10, DAP12, Syk one or more of.
  • the co-stimulatory signaling region is derived from CD2, CD3, CD7, CD27, CD28, CD30, CD40, CD83, CD244, 4-1BB, OX40, LFA-1
  • the co-stimulatory signaling region is derived from CD2, CD3, CD7, CD27, CD28, CD30, CD40, CD83, CD244, 4-1BB, OX40, LFA-1
  • ICOS LIGHT, NKG2C, NKG2D, DAP10, B7-H3, MyD88.
  • the above chimeric antigen receptor further comprises a leader peptide located at the N-terminal of the amino acid sequence of the chimeric antigen receptor; optionally, the leader peptide is derived from CD8 ⁇ .
  • the extracellular antigen recognition domain further comprises scFv antibodies against one of the following targets: CD138, NKG2D, CD38, BCMA, CD19, CD70, CD44v6, Lewis Y.
  • the extracellular antigen recognition domain sequentially comprises anti-BCMA scFv VL, anti-CS1 scFv VL, anti-CS1 scFv VH and anti-BCMA scFv VH, the anti-
  • the amino acid sequences of the scFv VH complementarity-determining regions CDR1, CDR2, and CDR3 of CS1 include the amino acid sequences shown in SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 respectively, and the scFv VL complementarity-determining region of the anti-CS1
  • the amino acid sequences of CDR1, CDR2, and CDR3 respectively include the amino acid sequences shown in SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6, and the amino acids of the scFv VH complementarity determining regions CDR1, CDR2, and CDR3 of the anti-BCMA
  • the sequences include the amino acid sequences shown in SEQ ID NO:
  • the VH sequence of the anti-CS1 scFv antibody includes the amino acid sequence shown in SEQ ID NO: 7, and the VL sequence of the anti-CS1 scFv antibody includes SEQ ID Amino acid sequence shown in NO:8.
  • the VH sequence of the scFv antibody against BCMA comprises the amino acid sequence shown in SEQ ID NO: 57
  • the VL sequence of the scFv antibody against BCMA comprises the sequence of SEQ ID NO: The amino acid sequence shown in NO:58.
  • the extracellular antigen recognition domain includes the amino acid sequence shown in SEQ ID NO:50.
  • the present application also provides an isolated nucleic acid molecule comprising the nucleotide sequence encoding the above-mentioned chimeric antigen receptor.
  • the present application also provides a vector comprising the above-mentioned isolated nucleic acid molecule.
  • the above-mentioned vector is an expression vector; in some embodiments, the vector is a viral vector; in some embodiments, it is a lentiviral vector.
  • the present application also provides an engineered immune effector cell, which comprises the above-mentioned chimeric antigen receptor, the above-mentioned isolated nucleic acid molecule, or the above-mentioned carrier.
  • the immune effector cells are selected from T lymphocytes, natural killer cells (NK cells), peripheral blood mononuclear cells (PBMC cells), pluripotent stem cells, and differentiated pluripotent stem cells.
  • the above-mentioned immune effector cells are T lymphocytes; optionally, the source of the T lymphocytes is autologous T lymphocytes or allogeneic T lymphocytes.
  • the present application also provides a pharmaceutical composition, which includes the above-mentioned engineered immune effector cells and pharmaceutically acceptable auxiliary materials.
  • the pharmaceutically acceptable excipients include protective agents.
  • the pharmaceutically acceptable adjuvant includes cell cryopreservation solution.
  • the above-mentioned pharmaceutical composition is an intravenous injection.
  • the present application also provides the use of the above-mentioned chimeric antigen receptor, nucleic acid molecule, carrier or immune effector cell in the preparation of medicines for treating diseases or conditions related to the expression of CS1.
  • the disease or disease associated with the expression of CS1 is cancer; optionally, the cancer is multiple myeloma; further optionally, the cancer is refractory or recurrent multiple myeloma.
  • the disease or disorder associated with the expression of CS1 may be an autoimmune disease.
  • the autoimmune disease may be selected from the following: systemic lupus erythematosus, rheumatoid arthritis, idiopathic thrombocytopenic purpura, myasthenia gravis or autoimmune hemolytic anemia.
  • the drug is an intravenous injection.
  • the present application also provides a method for treating diseases or disorders related to the expression of CS1, comprising the following steps: administering an effective amount of the above-mentioned immune effector cells or the pharmaceutical composition to a disease or disorder associated with the expression of CS1. subjects in need.
  • the disease or disorder associated with the expression of CS1 is cancer; optionally, the cancer is multiple myeloma; further optionally, the cancer is refractory or recurrent multiple myeloma.
  • the disease or condition associated with the expression of CS1 may be an autoimmune disease.
  • the autoimmune disease may be selected from the group consisting of systemic lupus erythematosus, rheumatoid arthritis, idiopathic thrombocytopenic purpura, myasthenia gravis, or autoimmune hemolytic anemia.
  • the administration is by intravenous injection.
  • the administering method is to administer an effective amount of the immune effector cells or the pharmaceutical composition to the subject in a single injection.
  • the effective amount of immune effector cells or the pharmaceutical composition is 1 ⁇ 10 5 to 1 ⁇ 10 7 cells/kg.
  • the present application also provides the above-mentioned immune effector cells or the above-mentioned pharmaceutical composition for treating diseases or diseases related to the expression of CS1.
  • the above-mentioned immune effector cells or the above-mentioned pharmaceutical composition, the disease or disease related to the expression of CS1 is cancer; optionally, the cancer is multiple myeloma; further optionally, the cancer is Refractory or relapsed multiple myeloma.
  • the disease or disorder associated with the expression of CS1 may be an autoimmune disease.
  • the autoimmune disease can be selected from the following: systemic lupus erythematosus, rheumatoid arthritis, idiopathic thrombocytopenic purpura, myasthenia gravis or autoimmune Immune hemolytic anemia.
  • Figure 1 shows a schematic diagram of the CS1 CAR structure in Example 1 of the present application.
  • Figure 2 shows the staining of UTD cells (T cells not transduced with CAR), CS1 CAR-T cells 21G-27G, and positive control CS1 CAR-T cells LUC90V2 with PE fluorescence-labeled CS1 antigen in Example 2 of the present application. The results of detecting the percentage of CAR molecules expressed on the surface.
  • Figure 3 shows the results of staining UTD cells, CS1 CAR-T cells 21G-27G, and positive control CAR-T cells 02G with fluorescently-labeled anti-human IgG antibodies in Example 2 of the present application to detect the percentage of CAR molecules expressed on their surfaces picture.
  • Figure 4 shows the release of IL-2 after CS1 CAR-T cells were activated by CS1 positive target cells detected in Example 3 of the present application.
  • K562 cells and K562-CS1 cells from left to right are UTD, 21G cells, The release of IL-2 from 22G cells, 23G cells, 24G cells, 25G cells and 26G cells.
  • Figure 5 shows the killing effect of CS1 CAR-T cells on CS1 positive target cells in Example 3 of the present application.
  • K562 cells and K562-CS1 cells from left to right are the killing effects of UTD, 21G cells, 22G cells, 23G cells, 24G cells, 25G cells, 26G cells and positive control LUC90V2 cells on target cells.
  • Figure 6A shows the continuous proliferation of CD3+ cells after CS1 CAR-T cells are activated by multiple rounds of antigen stimulation in Example 4 of the present application
  • Figure 6B shows the activation of CS1 CAR-T cells by multiple rounds of antigen stimulation in Example 4 of the present application , Continuous proliferation of CAR+ cells.
  • Fig. 7 shows a schematic diagram of the construction of the BCMA-CS1 bispecific CAR in Example 5 of the present application.
  • Figure 8 shows the results of flow cytometry for the detection of the positive rate of BCMA-CS1 bispecific CAR-T cells in each group in Example 6 of the present application.
  • Figure 9A, Figure 9B, and Figure 9C show the effects of BCMA-CS1 bispecific CAR-T cells, negative control cells, and positive control cells in Example 7 of the present application on double-negative target cells K562 and exogenous BCMA-positive cells K562- BCMA, exogenous CS1 positive cell K562-CS1 cell killing experiment results.
  • CAR Chimeric Antigen Receptor
  • extracellular antigen recognition domain refers to the antigen recognition domain (Antigen Recognition Domain, ARD).
  • CAR cell therapy products such as CAR-T cells
  • ARD Antigen Recognition Domain
  • CAR-T cells CAR-T cells
  • TCR Chain variable region
  • VLR variable lymphocyte receptors
  • the scFv antibody refers to the scFv antibody targeting CS1.
  • scFv antibodies can include one, two or more antibody heavy chain variable regions (VH) and one, two or more light chain variable regions (VL), connected by a peptide chain , such as: the connecting sequence GSTSGSGKPGSGEGSTKG composed of 18 amino acids.
  • VH antibody heavy chain variable regions
  • VL light chain variable regions
  • the regions are called hypervariable regions (HVR); in the V regions of the L chain and H chain
  • HVR hypervariable regions
  • CDRs complementarity determining regions
  • CDR division rules include Kabat, AbM, Chothia, Contact, and IMGT. These rules are well known to those skilled in the art. When applying the website that implements these rules, just input the VH and VL sequences and select the corresponding rules. CDR sequences according to different rules can be obtained. Those skilled in the art should understand that the protection scope of the present application covers the combination of CDR sequences obtained by analysis using different rules. In this application, the CDR is divided according to the IMGT rule.
  • telomere binding refers to the recognition and/or binding between CAR and a specific target, with greater affinity, avidity, easier, and/or bind the target for a greater duration.
  • humanized antibody is also referred to as a humanized engineered antibody, which is obtained by combining the complementarity-determining regions (CDRs) of non-human mammalian antibodies, such as mouse antibodies, rat antibodies, and rabbit antibodies. ) are transplanted into the CDRs of human antibodies for preparation, and conventional recombinant DNA techniques for preparing humanized antibodies are known (eg WO96/02576).
  • CDRs complementarity-determining regions
  • FRs framework regions
  • hinge region refers to the link between the extracellular antigen recognition domain and the transmembrane domain. This region allows CAR to recognize antigen by giving the antigen recognition domain a certain range of motion.
  • Currently used hinge regions are mainly derived from one or more of IgG1, IgG4, CD4, CD7, CD28, CD84, and CD8 ⁇ .
  • the typical hinge region also contains residues that are involved in CAR dimerization and contribute to enhanced antigen sensitivity.
  • transmembrane region refers to the transmembrane domain that connects the intracellular and extracellular components of the CAR structure. Different transmembrane domains can affect the expression and stability of CAR to a certain extent, but they are not directly involved in signal transmission, and the downstream signal transmission can be improved through interaction.
  • the transmembrane region can be derived from one or more of CD3, CD4, CD7, CD8 ⁇ , CD28, CD80, CD86, CD88, 4-1BB, CD152, OX40, and Fc70.
  • intracellular domain includes intracellular signaling regions and may also include co-stimulatory signaling regions.
  • intracellular signaling region refers to the activation of at least one normal effector function of an immune effector cell responsible for the expression of CAR.
  • the intracellular signaling region can be derived from one or more of CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, FcR ⁇ , FcR ⁇ , CD66d, DAP10, DAP12, and Syk.
  • the term "co-stimulatory signaling domain" exists because, in addition to the stimulation of antigen-specific signals, many immune effector cells also require co-stimulation to promote cell proliferation, differentiation and survival, and activation of cell activation. Effector function.
  • the CAR may also include one or more co-stimulatory signaling regions, wherein the co-stimulatory signaling regions may be derived from CD2, CD3, CD7, CD27, CD28, CD30, CD40, CD83, CD244, 4- One, two or more of 1BB, OX40, LFA-1, ICOS, LIGHT, NKG2C, NKG2D, DAP10, B7-H3, MyD88.
  • isolated generally means obtained from the natural state by artificial means. If an "isolated" substance or component occurs in nature, it may be that its natural environment has been altered, the substance has been isolated from its natural environment, or both. For example, an unisolated polynucleotide or polypeptide naturally exists in a living animal, and the same polynucleotide or polypeptide with high purity isolated from this natural state is called isolation. of.
  • isolated does not exclude artificial or synthetic substances obtained from the natural state by artificial means, nor does it exclude the presence of other impure substances that do not affect the activity of the substance.
  • guide peptide refers to a short peptide before the extracellular antigen recognition domain (such as scFv sequence), whose function is to guide the export of the recombinant protein synthesized in the cell to the outside of the cell.
  • scFv sequence extracellular antigen recognition domain
  • Commonly used guide peptides are human CD8 ⁇ signal peptide, or human GM-CSF receptor ⁇ signal peptide.
  • CS1 is also called SLAMF7 (signaling-lymphocyte-activating molecule F7) or CD319, which is a glycoprotein on the cell surface, expressed in plasma cells, NK cells, CD8+ T cells, activated In normal tissues such as B cells and dendritic cells, but not expressed in hematopoietic stem cells and non-hematopoietic organs, it can participate in the mutual adhesion between myeloma cells and bone marrow stromal cells.
  • SLAMF7 signalaling-lymphocyte-activating molecule F7
  • CD319 which is a glycoprotein on the cell surface, expressed in plasma cells, NK cells, CD8+ T cells, activated In normal tissues such as B cells and dendritic cells, but not expressed in hematopoietic stem cells and non-hematopoietic organs, it can participate in the mutual adhesion between myeloma cells and bone marrow stromal cells.
  • BCMA refers to B cell maturation antigen, a member of the tumor necrosis factor receptor superfamily. Human BCMA is expressed almost exclusively in plasma cells and multiple myeloma cells. BCMA may be a suitable tumor antigen target for immunotherapeutics against multiple myeloma. However, due to the heterogeneity of specific antigens on the surface of multiple myeloma cells, the selection of its antigen target is not necessarily single. By selecting appropriate targets, the anti-tumor activity of CAR-T cells can be optimized.
  • isolated nucleic acid molecule generally refers to an isolated form of nucleotides, deoxyribonucleotides or ribonucleotides of any length, which may be isolated from its natural environment or a synthetic analogue .
  • gene transduction/transfection methods mainly include viral and non-viral methods. Such as: through ⁇ -retroviral vectors, lentiviral vectors, adeno-associated viral vectors, plasmid DNA-dependent vectors, transposon-dependent gene transfer, and mRNA-mediated gene transduction.
  • vector generally refers to a nucleic acid delivery tool into which a polynucleotide encoding a protein can be inserted and the protein can be expressed.
  • the vector can transform, transduce or transfect the host cell, so that the genetic material elements carried by it can be expressed in the host cell.
  • vectors include: plasmids; phagemids; cosmids; artificial chromosomes such as yeast artificial chromosome (YAC), bacterial artificial chromosome (BAC) or P1-derived artificial chromosome (PAC); phage such as lambda phage or M13 phage and animal viruses.
  • Types of animal viruses used as vectors include retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpesviruses (such as herpes simplex virus), poxviruses, baculoviruses, papillomaviruses, papillary polyoma vacuoles Viruses (such as SV40).
  • retroviruses including lentiviruses
  • adenoviruses such as herpes simplex virus
  • poxviruses such as herpes simplex virus
  • baculoviruses such as herpes simplex virus
  • baculoviruses such as baculoviruses
  • papillomaviruses such as SV40
  • a vector may contain a variety of elements that control expression, including promoter sequences, transcription initiation sequences, enhancer sequences, selection elements, and reporter genes.
  • the vector may also contain an origin of replication.
  • Vectors may also
  • transposon refers to a discontinuous DNA segment that has the ability to migrate between chromosomal sites and carry genetic information, such as: Sleeping Beauty SB system and PB system derived from Lepidoptera insects.
  • electroporation can also be used to transduce mRNA into T cells.
  • Immune effector cell generally refers to a cell that participates in an immune response, eg, promotes an immune effector response.
  • Immune effector cells may be selected from the group consisting of: T lymphocytes, natural killer cells (NK cells), peripheral blood mononuclear cells (PBMC cells), pluripotent stem cells, T lymphocytes differentiated from pluripotent stem cells, NK cells, induced pluripotent stem cells (iPSC), T cells differentiated from induced pluripotent stem cells (iPSC-T), NK cells differentiated from induced pluripotent stem cells (iPSC-NK), embryonic stem cells one or more species.
  • NK cells natural killer cells
  • PBMC cells peripheral blood mononuclear cells
  • pluripotent stem cells T lymphocytes differentiated from pluripotent stem cells
  • NK cells induced pluripotent stem cells
  • iPSC induced pluripotent stem cells
  • iPSC-T T cells differentiated from induced pluripotent stem cells
  • the term "pharmaceutical composition” generally refers to a pharmaceutical composition suitable for administration to patients, which may contain the immune effector cells described in this application, and may also contain one or more pharmaceutically acceptable excipients, such as : one or more of carrier, protective agent, stabilizer, excipient, diluent, solubilizer, surfactant, emulsifier, preservative.
  • the pharmaceutically acceptable excipients include protective agents, such as cell cryopreservation solution.
  • the pharmaceutical composition of the present application is a cell suspension or frozen cells thereof.
  • subject generally refers to human or non-human animals, including but not limited to mice, rats, cats, dogs, rabbits, horses, pigs, cows, sheep or monkeys.
  • the term "about” usually refers to the fluctuation range acceptable to those skilled in the art above or below the specified value, such as: within the range of ⁇ 0.5%-10%, for example, 0.5% above or below the specified value %, 1%, 1.5%, 2%, 2.5%, 3%, 3.5%, 4%, 4.5%, 5%, 5.5%, 6%, 6.5%, 7%, 7.5%, 8%, 8.5%, 9%, 9.5% or 10% range.
  • Chimeric antigen receptors nucleic acids, vectors, immune effector cells, pharmaceutical compositions
  • the present application provides a chimeric antigen receptor targeting CS1, which comprises an extracellular antigen recognition domain, a hinge region, a transmembrane region and an intracellular domain; wherein: the extracellular antigen recognition domain comprises Anti-CS1 scFv antibody, the amino acid sequences of the VH complementarity determining regions CDR1, CDR2, and CDR3 of the scFv antibody include the amino acid sequences shown in SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3 respectively, and the The amino acid sequences of the VL complementarity determining regions CDR1, CDR2, and CDR3 of the scFv antibody include the amino acid sequences shown in SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively.
  • the VH sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO:7
  • the VL sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO:8.
  • the scFv antibody is a humanized antibody.
  • the VH sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO:9
  • the VL sequence of the scFv antibody includes the amino acid sequence shown in SEQ ID NO:10.
  • the scFv antibody is a rabbit antibody.
  • connecting region between VH and VL in the scFv antibody, and the connecting region is selected from one or more of the following: SEQ ID NOs: 59-61.
  • sequence of the scFv antibody is shown in SEQ ID NO: 11 or SEQ ID NO: 12.
  • the present application also includes substitution, deletion, addition and/or insertion of one or more amino acids in the amino acid sequence of any one of the above chimeric antigen receptors, and it has the equivalent of any one of the above Chimeric antigen receptor activity; optionally, the substitutions are conservative substitutions.
  • substitutions are conservative substitutions.
  • the humanized transformation of the FR region in the scFv is different from the amino acid sequence shown in SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9 or SEQ ID NO:10 .
  • those skilled in the art also know that in order to make the CDR region of the modified antibody retain a suitable antigen-binding site during the process of humanization, if necessary, one, two, three or none of the CDRs More than 10% of the amino acid sequence may be substituted, deleted, added and/or inserted, and these are also included in this application.
  • the hinge region is derived from one or more of IgG1, IgG4, CD4, CD7, CD28, CD84, and CD8 ⁇ ; optionally, the amino acid sequence of the hinge region is derived from CD8 ⁇ ; further optionally, the amino acid sequence of the hinge region comprises the amino acid sequence shown in SEQ ID NO: 13.
  • the transmembrane region is derived from one or more of CD3, CD4, CD7, CD8 ⁇ , CD28, CD80, CD86, CD88, 4-1BB, CD152, OX40, Fc70; alternatively, The amino acid sequence of the transmembrane region is derived from CD8 ⁇ ; further optionally, the amino acid sequence of the transmembrane region comprises the amino acid sequence shown in SEQ ID NO:14.
  • the intracellular domain comprises an intracellular signaling region; optionally, also includes a co-stimulatory signaling region; further optionally, wherein the intracellular signaling region is derived from CD3 ⁇ , CD3 ⁇ , one or more of CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, FcR ⁇ , FcR ⁇ , CD66d, DAP10, DAP12, Syk; still further optionally, the intracellular signaling region is derived from CD3 ⁇ ,
  • the amino acid sequence of the intracellular signal transduction region comprises the amino acid sequence shown in SEQ ID NO:15.
  • the co-stimulatory signaling region is derived from CD2, CD3, CD7, CD27, CD28, CD30, CD40, CD83, CD244, 4-1BB, OX40, LFA-1, ICOS, LIGHT, NKG2C, One, two, or more than three of NKG2D, DAP10, B7-H3, and MyD88; optionally, the costimulatory signal transduction region is derived from CD28 or 4-1BB; further optionally, the costimulatory signal transduction region
  • the amino acid sequence comprises the amino acid sequence shown in SEQ ID NO:16.
  • the chimeric antigen receptor further comprises a guide peptide located at the N-terminal of the chimeric antigen receptor amino acid sequence; optionally, wherein the guide peptide is derived from CD8 ⁇ ; further optionally, The amino acid sequence of the leader peptide comprises the amino acid sequence shown in SEQ ID NO:17.
  • the chimeric antigen receptor of the present invention comprises the amino acid sequence shown in SEQ ID NO:32.
  • the extracellular antigen recognition domain only comprises a scFv antibody targeting a single CS1 target.
  • the extracellular antigen recognition domain further comprises scFv antibodies against any of the following targets: CD138, NKG2D, CD38, BCMA, CD19, CD70, CD44v6, Lewis Y.
  • the extracellular antigen recognition domain sequentially comprises anti-BCMA scFv VL, anti-CS1 scFv VL, anti-CS1 scFv VH and anti-BCMA scFv VH, and the anti-CS1 scFv VH complementarity determining region
  • the amino acid sequences of CDR1, CDR2, and CDR3 respectively include the amino acid sequences shown in SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3, and the amino acids of the scFv VL complementarity determining regions CDR1, CDR2, and CDR3 of the anti-CS1
  • the sequences include the amino acid sequences shown in SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6, respectively, and the amino acid sequences of the anti-BCMA scFv VH complementarity determining regions CDR1, CDR2, and CDR3 include SEQ ID NO: 51.
  • amino acid sequences shown in SEQ ID NO:52 and SEQ ID NO:53 the amino acid sequences of the anti-BCMA scFv VL complementarity determining regions CDR1, CDR2, and CDR3 respectively include SEQ ID NO:54, SEQ ID NO:55 , the amino acid sequence shown in SEQ ID NO:56.
  • the VH sequence of the anti-CS1 scFv antibody includes the amino acid sequence shown in SEQ ID NO:7
  • the VL sequence of the anti-CS1 scFv antibody includes the amino acid sequence shown in SEQ ID NO:8 sequence.
  • the VH sequence of the anti-BCMA scFv antibody includes the amino acid sequence shown in SEQ ID NO:57
  • the VL sequence of the anti-BCMA scFv antibody includes the amino acid sequence shown in SEQ ID NO:58 sequence.
  • the extracellular antigen recognition domain includes the amino acid sequence shown in SEQ ID NO:50.
  • the present application also provides an isolated nucleic acid molecule comprising the nucleotide sequence encoding the above-mentioned chimeric antigen receptor.
  • the application provides an isolated nucleic acid molecule encoding a chimeric antigen receptor, which is selected from one of the following nucleic acid molecules: comprising the nucleotide sequence shown in SEQ ID NO: 18, or Nucleic acid molecule that is similar to the nucleotide sequence shown in :18 and encodes the same chimeric antigen receptor nucleotide sequence.
  • the similarity to the nucleotide sequence shown in SEQ ID NO: 18 refers to having at least 70%, 75%, 80%, 85% of the nucleotide sequence shown in SEQ ID NO: 18 , 90%, 95%, 96%, 97%, 98% or 99% sequence identity.
  • the nucleotide sequence shown in SEQ ID NO: 18 is similar to that shown in SEQ ID NO: 18 due to the wobble (degeneracy) of the third base of the nucleic acid codon.
  • Nucleotide sequences are different, but they encode the same chimeric antigen receptor nucleic acid molecule.
  • the nucleic acid molecule comprises the nucleotide sequence shown in SEQ ID NO:39.
  • the present application also provides a vector comprising the above-mentioned isolated nucleic acid molecule.
  • Vectors include: plasmids; phagemids; cosmids; artificial chromosomes such as yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC) or P1-derived artificial chromosomes (PAC); phages such as lambda phage or M13 phage and animal viruses, etc. .
  • Types of animal viruses used as vectors include retroviruses (including lentiviruses), adenoviruses, adeno-associated viruses, herpesviruses (such as herpes simplex virus), poxviruses, baculoviruses, papillomaviruses, papillary polyoma vacuoles Viruses (such as SV40).
  • the vector is an expression vector; optionally, the vector is a viral vector; further optionally, a lentiviral vector.
  • the present application also provides an engineered immune effector cell, which comprises the above-mentioned chimeric antigen receptor, the above-mentioned isolated nucleic acid molecule, or the above-mentioned carrier.
  • the immune effector cells are selected from T lymphocytes, natural killer cells (NK cells), peripheral blood mononuclear cells (PBMC cells), pluripotent stem cells, T cells differentiated from pluripotent stem cells, pluripotent NK cells differentiated from stem cells, induced pluripotent stem cells (iPSC), T cells differentiated from induced pluripotent stem cells (iPSC-T), NK cells differentiated from induced pluripotent stem cells (iPSC-NK), embryonic stem cells one or more of.
  • NK cells natural killer cells
  • PBMC cells peripheral blood mononuclear cells
  • pluripotent stem cells T cells differentiated from pluripotent stem cells
  • pluripotent NK cells differentiated from stem cells induced pluripotent stem cells (iPSC)
  • iPSC-T T cells differentiated from induced pluripotent stem cells
  • iPSC-NK NK cells differentiated from induced pluripotent stem cells
  • the immune effector cells are T lymphocytes; optionally, the source of the T lymphocytes is autologous T lymphocytes or allogeneic T lymphocytes.
  • the surface of the immune effector cells may express the chimeric antigen receptors described herein.
  • the present application also provides a pharmaceutical composition, which includes the above-mentioned engineered immune effector cells and pharmaceutically acceptable auxiliary materials.
  • the pharmaceutically acceptable auxiliary materials include: one or more of carriers, protective agents, stabilizers, excipients, diluents, solubilizers, surfactants, emulsifiers, and preservatives.
  • the pharmaceutically acceptable excipients include protective agents, such as cell cryopreservation solution.
  • the pharmaceutical composition is a cell suspension or frozen cells thereof.
  • the pharmaceutical composition is an intravenous injection.
  • the present application also provides a method for preparing immune effector cells, which includes the following steps: transducing the vector described in the present application into the immune effector cells.
  • the immune effector cells are selected from T lymphocytes, natural killer cells (NK cells), peripheral blood mononuclear cells (PBMC cells), pluripotent stem cells, T cells differentiated from pluripotent stem cells, pluripotent NK cells differentiated from stem cells, induced pluripotent stem cells (iPSC), T cells differentiated from induced pluripotent stem cells (iPSC-T), NK cells differentiated from induced pluripotent stem cells (iPSC-NK), embryonic stem cells one or more of.
  • NK cells natural killer cells
  • PBMC cells peripheral blood mononuclear cells
  • pluripotent stem cells T cells differentiated from pluripotent stem cells
  • pluripotent NK cells differentiated from stem cells induced pluripotent stem cells (iPSC)
  • iPSC-T T cells differentiated from induced pluripotent stem cells
  • iPSC-NK NK cells differentiated from induced pluripotent stem cells
  • the immune effector cells are T lymphocytes; optionally, the source of the T lymphocytes is autologous T lymphocytes or allogeneic T lymphocytes.
  • the present application also provides the use of the chimeric antigen receptor described in the present application, the nucleic acid molecule, the carrier and/or the immune effector cell for the preparation of a drug, wherein the drug For use in the treatment of a disease or condition associated with the expression of CS1.
  • the present application also provides a method for treating a disease or disorder associated with the expression of CS1, the method comprising administering an effective dose of the present invention to a subject in need of treating a disease or disorder associated with the expression of CS1.
  • the chimeric antigen receptor, the nucleic acid molecule, the carrier and/or the immune effector cell are provided.
  • the administration can be performed by different means, such as intravenous, intratumoral, intraperitoneal, subcutaneous, intramuscular, topical or intradermal administration.
  • the mode of administration may be administered to a subject by intravenous injection.
  • the effective dose of immune effector cells or the pharmaceutical composition can be administered to the subject once, or dividedly administered to the subject within a certain period of time, such as once a week, once every two weeks, Once every three weeks, once every four weeks, once a month, once every three months, or once every three to six months.
  • the dosage may be different; for patients with different disease severity, the dosage may also be different.
  • the administered dose may range from 1 ⁇ 10 5 CAR-positive T cells/kg to 1 ⁇ 10 7 CAR-positive T cells/kg, for example, 1 ⁇ 10 5 CAR-positive T cells/kg to 1 ⁇ 10 6 CAR-positive T cells/kg, 1 ⁇ 10 6 CAR-positive T cells/kg to 1 ⁇ 10 7 CAR-positive T cells/kg.
  • the administered dose can also be measured by the total administered dose, for example: the total administered dose does not exceed 5 ⁇ 10 8 CAR-positive T cells. In this application, the administration dose is based on the count of CAR-positive T cells, and details will not be repeated in the specific examples.
  • the subjects can include humans and non-human animals.
  • the subject may include, but is not limited to, mice, rats, cats, dogs, horses, pigs, cows, sheep, rabbits or monkeys.
  • the present application also provides the chimeric antigen receptor, the nucleic acid molecule, the vector and/or the immune effector cell, which can be used to treat diseases related to the expression of CS1 or illness.
  • the disease or condition associated with the expression of CS1 may include non-solid tumors, and optionally, the non-solid tumors are hematological tumors.
  • the disease or disorder associated with the expression of CS1 may include multiple myeloma.
  • the multiple myeloma is relapsed or refractory multiple myeloma.
  • the disease or disorder associated with expression of BCMA may be an autoimmune disease.
  • the autoimmune disease may be selected from the group consisting of systemic lupus erythematosus, rheumatoid arthritis, idiopathic thrombocytopenic purpura, myasthenia gravis, or autoimmune hemolytic anemia.
  • the following examples are only for explaining the chimeric antigen receptor, immune effector cell, preparation method and application of the present application, and are not intended to limit the scope of the present invention.
  • the Examples do not include detailed descriptions of conventional methods, such as those used to construct vectors and plasmids, to insert genes encoding proteins into such vectors and plasmids, or to introduce plasmids into host cells.
  • Such methods are well known to those of ordinary skill in the art and are described in numerous publications, including Sambrook, J., Fritsch, E.F. and Maniais, T. (1989) Molecular Cloning: A Laboratory Manual , 2nd edition, Cold Spring Harbor Laboratory Press.
  • the above-mentioned 21G ⁇ The nucleotide sequences of 27G scFv are as SEQ ID NO:25, SEQ ID NO:18, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30 shown). Since the functional verification of scFv at the protein level cannot reflect its function at the cellular level, we chose to screen candidate CS1 scFv sequences on the second-generation CAR structure. The schematic diagram of the CAR structure is shown in Figure 1.
  • CD8 ⁇ guide chain As the signal peptide (its amino acid sequence is shown in SEQ ID NO: 17), CS1scFv as the extracellular tumor antigen recognition region, and CD8 ⁇ as the hinge region and transmembrane region. Structure (the amino acid sequence is shown in SEQ ID NO:13 and SEQ ID NO:14), 4-1BB is the intracellular co-stimulatory signal (the amino acid sequence is shown in SEQ ID NO:16), and CD3 ⁇ is the T cell Activation signal (its amino acid sequence is shown in SEQ ID NO: 15).
  • the three packaging plasmids are pMD2.0G (purchased from Biovector, product number Biovector012259), pMDLg-/pRRE (purchased from Biovector, product number Biovector012251), and pRSV-Rev (purchased from Biovector, product number Biovector012253).
  • PBMC Peripheral blood mononuclear cells
  • the isolated T cells were treated with complete lymphocyte culture medium (X-VIVO15 medium+5%FBS+300IU/ml IL-2 or X-VIVO15 medium+5%FBS+5ng/ml IL-15+10ng/ml IL -7) Resuspend to make the final concentration (1 ⁇ 2) ⁇ 10 6 cells/ml, and add 5 ⁇ 10 ⁇ l of CD3/CD28 magnetic beads to stimulate, mix well and place in the incubator for culture, the culture condition is 37 °C + 5% CO 2 , the incubation time is at least 24 hours.
  • complete lymphocyte culture medium X-VIVO15 medium+5%FBS+300IU/ml IL-2 or X-VIVO15 medium+5%FBS+5ng/ml IL-15+10ng/ml IL -7) Resuspend to make the final concentration (1 ⁇ 2) ⁇ 10 6 cells/ml, and add 5 ⁇ 10 ⁇ l of CD3/CD28 magnetic beads to stimulate, mix well and place in the incubator for culture, the culture condition is 37 °C + 5%
  • polybrene polybrene
  • MOI lentiviral vector
  • the transduced cells were taken out, and the cell density was monitored to maintain the cells at (0.5-1) ⁇ 10 6 cells/ml for use in subsequent examples.
  • T cells obtained after infecting T cells with lentiviruses containing 21G-27G scFv were named CS1 CAR-T cells 21G-27G, respectively.
  • CS1 CAR-T cells 21G-27G were named.
  • the CAR protein molecules expressed on the surface of the 21G-27G CS1 CAR-T cells obtained in Example 1 were detected.
  • PE fluorescence-labeled CS1 antigen manufactured by ACRO Biosystems, product number: SL7-HP2H3
  • UTD cells T cells not transduced with CAR
  • LUC90V2 CAR-T cells LUC90V2 is Positive control CS1 CAR sequence
  • the CAR sequence is shown in SEQ ID NO: 63
  • the scFv in the CAR sequence is a mouse sequence
  • the method for obtaining the CAR-T cells is also the method in Example 1), by flow cytometry Cytometry was used to detect and analyze the positive ratio of CAR.
  • BCMA CAR-T cell 02G containing another humanized scFv was used as a positive control in this method (the sequence of BCMA 02G CAR is as follows: shown in SEQ ID NO:62).
  • Example 3 CS1 CAR-T cells were subjected to cytokine release experiments and cell killing experiments
  • Cytokine release experiment The CS1 CAR-T cells 21G-26G obtained in Example 1 (because there is no CAR protein expression on the surface of CS1 CAR-T cells 27G, so this cell will not be involved in subsequent experiments) and target cells After co-cultivating in X-VIVO15 medium for 24 hours according to the condition that the effect-to-target ratio of CAR+ cells and target cells was 1:1, the concentration of IL-2 in the cell supernatant was detected by ELISA method (manufacturer: Dakowi, article number: 1110202).
  • K562 is the CS1-negative target cell
  • K562-CS1 is the positive target cell expressing CS1 exogenously
  • the UTD group is used as the negative control.
  • CS1 CAR-T cells 21G-26G obtained in Example 1 and target cells were co-cultured in X-VIVO15 medium for 48 hours according to the condition of CAR+ cells and target cell effect-to-target ratio of 1:1, Detect target cells by detecting stably expressed luciferase activity in target cells (construct a lentiviral expression vector containing green fluorescent protein GFP and luciferase Luc coding region, then package lentivirus, transduce K562-CS1 cells with lentivirus, Use the GFP signal to sort positive monoclonal cells by flow cytometry, through culture expansion and GFP expression identification, after confirming that they are monoclonal, the cell preparation is completed) the survival ratio (the detection reagent is purchased from Promega, product number: E2520), the results are as follows As shown in Figure 5: CS1 CAR-T cells 21G, 22G, 23G, 26G and the positive control CS1 CAR-T cell LUC90V2 all had good killing effect
  • Example 4 Sustained proliferation of CS1 CAR-T cells
  • Antigen stimulation can activate CAR-T cells to proliferate CAR-T cells, and the continuous activation of T cells will lead to cell exhaustion, and the proliferation ability and effector function of exhausted T cells will decrease.
  • CS1 CAR-T cells 21G The proliferation of CD3+ cells (i.e. the proliferation of T cells, CD3 is a marker to distinguish whether they are T cells) and the proliferation of CAR+ cells after multiple rounds of antigen stimulation experiments of ⁇ 26G, to verify the continuous proliferation of CS1 CAR-T cells .
  • the CAR-positive ratio of CS1 CAR-T cells in each group was adjusted to a level consistent with that of the group of CAR-T cells with the lowest CAR-positive ratio using T cells that were not transduced with CAR.
  • CS1 CAR-T cells in each group were co-cultured with CS1 endogenous positive target cells MM.1S (human multiple myeloma cells) in a 24-well plate according to the effect-to-target ratio of 1:2. , 2ml X-VIVO15 medium per well, repeat 3 wells for each group of cells.
  • CDR1, CDR2, and CDR3 of VH in 22G scFv are shown in SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3, respectively, and CDR1, CDR2, and CDR3 of VL in 22G scFv are shown in SEQ ID NO:4, Shown in SEQ ID NO:5 and SEQ ID NO:6.
  • the 02G scFv selected for the BCMA target (the CDR1, CDR2, and CDR3 of its VH are shown in SEQ ID NO:51, SEQ ID NO:52, and SEQ ID NO:53, respectively, and the CDR1, CDR2, and CDR3 of its VL are shown in Shown in SEQ ID NO:54, SEQ ID NO:55, SEQ ID NO:56, the amino acid sequence of scFv VH is shown in SEQ ID NO:57, and the amino acid sequence of scFv VL is shown in SEQ ID NO:58), for After the 22G scFv was selected for the CS1 target, the construction of the BCMA-CS1 bispecific CAR began.
  • BCMA-CS1 bispecific CAR Considering the changes in the sequence of target BCMA scFv and CS1 scFv, the sequence of VH and VL in each target scFv, the type of linker, and the different structures of tandem and loop, there are many structural designs for BCMA-CS1 bispecific CAR.
  • One option tried four tandem structures and two loop structures as shown in Figure 7, and built them into the second-generation CAR structure.
  • CD8 ⁇ guide chain as signal peptide
  • BCMA-CS1 bispecific scFv as extracellular tumor antigen recognition region
  • hinge region and transmembrane region as CD8 ⁇ structure
  • 4-1BB intracellular co-stimulatory signal
  • CD3 ⁇ T Cell activation signal.
  • Example 1 In addition to replacing the CAR sequence, the method for preparing CS1 CAR-T cells in Example 1 was still used to prepare BCMA-CS1 bispecific CAR-T cells 41A, 41B, 42A, 42B, 43A, 43B (the sequence of the corresponding scFv Respectively such as: SEQ ID NO:45, SEQ ID NO:46, SEQ ID NO:47, SEQ ID NO:48, SEQ ID NO:49, SEQ ID NO:50), the amino acid sequence of the components in the second generation CAR structure and Example 1 is the same.
  • BCMA-CS1 bispecific CAR-T cells 41A, 41B, 42A, 42B, 43A, and 43B were tested for CAR positive rate, among which: UTD cells were used as negative controls, and BCMA CAR-T cells 02G (replacing the CAR sequence, still using Prepared by the method for preparing CS1 CAR-T cells in Example 1) and CS1 CAR-T cells 22G (prepared in Example 1) were used as positive controls, and FITC fluorescently labeled BCMA antigen (manufacturer: ACRO Biosystems, article number: BCA-HF254) and PE fluorescently labeled CS1 antigen (same as Example 2) were stained, and the results are shown in Figure 8: CAR-T cells with four tandem structures (41A, 41B, 42A, 42B) were almost not detected When it comes to CAR-positive cells, both 43A and 43B can clearly detect CAR-positive cells, but most of the positive cells of 43A only recognize BCMA antigens, while the positive cells of 43B
  • Example 6 since only BCMA-CS1 bispecific CAR-T cells 43A and 43B clearly detected CAR-positive cells, 43A, 43B and BCMA CAR-T cells 02G, CS1 CAR-T cells The CAR positive rate of 22G was adjusted to be consistent. Since the positive rate of CAR in other groups was very low, the positive rate of CAR in all groups was not uniformly adjusted to the lowest level. In the end, there were no adjusted cells in each group (41A, 41B, 42A, 42B) Only the same number of T cells can be added in the experiment, but the same number of CAR+T cells cannot be satisfied.
  • the obtained BCMA-CS1 bispecific CAR-T cells 41A, 41B, 42A, 42B, 43A, 43B and target cells were placed in X-VIVO15 medium according to the conditions of different effect-to-target ratios between CAR+ cells and target cells.
  • the survival rate of target cells was detected by detecting the stably expressed luciferase activity in the target cells (same as in Example 3). and the killing ability of target cells expressing exogenous CS1, and UTD cells were used as negative controls, and BCMA CAR-T cells 02G and CS1 CAR-T cells 22G were used as positive controls.
  • SEQ ID NO: 1 GFSLSNYG, which is 22G scFv VH CDR1;
  • SEQ ID NO:2 IGTIGAT, which is 22G scFv VH CDR2;
  • SEQ ID NO: 3 ARGIYGDIYVYAFDI, which is 22G scFv VH CDR3;
  • SEQ ID NO:4 QSVRDNGD, which is 22G scFv VL CDR1;
  • SEQ ID NO:5 DVS, which is 22G scFv VL CDR2;
  • SEQ ID NO:6 AGGYIAGSDRWV, which is 22G scFv VL CDR3;
  • SEQ ID NO: 7 amino acid sequence of humanized 22G scFv VH;
  • SEQ ID NO: 8 amino acid sequence of humanized 22G scFv VL;
  • SEQ ID NO: 9 amino acid sequence of rabbit source 22G scFv VH;
  • SEQ ID NO: 10 amino acid sequence of rabbit source 22G scFv VL;
  • SEQ ID NO:11 amino acid sequence of humanized 22G scFv
  • SEQ ID NO:12 Amino acid sequence of rabbit-derived 22G scFv;
  • SEQ ID NO:13 the amino acid sequence of the hinge region
  • SEQ ID NO:14 the amino acid sequence of the transmembrane region
  • SEQ ID NO:15 the amino acid sequence of the intracellular signal transduction region
  • SEQ ID NO:16 the amino acid sequence of costimulatory signal transduction region
  • SEQ ID NO:17 the amino acid sequence of the leader peptide (i.e. signal peptide);
  • SEQ ID NO: 18 nucleotide sequence encoding 22G scFv amino acid sequence
  • SEQ ID NO: 19 21G scFv amino acid sequence
  • SEQ ID NO:20 23G scFv amino acid sequence
  • SEQ ID NO:21 24G scFv amino acid sequence
  • SEQ ID NO:22 25G scFv amino acid sequence
  • SEQ ID NO:23 26G scFv amino acid sequence
  • SEQ ID NO:24 27G scFv amino acid sequence
  • SEQ ID NO:25 nucleotide sequence encoding 21G scFv amino acid sequence
  • SEQ ID NO:26 nucleotide sequence encoding 23G scFv amino acid sequence
  • SEQ ID NO:27 nucleotide sequence encoding 24G scFv amino acid sequence
  • SEQ ID NO:28 nucleotide sequence encoding 25G scFv amino acid sequence
  • SEQ ID NO:29 nucleotide sequence encoding 26G scFv amino acid sequence
  • SEQ ID NO: 30 nucleotide sequence encoding 27G scFv amino acid sequence
  • SEQ ID NO:31 21G CAR amino acid sequence
  • SEQ ID NO:32 22G CAR amino acid sequence
  • SEQ ID NO:33 23G CAR amino acid sequence
  • SEQ ID NO:34 24G CAR amino acid sequence
  • SEQ ID NO:35 25G CAR amino acid sequence
  • SEQ ID NO:36 26G CAR amino acid sequence
  • SEQ ID NO:37 27G CAR amino acid sequence
  • SEQ ID NO:38 nucleotide sequence encoding 21G CAR amino acid sequence
  • SEQ ID NO:39 nucleotide sequence encoding 22G CAR amino acid sequence
  • SEQ ID NO:40 nucleotide sequence encoding 23G CAR amino acid sequence
  • SEQ ID NO:41 nucleotide sequence encoding 24G CAR amino acid sequence
  • SEQ ID NO:42 Nucleotide sequence encoding 25G CAR amino acid sequence
  • SEQ ID NO:43 Nucleotide sequence encoding 26G CAR amino acid sequence
  • SEQ ID NO:44 Nucleotide sequence encoding 27G CAR amino acid sequence
  • SEQ ID NO:45 bispecific scFv 41A amino acid sequence
  • SEQ ID NO:46 bispecific scFv 41B amino acid sequence
  • SEQ ID NO:47 bispecific scFv 42A amino acid sequence
  • SEQ ID NO:48 bispecific scFv 42B amino acid sequence
  • SEQ ID NO:49 bispecific scFv 43A amino acid sequence
  • SEQ ID NO:50 bispecific scFv 43B amino acid sequence
  • SEQ ID NO:51 GFSLSTYH, which is 02G scFv VH CDR1;
  • SEQ ID NO:52 ISSSGST, which is 02G scFv VH CDR2;
  • SEQ ID NO:53 ARDLDYVIDL, which is 02G scFv VH CDR3;
  • SEQ ID NO:54 PSVYNNY, which is 02G scFv VL CDR1;
  • SEQ ID NO:55 ETS, which is 02G scFv VL CDR2;
  • SEQ ID NO:56 AGTYVSGDRRA, which is 02G scFv VL CDR3;
  • SEQ ID NO:57 VH amino acid sequence of humanized 02G scFv antibody
  • SEQ ID NO:58 VL amino acid sequence of humanized 02G scFv antibody
  • SEQ ID Nos:59-61 the amino acid sequence of the linking region connecting VH and VL;
  • SEQ ID NO:62 BCMA 02G CAR
  • SEQ ID NO:63 LUC90V2 positive control CS1 CAR sequence.

Abstract

La présente invention concerne un récepteur antigénique chimérique ciblant CS1, comprenant un domaine de reconnaissance d'antigène extracellulaire, une région charnière, une région transmembranaire et un domaine intracellulaire, le domaine de reconnaissance d'antigène extracellulaire comprenant un anticorps scFv dirigé contre CS1 ; des séquences d'acides aminés de régions déterminant la complémentarité VH CDR1, CDR2 et CDR3 de l'anticorps scFv comprennent respectivement des séquences d'acides aminés telles que représentées dans SEQ ID NO : 1, SEQ ID NO : 2 et SEQ ID NO : 3 ; et des séquences d'acides aminés de régions déterminant la complémentarité VL CDR1, CDR2 et CDR3 de l'anticorps scFv comprennent respectivement des séquences d'acides aminés telles que représentées dans SEQ ID NO : 4, SEQ ID NO : 5 et SEQ ID No: 6.
PCT/CN2022/143236 2021-12-31 2022-12-29 Récepteur antigénique chimérique ciblant cs1, récepteur antigénique chimérique bispécifique ciblant bcma/cs1 et utilisation associée WO2023125766A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111678338.9 2021-12-31
CN202111678338.9A CN116410331B (zh) 2021-12-31 2021-12-31 靶向cs1的嵌合抗原受体、靶向bcma/cs1的双特异性嵌合抗原受体及其应用

Publications (1)

Publication Number Publication Date
WO2023125766A1 true WO2023125766A1 (fr) 2023-07-06

Family

ID=86998093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/143236 WO2023125766A1 (fr) 2021-12-31 2022-12-29 Récepteur antigénique chimérique ciblant cs1, récepteur antigénique chimérique bispécifique ciblant bcma/cs1 et utilisation associée

Country Status (2)

Country Link
CN (1) CN116410331B (fr)
WO (1) WO2023125766A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107429253A (zh) * 2014-12-05 2017-12-01 希望之城公司 Cs1靶向性嵌合抗原受体修饰的t细胞
WO2020009868A1 (fr) * 2018-07-03 2020-01-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Récepteurs d'antigènes chimériques anti-slamf7
WO2021045975A1 (fr) * 2019-09-04 2021-03-11 Promab Biotechnologies, Inc. Anticorps cs1 et cellules anti-cs1-car-t
CN112566643A (zh) * 2018-06-12 2021-03-26 加利福尼亚大学董事会 用于治疗癌症的单链双特异性嵌合抗原受体
CN112778427A (zh) * 2021-01-29 2021-05-11 武汉思安医疗技术有限公司 双特异性cs1-bcma car-t细胞及其应用

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2711118C2 (ru) * 2012-11-05 2020-01-15 Деленекс Терапьютикс Аг Элементы, связывающиеся с ил-1 бета
ES2777940T3 (es) * 2013-05-03 2020-08-06 Ohio State Innovation Foundation Células efectoras inmunitarias genomanipuladas con un receptor de antígeno químico específico de CS1
CA2947646A1 (fr) * 2014-05-02 2015-11-05 Cellectis Recepteur antigenique chimerique a chaines multiples specifique de cs1
US20190135894A1 (en) * 2015-06-25 2019-05-09 iCell Gene Therapeuticics LLC COMPOUND CHIMERIC ANTIGEN RECEPTOR (cCAR) TARGETING MULTIPLE ANTIGENS, COMPOSITIONS AND METHODS OF USE THEREOF
JP7146632B2 (ja) * 2015-07-21 2022-10-04 ノバルティス アーゲー 免疫細胞の有効性および増大を改善する方法
CN116082505A (zh) * 2016-02-25 2023-05-09 细胞医学公司 Pd-l1的结合成员
CN108276493B (zh) * 2016-12-30 2023-11-14 南京传奇生物科技有限公司 一种嵌合抗原受体及其应用
EP3346001A1 (fr) * 2017-01-06 2018-07-11 TXCell Population de cellules t régulatrices monospécifiques avec cytotoxicité pour cellules b
EP3700933A1 (fr) * 2017-10-25 2020-09-02 Novartis AG Anticorps ciblant cd32b et leurs procédés d'utilisation
WO2019195017A1 (fr) * 2018-04-03 2019-10-10 Promab Biotechnologies, Inc. Lymphocytes t-car anti-bcma
US10640562B2 (en) * 2018-07-17 2020-05-05 Mcmaster University T cell-antigen coupler with various construct optimizations
WO2020025039A1 (fr) * 2018-08-03 2020-02-06 南京驯鹿医疗技术有限公司 Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée
US20200190163A1 (en) * 2018-10-26 2020-06-18 Lijun Wu Humanized bcma-car-t cells
CN109485734B (zh) * 2018-12-30 2020-05-12 广州百暨基因科技有限公司 一种靶向bcma和cd19的双特异性嵌合抗原受体及其应用
WO2020227595A1 (fr) * 2019-05-09 2020-11-12 Atara Biotherapeutics, Inc. Cellules car-t ciblées par clec4
CN112279923B (zh) * 2019-07-22 2023-07-18 南京助天中科科技发展有限公司 一种嵌合抗原受体及其应用
CN112079934B (zh) * 2019-12-17 2021-01-29 合源生物科技(天津)有限公司 一种靶向cd19的嵌合抗原受体及其用途
CN112979820B (zh) * 2019-12-17 2022-06-17 中国医学科学院血液病医院(中国医学科学院血液学研究所) 靶向cd123的嵌合抗原受体及含有靶向cd123嵌合抗原受体的双靶点嵌合抗原受体
CN111171158B (zh) * 2020-01-17 2020-10-30 南京蓝盾生物科技有限公司 同时靶向bcma和cd38的嵌合抗原受体及其应用
CN113278071B (zh) * 2021-05-27 2021-12-21 江苏荃信生物医药股份有限公司 抗人干扰素α受体1单克隆抗体及其应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107429253A (zh) * 2014-12-05 2017-12-01 希望之城公司 Cs1靶向性嵌合抗原受体修饰的t细胞
CN112566643A (zh) * 2018-06-12 2021-03-26 加利福尼亚大学董事会 用于治疗癌症的单链双特异性嵌合抗原受体
WO2020009868A1 (fr) * 2018-07-03 2020-01-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Récepteurs d'antigènes chimériques anti-slamf7
WO2021045975A1 (fr) * 2019-09-04 2021-03-11 Promab Biotechnologies, Inc. Anticorps cs1 et cellules anti-cs1-car-t
CN112778427A (zh) * 2021-01-29 2021-05-11 武汉思安医疗技术有限公司 双特异性cs1-bcma car-t细胞及其应用

Also Published As

Publication number Publication date
CN116410331B (zh) 2024-01-30
CN116410331A (zh) 2023-07-11

Similar Documents

Publication Publication Date Title
JP7047172B2 (ja) 融合タンパク質を用いたtcrの再プログラミングのための組成物及び方法
JP7061235B2 (ja) 抗cld18a2ナノ抗体及びその応用
US11242376B2 (en) Compositions and methods for TCR reprogramming using fusion proteins
AU2016211438C1 (en) Chimeric antigen receptors, compositions, and methods
JP2023052446A (ja) 融合タンパク質を用いてt細胞受容体をリプログラミングするための組成物及び方法
WO2017063162A1 (fr) Anticorps anti-ox40 et son application
CN115135674A (zh) 树突细胞激活性嵌合抗原受体和其用途
CN115052902A (zh) 淋巴细胞-抗原提呈细胞共刺激因子及其应用
JP7459046B2 (ja) Steap1に対するキメラ受容体及びその使用方法
EP4023678A1 (fr) Récepteur d'antigène chimérique et cellule effectrice immunitaire exprimant un récepteur d'antigène chimérique
CN113039209A (zh) 用于使用融合蛋白进行tcr重编程的组合物和方法
CN115850476B (zh) 一种cll1抗体及其应用
CN115850505A (zh) 一种靶向cll1的嵌合抗原受体及其应用
WO2023125766A1 (fr) Récepteur antigénique chimérique ciblant cs1, récepteur antigénique chimérique bispécifique ciblant bcma/cs1 et utilisation associée
WO2023088359A1 (fr) Récepteur antigénique chimérique ciblant bcma et son utilisation
WO2023226921A1 (fr) Récepteur antigénique chimérique bispécifique ciblant bcma-cd19 et son utilisation
WO2024032247A1 (fr) Anticorps anti-cll1 et son utilisation
KR102393776B1 (ko) Cd22에 특이적인 인간화 항체 및 이를 이용한 키메라 항원 수용체
US20240123068A1 (en) Cd19 binders, car-t constructs comprising the same, and methods of using the same
US11802159B2 (en) Humanized anti-GDNF family alpha-receptor 4 (GRF-alpha-4) antibodies and chimeric antigen receptors (CARs)
US20240052031A1 (en) Cea6 binding molecules and uses thereof
US20240050569A1 (en) Mesothelin binding molecules and uses thereof
US20240076400A1 (en) Epcam binding molecules and uses thereof
KR20240034205A (ko) 항EGFRviii 항체, 폴리펩타이드, 상기 폴리펩타이드를 발현하는 세포, 상기 세포를 포함하는 의약 조성물, 상기 세포의 제조 방법 및 상기 폴리펩타이드를 코딩하는 염기서열을 포함하는 폴리뉴클레오티드 또는 벡터
WO2023010068A2 (fr) Cellules génétiquement modifiées sécrétant un anticorps multispécifique

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: 22915016

Country of ref document: EP

Kind code of ref document: A1