WO2024051641A1 - Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof - Google Patents

Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof Download PDF

Info

Publication number
WO2024051641A1
WO2024051641A1 PCT/CN2023/116708 CN2023116708W WO2024051641A1 WO 2024051641 A1 WO2024051641 A1 WO 2024051641A1 CN 2023116708 W CN2023116708 W CN 2023116708W WO 2024051641 A1 WO2024051641 A1 WO 2024051641A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
cells
tumor
tumor cell
cell
Prior art date
Application number
PCT/CN2023/116708
Other languages
French (fr)
Chinese (zh)
Inventor
高明明
沈浩
靳利远
张伟伟
Original Assignee
复星凯特生物科技有限公司
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 复星凯特生物科技有限公司 filed Critical 复星凯特生物科技有限公司
Publication of WO2024051641A1 publication Critical patent/WO2024051641A1/en

Links

Classifications

    • 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
    • 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
    • A61K39/001103Receptors for growth factors
    • A61K39/001104Epidermal growth factor receptors [EGFR]
    • 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
    • A61K39/001103Receptors for growth factors
    • A61K39/00111Hepatocyte growth factor receptor [HGFR or c-met]
    • 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
    • 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
    • C07K16/28Immunoglobulins [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
    • C07K16/40Immunoglobulins [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
    • 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
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • 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

Definitions

  • the invention belongs to the field of biotechnology.
  • the present invention relates to anti-EGFR and cMet bispecific chimeric antigen receptors and their uses.
  • CAR chimeric antigen receptors
  • CAR molecules include three main structural components, namely the extracellular antigen-binding functional domain; the transmembrane domain to transmit antigen recognition signals; and the intracellular signaling domain.
  • the first generation of CAR only contains CD3 ⁇ activation signal. Due to the lack of costimulatory signal functional domain, it cannot fully activate CAR T cells, shows limited cytotoxicity, and cannot effectively expand or persist in the body.
  • the second-generation CAR also introduced the costimulatory functional domain of CD28 or 4-1BB, which enhanced the cytotoxicity of CAR T and improved its persistence.
  • CD28 or 4-1BB costimulatory functional domain of CD28 or 4-1BB
  • CAR T Although it is reported to have stronger persistence, it may induce excessive release of cytokines. At present, its clinical efficacy has not been verified. In order to expand the efficacy of CAR T and apply it to a wider range of malignant tumors, especially solid tumors, innovative engineering construction of CAR T is needed.
  • CRS cytokine release syndrome
  • neurological toxicity on-target off-tumor toxicity
  • tumor lysis syndrome a chronic respiratory disease characterized by CAR T cell therapy.
  • CRS cytokine release syndrome
  • neurological toxicity on-target off-tumor toxicity
  • tumor lysis syndrome a chronic respiratory disease characterized by CRS pulmonary disease.
  • CRS cytokine release syndrome
  • Tumor-related antigens are highly expressed in tumor cells, they are also expressed at non-specific low levels in normal tissues or cells, such as CD19, Her2, ROR1, Muc1, etc.
  • Target toxicity means that CAR T cells are activated by recognizing antigens expressed by normal tissues and cause damage to normal tissues.
  • CAR T treats CD19-positive malignant lymphoma because there are CD19-expressing B cell progenitor cells in the bone marrow. cells, which are eliminated together with tumor cells, resulting in B cell dysplasia and reduced hemoglobin. Clinically, immunoglobulin is infused to replace the antibody function produced by B cells.
  • CAR T cells targeting carbonic anhydrase IX to treat renal cancer several patients developed liver enzyme abnormalities. These adverse reactions were attributed to CAR T cell infiltration and action on carbonic anhydrase-expressing cells. Bile duct epithelium. A patient with colorectal cancer developed fatal lung damage after receiving an infusion of Her2-targeting CAR T cells because lung epithelial cells also expressed low levels of Her2.
  • the purpose of the present invention is to provide a new type of chimeric antigen receptor T cell that can recognize multiple antigens at the same time, enhance targeting specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor (on target/off-tumor) off tumor) toxicity and improve the safety of cell therapy.
  • a first aspect of the present invention provides an engineered immune cell, the engineered immune cell expresses a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the third Two CARs target a second tumor cell marker, and the first tumor cell marker is selected from the following group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof; the second tumor cell The marker is selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof.
  • the immune cell contains a first CAR and a second CAR.
  • the immune cells are NK cells, macrophages or T cells, preferably T cells.
  • the first CAR and the second CAR are located on the cell membrane of the immune cell.
  • the first CAR and the second CAR contain antigen-binding domains targeting tumor cell markers.
  • the antigen-binding domain is an antibody or antigen-binding fragment.
  • the antigen-binding fragment is Fab or scFv or single domain antibody sdFv.
  • the structure of the first CAR is shown in Formula I: L1-S1-H1-TM1-C1-CD3 ⁇ (I)
  • the "-" is a connecting peptide or peptide bond
  • L1 is none or the first signal peptide sequence
  • S1 is an antigen-binding domain targeting a first tumor cell marker selected from the group consisting of: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof;
  • H1 is none or the first hinge area
  • TM1 is the first transmembrane domain
  • C1 is no or the first costimulatory signal molecule
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇ .
  • the antigen-binding domain targeting the first tumor cell marker includes an antibody single-chain variable region sequence targeting the tumor cell marker.
  • the structure of the single-chain variable region sequence of the antibody targeting the first tumor cell marker is shown in Formula A1 or A2: V H1 -V L1 (A1); or V L1 -V H1 (A2);
  • V L1 is the light chain variable region of the anti-first tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-first tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or Peptide bonds.
  • V L1 and V H1 are connected through a flexible joint.
  • the flexible linker is 1-5 (preferably, 2-4) consecutive sequences represented by GGGGS.
  • amino acid sequence of the flexible linker is shown in positions 120-134 of SEQ ID NO.: 1.
  • amino acid sequence of V L1 is shown at positions 135-247 of SEQ ID NO.: 1
  • amino acid sequence of V H1 is shown at positions 1-119 of SEQ ID NO.: 1.
  • the single-chain variable region sequence of the antibody targeting the first tumor cell marker is shown in SEQ ID NO.: 1.
  • amino acid sequence of the first CAR is as shown in any one of SEQ ID NO.: 2-4.
  • the structure of the second CAR is shown in Formula II: L2-S2-H2-TM2-C2-Z2 (II)
  • the "-" is a connecting peptide or peptide bond
  • L2 is none or the second signal peptide sequence
  • S2 is an antigen-binding domain targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof;
  • H2 is no or second hinge region
  • TM2 is the second transmembrane domain
  • C2 is the second costimulatory signal molecule
  • Z2 is a cytoplasmic signaling sequence that is absent or derived from CD3 ⁇ .
  • the antigen-binding domain targeting the second tumor cell marker includes an antibody single-chain variable region sequence targeting the tumor cell marker.
  • the structure of the single-chain variable region sequence of the antibody targeting the second tumor cell marker is shown in Formula A3 or A4: V H2 -V L2 (A3); or V L2 -V H2 (A4);
  • V L2 is the light chain variable region of the anti-second tumor cell marker antibody
  • V H2 is the heavy chain variable region of the anti-second tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or Peptide bonds.
  • V L2 and V H2 are connected through a flexible joint.
  • the flexible linker is 1-5 (preferably, 2-4) consecutive sequences represented by GGGGS.
  • amino acid sequence of the flexible linker is as shown at positions 120-134 in SEQ ID NO.: 5.
  • amino acid sequence of the flexible linker is as shown at positions 120-134 in SEQ ID NO.: 6.
  • amino acid sequence of V L2 is shown in positions 135-241 of SEQ ID NO.:5
  • amino acid sequence of V H2 is shown in positions 1-119 of SEQ ID NO.:5.
  • amino acid sequence of V L2 is shown at positions 135-241 of SEQ ID NO.:6, and the amino acid sequence of V H2 is shown at positions 1-119 of SEQ ID NO.:6.
  • the single-chain variable region sequence of the antibody targeting the second tumor cell marker is shown in SEQ ID NO.: 5 or 6.
  • amino acid sequence of the second CAR is as shown in any one of SEQ ID NO.: 7-14.
  • the single-chain variable region of the antibody targeting the first or second tumor cell marker The sequences are murine, human, human and murine chimeric, or fully humanized single-chain antibody variable region fragments.
  • each of L1 and L2 is independently a signal peptide selected from the following group of proteins: CD8a, CD8, CD28, GM-CSF, CD4, CD137, or a combination thereof.
  • the L1 is a signal peptide derived from GM-CSF.
  • the L2 is a signal peptide derived from CD8a.
  • amino acid sequences of L1 and L2 are independently as shown in SEQ ID NO.: 15 and 16.
  • the H1 and H2 are each independently a hinge region of a protein selected from the following group: CD8, Ig (immunoglobulin) hinge, or a combination thereof.
  • H1 and H2 are each independently a hinge region derived from CD8.
  • amino acid sequences of H1 and H2 are independently as shown in SEQ ID NO.: 17.
  • the TM1 and TM2 are each independently a transmembrane region of a protein selected from the following group: CD8, CD28, CD8a, CD33, CD37, CD8 ⁇ , CD5, CD16, ICOS, CD9, CD22, CD134 , CD137, CD154, CD19, CD45, CD4, CD3 ⁇ , or combinations thereof.
  • the TM1 and TM2 are transmembrane regions derived from CD8.
  • the TM2 is a transmembrane region derived from CD28.
  • amino acid sequences of TM1 and TM2 are shown in SEQ ID NO.: 18.
  • amino acid sequence of TM2 is shown in SEQ ID NO.: 19.
  • the C1 and C2 are each independently a costimulatory signal molecule selected from the following group of proteins: CD28, 4-1BB (CD137), CD30, CD40, CD70, CD134, LIGHT, DAP10, CDS , ICAM-1, OX40, or combinations thereof.
  • the C1 is none; or a costimulatory signal molecule derived from CD28 and/or 4-1BB.
  • the C2 is a costimulatory signal molecule derived from CD28 and/or 4-1BB.
  • amino acid sequences of C1 and C2 are independently as shown in SEQ ID NO.: 20.
  • amino acid sequences of C1 and C2 are independently as shown in SEQ ID NO.: 21.
  • amino acid sequence of CD3 ⁇ is shown in SEQ ID NO.: 22.
  • a second aspect of the present invention provides a method for preparing the engineered immune cells described in the first aspect of the present invention, comprising the following steps:
  • (B) Modify the immune cells so that the immune cells express a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the second CAR targets a first tumor cell marker. Two tumor cell markers, thereby obtaining the engineered immune cells described in the first aspect of the present invention.
  • step (A) also includes isolating and/or activating immune cells to be modified.
  • step (B) includes (B1) introducing a first expression cassette expressing a first CAR targeting the first tumor cell marker into the immune cell; and (B2) introducing A second expression cassette expressing a second CAR targeting a second tumor cell marker is introduced into the immune cell; wherein step (B1) can be performed before, after, simultaneously, or alternately with step (B2).
  • step (B) the first expression cassette and/or the second expression cassette is introduced into the nucleus of the immune cell.
  • step (B) when the immune cells to be modified in step (A) already express the first CAR and the second CAR, step (B) can be omitted.
  • the immune cells are NK cells, macrophages or T cells.
  • the first expression cassette contains a nucleic acid sequence encoding the first CAR.
  • the second expression cassette contains a nucleic acid sequence encoding the second CAR.
  • 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.
  • first expression cassette and the second expression cassette are located in different vectors.
  • the vector is a viral vector.
  • the vector is selected from the following group: DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, other gene transfer systems, or combinations thereof.
  • the vector is a retroviral vector.
  • the method further includes the step of testing the function and effectiveness of the obtained engineered immune cells.
  • a third aspect of the present invention provides a preparation, which contains the engineered immune cells described in the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
  • the preparation is a liquid preparation.
  • the dosage form of the preparation includes 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 chemotherapy 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 for preparing drugs or preparations that selectively kill tumors.
  • the tumors include tumors that highly express tumor cell markers (such as EGFR, cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, EpCAM).
  • tumor cell markers such as EGFR, cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, EpCAM.
  • the tumors include tumors that simultaneously express tumor cell markers (such as EGFR, cMet, Her2, and HER3).
  • tumor cell markers such as EGFR, cMet, Her2, and HER3.
  • the tumor includes a tumor that expresses both EGFR and cMet.
  • the tumor is selected from the group consisting of hematological tumors, solid tumors, or combinations thereof.
  • the tumor is a solid tumor.
  • the blood tumor is selected from the following group: acute myelogenous leukemia, acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodysplastic syndrome, or combinations thereof.
  • acute myelogenous leukemia acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodys
  • 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 lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, and chondrosarcoma.
  • the fifth aspect of the present invention provides a kit for selectively killing tumors.
  • the kit contains a container, and located in the container:
  • first and second nucleic acid sequences are independent or connected.
  • first and second nucleic acid sequences are located in the same or different containers.
  • 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 cells described in the first aspect of the present invention or the preparation described in the third aspect of the present invention is administered to the 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, which includes administering a safe and effective amount of the engineered immune cells described in the first aspect of the present invention or the preparation described in 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 tumors to the subject in need of treatment.
  • the other drugs include CAR-T drugs.
  • the disease is cancer or tumor.
  • the tumors include tumors that highly express tumor cell markers (such as EGFR, cMet, HER2, HER3, MUCl, ROR1, PD-L1, CD47).
  • tumor cell markers such as EGFR, cMet, HER2, HER3, MUCl, ROR1, PD-L1, CD47.
  • the tumors include tumors that simultaneously express tumor cell markers (such as EGFR, cMet, EpCAM, HER2, HER3).
  • tumor cell markers such as EGFR, cMet, EpCAM, HER2, HER3.
  • the tumor includes a tumor that expresses both EGFR and cMet.
  • the tumor is selected from the following group: hematological tumors, solid tumors, or combinations thereof.
  • the tumor is a solid tumor.
  • the blood tumor is selected from the following group: acute myelogenous leukemia, acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodysplastic syndrome, or combinations thereof.
  • acute myelogenous leukemia acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodys
  • 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 lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, and 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, nasopharyngeal cancer, thymic cancer, or combinations thereof.
  • An eighth aspect of the present invention provides a fusion protein, the fusion protein comprising a first CAR targeting a first tumor cell marker and a second CAR targeting a second tumor cell marker, the first tumor cell marker
  • the material is selected from the following group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof
  • the second tumor cell marker is selected from the following group: EGFR, EpCAM, Her2, Her3, or a combination thereof .
  • first CAR and the second CAR are connected through a connecting peptide.
  • the connecting peptide includes a self-cleaving protein.
  • the self-cleaving protein is selected from the following group: 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: L1-S1-H1-TM1-C1-CD3 ⁇ -Z3-L2-S2-H2-TM2-C2-Z2 (III)
  • Each "-" is independently a connecting peptide or peptide bond
  • L1 is none or the first signal peptide sequence
  • L2 is none or the second signal peptide sequence
  • S1 is an antigen-binding domain targeting a first tumor cell marker selected from the group consisting of: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof;
  • S2 is an antigen-binding domain targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof;
  • H1 is none or the first hinge area
  • H2 is no or second hinge region
  • TM1 is the first transmembrane domain
  • TM2 is the second transmembrane domain
  • C1 is no or the first costimulatory signal molecule
  • C2 is the second costimulatory signal molecule
  • CD3 ⁇ is a cytoplasmic signaling sequence derived from CD3 ⁇
  • Z2 is a cytoplasmic signaling sequence that is absent or derived from CD3 ⁇ ;
  • Z3 is the connecting peptide.
  • amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
  • the ninth aspect of the present invention provides a polynucleotide encoding the fusion protein according to the eighth aspect of the present invention.
  • polynucleotide is selected from the following group:
  • the 5' end and/or 3' end of the polynucleotide shown in (b) is truncated or 1-60 (preferably 1-30, more preferably 1-10) nucleotides are added polynucleotide;
  • polynucleotide sequence is as shown in any one of SEQ ID NO.: 27-30.
  • a tenth aspect of the present invention provides a vector, which includes the polynucleotide described in the ninth aspect of the present invention.
  • the vector includes DNA and RNA.
  • the vector is selected from the following group: plasmids, viral vectors, transposons, or combinations thereof.
  • the vector includes DNA virus and retroviral vector.
  • the vector is selected from the following group: lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated virus vectors, or combinations thereof.
  • the vector is a retroviral vector.
  • the vector contains one or more promoters operably linked to the nucleic acid sequence, enhancer, intron, transcription termination signal, polyadenylation sequence, origin of replication , selectable markers, nucleic acid restriction sites, and/or homologous recombination site ligation.
  • the vector is a vector containing or inserted with the polynucleotide described in 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 And logic BiCAR targeting EGFR and cMet.
  • the composition includes two independent CAR molecules, the CAR1 molecule is an anti-cMet scFv tandem CD3 ⁇ activation signal domain, and the CAR2 molecule is an anti-EGFR scFv tandem CD28 or 4-1BB costimulatory signal domain;
  • Figure 1B shows " and "Schematic diagram of the molecular construction of logical BiCAR expression, CAR1 molecules and CAR2 molecules are connected through P2A;
  • Figure 1C shows the construction diagram of the second generation fully functional CAR targeting cMet, anti-cMET scFv tandem CD28 or 4-1BB costimulatory signal domain and CD3 ⁇ activation signaling domain.
  • Figure 2 shows the flow cytometry analysis of CAR transduction efficiency of Mock T (2A), O-28z (2B) CAR and "And" logic BiCAR T (2C) cells.
  • Figure 3 shows that Mock T, O-28z CAR and BiCAR T cells prepared from peripheral blood mononuclear cells (PBMC) of different healthy donors #1 (3A) and #2 (3B) have different effects on targets in vitro. Compared with the lytic toxicity to target cells after incubation with NCI-H1975 human lung adenocarcinoma cell line co-expressing EGFR/cMet for 24 hours.
  • PBMC peripheral blood mononuclear cells
  • Figure 4 shows that Mock T, O-28z CAR and BiCAR T cells prepared from peripheral blood mononuclear cells (PBMC) of different healthy donors #1 (4A) and #2 (4B) were compared with each other at different effect-target ratios.
  • PBMC peripheral blood mononuclear cells
  • Figure 5 shows the lytic toxicity of Mock T and BiCAR-T cells to target cells after incubation for 24 hours with the human gastric cancer cell line SNU5 cells co-expressing EGFR/cMET under different effective-target ratio conditions.
  • Figure 6 shows the comparison of Mock T and BiCAR-T cells under different effective target ratio conditions with control 293T-cells, 293T stable transfection strain expressing cMET alone, 293T stably transducing strain expressing EGFR alone and 293T co-expressing EGFR/cMet. Lytic toxicity of the stably transformed strains to target cells after co-incubation for 24 hours.
  • FIG 7 shows the line graph of tumor growth size in tumor-bearing immunodeficient mice after tail vein injection of CAR T cells.
  • the results show that O-28z CAR and BiCAR T cells significantly killed or even eliminated tumor cells in the mice, and were small. No abnormalities were observed in the rats.
  • a first CAR targeting a first tumor cell marker such as cMet, Her2, Her3, Muc1, ROR1, PD-L1, CD47
  • a second targeting tumor cell marker such as EGFR, EpCAM, Her2, Her3
  • the immune cells of the second CAR of tumor cell markers can be fully activated, enhance targeting specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor toxicity, improving cell therapy. security.
  • the term "about” when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value.
  • the expression “about 100” includes all values between 99 and 101 and between (eg, 99.1, 99.2, 99.3, 99.4, etc.).
  • the term “contains” or “includes” can be open, semi-closed and closed. In other words, the term also includes “consisting essentially of,” or “consisting of.”
  • a “chimeric antigen receptor (CAR)” is a fusion protein comprising an extracellular domain capable of binding an antigen, a transmembrane domain derived from a different polypeptide, and at least one cellular domain. inner domain.
  • CAR Chimeric antigen receptor
  • CIR chimeric immune receptor
  • the "extracellular domain capable of binding antigen” refers to any oligopeptide or polypeptide capable of binding a certain antigen.
  • “Intracellular domain” refers to any oligopeptide or polypeptide known as a domain that transmits signals to activate or inhibit biological processes within a cell.
  • domain refers to a region of a polypeptide that is independent of other regions and folds into a specific structure.
  • administering refers to the application of an exogenous drug, therapeutic agent, diagnostic agent or composition to an animal, human, subject, cell, tissue, organ or biological fluid.
  • administering may refer to therapeutic, pharmacokinetic, diagnostic, research and experimental methods.
  • Treatment of cells includes contact of reagents with cells, contact of reagents with fluids, and contact of fluids with cells.
  • administering and “treatment” also mean in vitro and ex vivo treatment of cells by a reagent, a diagnostic, a binding composition or by another cell.
  • Treatment when applied to humans, animals or research subjects, means therapeutic treatment, prophylactic or prophylactic measures, research and diagnostics.
  • treatment refers to the administration of an internal or external therapeutic agent, including any CAR of the invention and compositions thereof, to a patient with one or more symptoms of a disease for which the therapeutic agent is known to be effective. These symptoms are therapeutic.
  • a therapeutic agent is administered to a patient in an amount effective to alleviate one or more symptoms of the disease (a therapeutically effective amount).
  • the terms “optionally” or “optionally” mean that the subsequently described event or circumstance may occur but does not necessarily occur.
  • “optionally including 1-3 antibody heavy chain variable regions” means that the antibody heavy chain variable regions of a specific sequence may be present but are not required to be present, and may be 1, 2 or 3.
  • Sequence identity means the degree of identity between two nucleic acids or two amino acid sequences when optimally aligned and compared with appropriate substitutions, insertions, deletions and other mutations.
  • 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%.
  • Epidermal factor growth receptor belongs to the tyrosine kinase family, which includes three other members: ERBB2/HER2, ERBB3/HER3 and ERBB4/HER4. It is anchored in the cytoplasmic membrane and includes an extracellular ligand-binding functional domain, a hydrophobic transmembrane region and an intracellular tyrosine kinase domain. After ligands of EGFR (such as EGF, TGF-a, etc.) bind to it, they will form homodimers or heterodimers with other family members, leading to autophosphorylation of tyrosine residues, thereby activating multiple A downstream signaling pathway that regulates cell proliferation, survival and apoptosis.
  • the EGFR signaling network plays an important role in the maintenance and growth of epithelial tissue, and its abnormal activation and activity are related to the occurrence, progression and poor prognosis of various cancers.
  • EGFR is overexpressed in a variety of tumors, including 25-77% of colorectal cancers, 80-100% of head and neck cancers, 40-80% of non-small cell lung cancer (NSCLC), 50-90% of renal cancers, 30- 50% of pancreatic cancers, 14-91% of breast cancers, 40-63% of glioblastomas, 35-70% of ovarian cancers, etc.
  • NSCLC non-small cell lung cancer
  • Gene mutations and overexpression of EGFR abnormally activate downstream pathways and are closely related to carcinogenesis and cancer progression.
  • TKIs tyrosine kinase inhibitors
  • monoclonal antibodies cetuximab and panitumumab
  • EGFR is also widely expressed in normal human organs and tissues, including skin, lungs, liver, kidneys, etc.
  • the real-world adverse reactions of approved monoclonal antibody drugs and the clinical on-target toxicity of CAR T cell therapy drugs developed for EGFR are mainly manifested as skin toxicity.
  • EGFR proto-oncogene driver mutations are an important factor in NSCLC carcinogenesis and disease progression. They are detected in 50-60% of Asian patients and are clinically used as biomarkers for prognosis and disease prediction.
  • EGFR-TKI drugs (gefitinib, erlotinib, etc.) have good efficacy in patients with advanced NSCLC with EGFR-sensitive mutations. However, after a median treatment time of 6-12 months, most patients have no response to EGFR-TKI. TKI resistance develops. The resistance mechanism mainly includes acquired mutations of EGFR. About 50% of drug-resistant patients have the T790M mutation.
  • the resistance to third-generation TKI drugs such as osimertinib is due to the C797S mutation; and the activation of the alternative pathway cMet and Her2 signaling pathways. About 30% of patients treated with osimertinib will develop acquired cMet gene amplification, thus avoiding the target EGFR, leading to TKI resistance.
  • NSCLC is a typical case of multiple driver gene mutations. Genomic instability, tumor heterogeneity, and primary or acquired drug resistance generated during disease progression have created bottlenecks in the application of targeted therapy for advanced NSCLC. Therefore, multi-target Targeted therapy will have opportunity to achieve better clinical outcomes.
  • cMet Mesenchymal epidermal transformation factor
  • HGF hepatocyte factor
  • cMet is a type of receptor tyrosine kinase. After binding to the ligand hepatocyte factor (HGF), it leads to dimerization and phosphorylation of the receptor, activating a variety of different cell signaling pathways. Involved in cell proliferation, movement, migration, invasion and angiogenesis.
  • cMet is normally expressed in a variety of human tissues and organs, including liver, esophagus, stomach, colorectum, skin, ovary and endometrium. cMet is very important for controlling cell homeostasis under normal physiological conditions. When cMet is overactivated, it will initiate the transformation of normal cells into tumor cells, resulting in epithelial-mesenchymal transition. Gene mutation, gene amplification or overexpression of cMet can lead to abnormal activation of its signaling pathway and promote tumor progression.
  • cMet is overexpressed in a variety of cancers, including lung, breast, ovarian, renal, colorectal, thyroid, liver, and gastric cancers. Overexpression of cMet is directly related to poor survival. In some cancers, such as lung cancer, colorectal cancer, ovarian cancer, etc., cMet and/or its ligand HGF are used as clinical prognostic indicators. Among cancers with abnormal cMet genes, gastric cancer and colorectal cancer are typical. In gastric cancer, about 10-20% of cMet gene copy number amplifications and about 25% of cMet gene mutations; in colorectal cancer patients, the cMet mutation rate is about 15%. cMet is overexpressed in 25%-75% of NSCLC. In EGFR-TKI-resistant NSCLC, 15%-30% have acquired amplification of the cMet gene, and cMet is also used as a detection marker for TKI resistance in NSCLC patients with EGFR mutations.
  • Targeted drugs for cMet all inhibit the HGF/cMet signaling pathway.
  • Those in clinical trials mainly include receptor enzyme inhibitors (such as Tivantinib, Capmatinib, etc.), monoclonal antibodies (such as Onartuzumab), and antibody drug conjugates ( For example Telisotuzumab vedotin).
  • receptor enzyme inhibitors such as Tivantinib, Capmatinib, etc.
  • monoclonal antibodies such as Onartuzumab
  • antibody drug conjugates For example Telisotuzumab vedotin. Due to cMet mutations in many types of cancer leading to its overexpression, acquired gene amplification caused by EGFR-TKI resistance in NSCLC, and resistance in the development of cMet inhibitors, multiple targets for specific indications and patient groups Developing or combining treatments will have the opportunity to improve clinical efficacy.
  • the antigen-binding domain of the chimeric antigen receptor CAR specifically binds to tumor cell markers, such as EGFR, cMet, Her2, Her3, Mucl, ROR1, or PD-L1, etc.
  • the antigen-binding domain of CAR is a single-chain variable fragment (scFv) formed by connecting the heavy chain and light chain of a monoclonal antibody through flexible linkers of different lengths. svFv sequences are usually derived from mouse, humanized or fully human monoclonal antibodies.
  • the antigen-binding domain also includes smaller, alpaca-derived heavy chain Nanobodies due to the lack of light chains. (VH/nanobody).
  • the affinity of scFv or VH to the target antigen is crucial for regulating CAR T cell function, but too high affinity may cause overactivation of CAR T cells and lead to cell death.
  • affinity the density and epitope of the target antigen are also important factors affecting antigen recognition and efficacy.
  • the high expression of antigen-independent scFv and CAR molecules on the cell membrane will induce the aggregation of CAR molecules, resulting in antigen-independent signal transduction (tonic signaling) and off-target activation, which may ultimately lead to early exhaustion of CAR T cells. .
  • the CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain naturally associated with one of the domains in the CAR is used.
  • transmembrane domains may 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, thus minimizing interaction with the receptor complex. Interactions with other members.
  • the transmembrane domain may be derived from natural or synthetic sources. In natural sources, this 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 CD8 or CD28.
  • the intracellular domain or additional intracellular signaling domain of the CAR of the 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 a cell's specialized function.
  • the effector function of a T cell may be cytolytic activity or auxiliary activity including cytokine secretion.
  • intracellular signaling domain therefore refers to the portion of a protein that transduces effector function signals and directs the cell to perform specialized functions. Although typically the entire intracellular signaling domain can be used, in many instances it is not necessary to use the entire chain.
  • intracellular signaling domain generally refers to any truncated portion of an intracellular signaling domain that is sufficient to transduce an effector function signal.
  • intracellular signaling domains for use in CARs of the present invention include cytoplasmic sequences of T cell receptors (TCRs) and co-receptors that act cooperatively to initiate signal transduction upon antigen receptor binding, as well as these sequences any derivative or variant and any synthetic sequence having the same functional capabilities.
  • TCRs T cell receptors
  • co-receptors that act cooperatively to initiate signal transduction upon antigen receptor binding
  • the cytoplasmic domain of the CAR can be designed to include CD3 ⁇ signaling itself domain, or may be combined with any other desired cytoplasmic domain(s) useful in the context of the CAR of the invention.
  • the cytoplasmic domain of a CAR may include a CD3 zeta chain portion and a costimulatory signaling region.
  • the costimulatory signaling region refers to the portion of the CAR that includes the intracellular domain of the costimulatory molecule.
  • Costimulatory molecules are cell surface molecules required for an effective lymphocyte response to antigen, rather than antigen receptors or their ligands.
  • 4-1BB CD137
  • CD28 etc. are included.
  • cytoplasmic signaling sequences within the cytoplasmic signaling 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 linkage.
  • Glycine-serine doublets provide particularly suitable linkers.
  • the cytoplasmic domain in the CAR of the invention is designed to include 4-1BB, and/or the signaling domain of CD28 (costimulatory molecule) and the signaling domain of CD3 ⁇ .
  • Chimeric antigen receptors are composed of an extracellular antigen recognition region, usually scFv (single-chain variable fragment), a transmembrane region, and an intracellular costimulatory signal region.
  • the design of CARs has gone through the following process: the first-generation CAR has only one intracellular signaling component, CD3 ⁇ or Fc ⁇ RI molecule. Since there is only one activation domain in the cell, it can only cause short-term T cell proliferation and less cytokine secretion. , but cannot provide long-term T cell proliferation signals and sustained anti-tumor effects in vivo, so it has not achieved good clinical efficacy.
  • the second-generation CARs introduce a co-stimulatory molecule, such as CD28, 4-1BB, OX40, and ICOS, based on the original structure. Compared with the first-generation CARs, their functions are greatly improved, further enhancing the persistence of CAR-T cells and their ability to target tumor cells. of lethality. On the basis of second-generation CARs, some new immune costimulatory molecules such as CD27 and CD134 are connected in series to develop into third- and fourth-generation CARs.
  • a co-stimulatory molecule such as CD28, 4-1BB, OX40, and ICOS
  • the extracellular segment of CARs can recognize a specific antigen and then transduce the signal through the intracellular domain, causing cell activation and proliferation, cytolytic toxicity and secretion of cytokines, thereby eliminating target cells.
  • the patient's autologous cells or allogeneic donors
  • CAR immune cells or allogeneic donors
  • the probability of developing 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 has been unachievable by any previous treatment method, triggering an upsurge in clinical research in 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).
  • cell The endodomain includes costimulatory signaling regions and/or zeta chain portions.
  • a costimulatory signaling domain refers to the portion of the intracellular domain that includes costimulatory molecules. Costimulatory molecules are cell surface molecules that are required for effective lymphocyte response to antigen, rather than antigen receptors or their ligands.
  • Linkers can be incorporated between the extracellular and transmembrane domains of the CAR, or between the cytoplasmic and transmembrane domains of the CAR.
  • the term "linker” generally refers to any oligopeptide or polypeptide that serves to connect a 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 expressed in T cells, the CAR of the present invention is capable of antigen recognition based on antigen-binding specificity. When it binds to its cognate antigen, it affects tumor cells, causing the tumor cells to fail to grow, be driven to death, or otherwise affected, and cause the patient's tumor burden to shrink or be eliminated.
  • the antigen binding domain is preferably fused to an intracellular domain from one or more of the costimulatory molecules and/or zeta chains.
  • the antigen binding domain is fused to an intracellular domain in combination with a 4-1BB signaling domain and/or a CD3 ⁇ signaling domain.
  • antigen-binding domain and “single-chain antibody fragment” all refer to a Fab fragment, a Fab' fragment, an F(ab')2 fragment, or a single Fv fragment with antigen-binding activity.
  • Fv antibodies are the smallest antibody fragments that contain an antibody heavy chain variable region, a light chain variable region, but no constant region, and have all antigen-binding sites. Typically, Fv antibodies also contain a polypeptide linker between the VH and VL domains and are capable of forming 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 a complete antibody.
  • Single chain antibodies are preferably one amino acid chain sequence encoded by one nucleotide chain.
  • the scFv includes specific recognition of tumor cell markers with high expression (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin18.2, Mucin 1 (MUC1 ), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138), preferably single chain antibodies.
  • the antigen-binding portion of the CAR of the present invention targets the first tumor cell marker and the second tumor cell marker.
  • the antigen-binding portion of the CAR of the present invention is a first scFv targeting cMet and a second scFv targeting EGFR.
  • the structure of the first scFv is shown in Formula A1 or A2: V H1 -V L1 (A1); or V L1 -V H1 (A2);
  • V L1 is the light chain variable region of the anti-first tumor cell marker antibody
  • V H1 is the heavy chain variable region of the anti-first tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or Peptide bonds.
  • amino acid sequence of V L1 is such as positions 135-247 in SEQ ID NO.:1 is shown, and the amino acid sequence of V H1 is shown in positions 1-119 of SEQ ID NO.: 1.
  • the structure of the second scFv is shown in formula A3 or A4: V H2 -V L2 (A3); or V L2 -V H2 (A4);
  • V L2 is the light chain variable region of the anti-second tumor cell marker antibody
  • V H2 is the heavy chain variable region of the anti-second tumor cell marker antibody
  • "-" is the connecting peptide (or flexible linker) or Peptide bonds.
  • amino acid sequence of V L2 is shown in positions 135-241 of SEQ ID NO.:5
  • amino acid sequence of V H2 is shown in positions 1-119 of SEQ ID NO.:5.
  • the first scFV and the second scFv comprise variant forms, and the variants have ⁇ 80%, ⁇ 85%, ⁇ 90%, ⁇ 95%, ⁇ 98% or ⁇ 99% homology.
  • the first scFV and the second scFV of the invention also include conservative variants thereof, which means that compared with the amino acid sequences of the first scFV and the second scFV of the invention respectively, there are at most 10, preferably At most 8, more preferably at most 5, most preferably at most 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%, and more preferably 1-33%. More preferably, it is 5-30%, more preferably, it is 10-25%, and even more preferably, it is 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, Optimally 1.
  • the CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR.
  • a transmembrane domain naturally associated with one of the domains in the CAR is used.
  • transmembrane domains may 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 interaction with the receptor complex. Interactions with other members.
  • the intracellular domain in the first CAR of the present invention includes the transmembrane region of CD8 and the signaling domain of CD3 ⁇ .
  • the intracellular domain in the second CAR of the present invention includes the transmembrane region of CD8 or CD28, and the costimulatory factor of 4-1BB or CD28.
  • amino acid sequence of the first CAR is as shown in any one of SEQ ID NO.: 2-4.
  • amino acid sequence of the second CAR is shown in any one of SEQ ID NO.: 7-14.
  • the amino acid sequence of the fusion protein containing the first CAR and the second CAR is as shown in any one of SEQ ID NO.: 23-26.
  • positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3 ⁇ , and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembrane region of CD28); positions 807-847 are costimulatory elements (such as CD28).
  • positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembr
  • positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3 ⁇ , and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembrane region of CD28); positions 807-847 are costimulatory elements (such as CD28).
  • positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembr
  • CAR-T cells As used herein, the terms “CAR-T cells”, “CAR-T” and “CAR-T cells of the present invention” all refer to the CAR-T cells of the present invention.
  • the CAR-T cells of the present invention can target tumor surface antigens. (such as PSMA), used to treat tumors with high expression or positive PSMA, especially solid tumors.
  • PSMA tumor surface antigens.
  • CAR-T cells have the following advantages over other T cell-based treatments: (1) The action process of CAR-T cells is not restricted by MHC; (2) Since many tumor cells express the same tumor antigen, targeting a certain 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) Use patients' autologous The 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 fusion protein containing the first CAR and the second CAR (i.e., logic BiCAR) of the present invention includes (i) an extracellular domain, which includes an antigen targeting a first tumor cell surface antigen; (ii) a third a hinge region; (iii) a first transmembrane domain; (iv) an optional first costimulator; and (iv) a signaling domain of CD3 ⁇ ; and; (v) a linking peptide (e.g., a self-cleaving protein ); (vi) an extracellular domain comprising an antigen targeting a second tumor cell surface antigen; (ii) a second hinge region; (iii) a second transmembrane domain; and (iv) a second costimulatory factor .
  • an extracellular domain which includes an antigen targeting a first tumor cell surface antigen
  • a third a hinge region e.g., a first transmembrane domain
  • an optional first costimulator e.g
  • 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.
  • the CAR-M cells of the present invention can target tumor surface antigens. (such as cMet and EGFR), used to treat tumors with high expression or positivity of tumor antigens (such as cMet and EGFR), especially solid tumors.
  • tumor surface antigens such as cMet and EGFR
  • tumor antigens such as cMet and EGFR
  • Macrophages are the main effectors and regulators of the innate immune system. They have phagocytic ability, can secrete pro-inflammatory factors, and present antigens to T cells to activate the immune system.
  • CAR-M 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 antigens. , present tumor cell antigens and activate endogenous T cells.
  • CAR-NK cells As used herein, the terms “CAR-NK cells”, “CAR-NK” and “CAR-NK cells of the present invention” all refer to the CAR-NK cells of the present invention.
  • the CAR-NK cells of the present invention can target tumor surface antigens (such as cMet and EGFR) and are used to treat tumors with high cMet and EGFR expression or positivity, especially solid tumors.
  • tumor surface antigens such as cMet and EGFR
  • Natural killer (NK) cells are a major type of immune effector cells that protect the body from viral infection and tumor cell invasion through non-antigen-specific pathways.
  • 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, such as: (1) they directly kill tumor cells by releasing perforin and granzyme, but have no killing effect on normal cells of the body; (2) they release A very small amount of cytokines thus reduces the risk of cytokine storm; (3) It is easy to amplify in vitro and develop into "off-the-shelf" products. Otherwise, it is similar to CAR-T cell therapy.
  • exogenous T cell antigen receptor refers to the ⁇ chain and ⁇ chain of TCR cloned from tumor reactive T cells through gene transfer technology, and through genetic engineering means, lentivirus or Retrovirus is used as a vector to transfer exogenous TCR into T cells.
  • Exogenous TCR-modified T cells can specifically recognize and kill tumor cells, and by optimizing the affinity of TCR and tumor-specific antigens, the affinity of T cells with tumors can be increased and the anti-tumor effect can be improved.
  • Nucleic acid sequences encoding the desired molecules can be obtained using recombinant methods known in the art, such as, for example, by screening libraries from cells expressing the gene, by obtaining the gene from a vector known to include the gene, or by using standard technology to isolate directly from cells and tissues containing the gene. Alternatively, the gene of interest can be produced synthetically.
  • the invention also provides vectors into which the expression cassette of the invention is inserted.
  • Vectors derived from retroviruses such as lentiviruses are suitable tools to achieve long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in daughter cells.
  • Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses, such as murine leukemia virus, in that 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 invention is typically operably linked to a promoter and incorporated into an expression vector.
  • This vector is suitable for replication and integration into eukaryotic cells.
  • Typical cloning vectors contain transcriptional and translational terminators, initial sequences, and promoters that can be used to regulate expression of the desired nucleic acid sequence.
  • the expression constructs of the present invention can also be used for nucleic acid immunization and gene therapy using standard gene delivery protocols. 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 incorporated by reference in their entirety.
  • the present invention provides gene therapy vectors.
  • the nucleic acid can be cloned into many types of vectors.
  • the nucleic acid can be cloned into such vectors, which include, but are not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids.
  • Specific vectors of interest include expression vectors, replication vectors, probe generation vectors and sequencing vectors.
  • the expression vector can be provided to the cell in the form of a viral vector.
  • Viral vector technology is well known in the art and is described, for example, in 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, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses.
  • a suitable vector will contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (eg, WO01/96584; WO01/29058; and U.S. Patent No. 6,326,193).
  • retroviruses provide a convenient platform for gene delivery systems.
  • the selected genes can be inserted into the vector and packaged into retroviral particles using techniques known in the art.
  • the recombinant virus can then be isolated and delivered to cells of the subject in vivo or ex vivo.
  • retroviral systems are known in the art.
  • adenoviral vectors are used.
  • Many adenoviral vectors are known in the art.
  • lentiviral vectors are used.
  • promoter elements can modulate the frequency with which transcription is initiated.
  • these are located in a region of 30-110 bp 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 so that promoter function is maintained when the elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased by 50 bp before activity begins to decrease.
  • individual elements are shown to act cooperatively or independently to initiate transcription.
  • 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 thereto.
  • Another example of a suitable promoter is elongation growth factor-1 ⁇ (EF-1 ⁇ ).
  • constitutive promoter sequences may also be used, including, but not limited to, simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLVqi kinesin, avian leukemia virus promoter, Epstein-Barr virus immediate early promoter, Ruth's 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 use of constitutive promoters. Inducible promoters are also considered part of the invention.
  • an inducible promoter provides a molecular switch capable of turning on expression of a polynucleotide sequence operably linked to the inducible promoter when such expression is desired, or turning off expression when expression is undesirable.
  • inducible promoters include, but are not limited to, metallothionein promoters, glucocorticoid promoters, progesterone promoters, and tetracycline promoters.
  • the expression vector introduced into the cell may also contain either or both a selectable marker gene or a reporter gene to facilitate the identification of populations of cells that are transfected or infected by the viral vector. Identify and select expressing cells.
  • the selectable marker can be carried on a separate stretch of DNA and used in co-transfection procedures. Both the selectable marker and the reporter gene can be flanked by appropriate regulatory sequences to enable expression 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 is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is clearly indicated by some readily detectable property, such as enzymatic activity. Expression of the reporter gene is measured at appropriate times after the DNA has been introduced into the recipient cell.
  • 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.
  • the construct with a minimum of 5 flanking regions that shows the highest level of reporter gene expression is identified as the promoter.
  • Such promoter regions can be ligated to a reporter gene and used to evaluate the ability of an agent to regulate promoter-driven transcription.
  • the vector can be readily introduced into a host cell, eg, a mammalian, bacterial, yeast or insect cell, by any method known in the art.
  • expression vectors can be transferred into host cells by physical, chemical or biological means.
  • Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. 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). Preferred methods for introducing polynucleotides into host cells The method 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 lentiviruses, poxviruses, herpes simplex virus I, adenovirus and adeno-associated virus, among others. See, for example, US Patent Nos. 5,350,674 and 5,585,362.
  • colloidal dispersion systems such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems, including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • Exemplary colloidal systems useful as delivery vehicles in vitro and in vivo are liposomes (eg, artificial membrane vesicles).
  • an exemplary delivery vehicle is liposomes.
  • lipid formulations to introduce nucleic acids into host cells (in vitro, ex vivo, or in vivo).
  • the nucleic acid can be associated with a lipid.
  • Nucleic acids associated with lipids can be encapsulated into the aqueous interior of the liposomes, dispersed within the lipid bilayer of the liposomes, attached via linker molecules associated with both the liposomes and the oligonucleotides to liposomes, entrapped in liposomes, complexed with liposomes, dispersed in a solution containing lipids, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or Complexed with micelles, or otherwise associated with lipids.
  • the lipid, lipid/DNA or lipid/expression vector associated with the composition is not limited to any specific structure in solution.
  • Lipids are fatty substances, which may be naturally occurring or synthetic lipids.
  • lipids include lipid droplets that occur naturally in the cytoplasm as well as compounds containing long-chain aliphatic hydrocarbons and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes.
  • the vector is a retroviral vector.
  • the invention provides an engineered immune cell according to the first aspect of the 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, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine ; antioxidant; chelating agent such as EDTA or glutathione; adjuvant agents (e.g., aluminum hydroxide); and preservatives.
  • buffers such as neutral buffered saline, sulfate buffered saline, and the like
  • carbohydrates such as glucose, mannose, sucrose or dextran, mannitol
  • proteins polypeptides or amino acids
  • antioxidant chelating agent
  • adjuvant agents e.g., aluminum hydroxide
  • preservatives e.g., aluminum hydroxide
  • the present invention encompasses the therapeutic use of cells (eg, T cells) transduced with a retroviral vector (RVV) encoding an expression cassette of the invention.
  • the transduced T cells can target tumor cell marker proteins (such as cMet and EGFR), cooperatively activate T cells, trigger cellular immune responses, and thereby selectively kill tumor cells, such as tumor cells with high expression of cMet and EGFR.
  • tumor cell marker proteins such as cMet and EGFR
  • the present invention also provides a method of stimulating a T cell-mediated immune response to a target cell population or tissue of a mammal, comprising the steps of administering a CAR-T cell of the present invention to the mammal.
  • the present invention includes a type of cell therapy in which a patient's autologous T cells (or allogeneic donors) are isolated, activated and genetically modified to produce CAR-T cells, and then injected into the same patient.
  • This method has a very low probability of suffering from graft-versus-host disease, and the antigen is recognized by T cells in an MHC-free manner.
  • one CAR-T can treat all cancers that express this antigen.
  • CAR-T cells are able to replicate in vivo, producing long-term persistence that can lead to sustained tumor control.
  • CAR-T cells of the invention can undergo robust in vivo T cell expansion for an extended amount of time.
  • CAR-mediated immune responses can be part of an adoptive immunotherapy step, in which CAR-modified T cells induce an immune response specific for the antigen-binding domain in the CAR.
  • CAR-T cells expressing tumor cell markers such as cMet, EGFR
  • elicit a specific immune response against cells expressing tumor cell markers such as cMet, EGFR
  • the data disclosed herein specifically disclose including an antigen-binding domain targeting a first tumor cell surface antigen, the hinge and transmembrane regions, and the CD3 ⁇ signaling domain, P2A, an antigen-binding domain targeting a second tumor cell surface antigen, hinge and transmembrane regions, and 4-1BB/CD28 retroviral vectors, but the invention should be construed to include any number of changes in each part of the construct composition.
  • Treatable cancers include tumors that are not vascularized or substantially unvascularized, as well as tumors that are vascularized.
  • Cancer may include non-solid tumors (such as hematological tumors, such as leukemias and lymphomas) or solid tumors.
  • Cancer types treated with the CARs of the invention include, but are not limited to, carcinomas, blastomas, and sarcomas, and certain leukemias or lymphoid malignancies, benign and malignant tumors, and malignancies, such as sarcomas, carcinomas, and melanomas. Also includes adult neoplasms/cancers and pediatric neoplasms/cancers.
  • Hematologic cancers are cancers of the blood or bone marrow.
  • hematological (or hematogenous) cancers include leukemias, including acute leukemias such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia, and myeloblastoid, promyelocytic, myelomonocytic types , monocytic and red and white blood diseases), chronic leukemias (such as chronic myeloid (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high-grade forms), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disorders, myelodysplastic syndromes, hairy cell leukemia, and myelodysplasia.
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or areas of fluid. Solid tumors can be benign or malignant. Different types of solid tumors are named after the cell types that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include prostate cancer, liver cancer, fibrosarcoma, myxosarcoma, liposarcoma mesothelioma, lymphoid malignancies, pancreatic cancer, ovarian cancer.
  • the CAR-modified T cells of the present invention may also be used as a type of vaccine for ex vivo immunization and/or in vivo therapy of mammals.
  • the mammal is human.
  • cells are isolated from a mammal (preferably human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein.
  • CAR-modified cells can be administered to mammalian recipients to provide therapeutic benefit.
  • the mammalian recipient can be human, and the CAR-modified cells can be autologous to the recipient.
  • the cells may be allogeneic, syngeneic, or xenogeneic relative to the recipient.
  • the present invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient.
  • the invention provides methods of treating tumors comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-modified T cell of the invention.
  • the CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition in combination with diluents and/or with other components or other cytokines or cell populations.
  • a pharmaceutical composition of the present invention may comprise a 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, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelates Adjuvants such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives.
  • the compositions of the present invention are preferably formulated for intravenous administration.
  • compositions of the present invention may be administered in a manner suitable for the disease to be treated (or prevented).
  • the amount and frequency of administration will be determined by factors such as the patient's condition, and the type and severity of the patient's condition. Degree—Although appropriate dosage can be determined by clinical trials.
  • compositions of the invention to be administered can be determined by the physician, who takes into account the patient (subject) ) age, weight, tumor size, degree of infection or metastasis, and individual differences in disease. It may generally be stated that pharmaceutical compositions comprising T cells described herein may be administered at a dose of 10 4 to 10 9 cells/kg body weight, preferably 10 5 to 10 6 cells/kg body weight (including all integers within those ranges). value) application. T cell compositions can also be administered multiple times at these dosages.
  • Cells can be administered using infusion techniques well known in immunotherapy (see, eg, Rosenberg et al., New Eng. J. of Med. 319:1676, 1988).
  • the optimal dosage and treatment regimen for a particular patient can be readily determined by one skilled in the medical field by monitoring the patient for signs of disease and adjusting treatment accordingly.
  • compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intraspinally, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally.
  • the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection.
  • the T cell composition of the invention is preferably administered by i.v. injection.
  • the composition of T cells can be injected directly into the tumor, lymph node or site 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., before , simultaneously or subsequently) administered to a patient, such forms of treatment include, but are not limited to, treatment with agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as for ARA-C) or natalizumab treatment in patients with MS or elfalizumab treatment in patients with psoriasis or other treatments in patients with PML.
  • agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as for ARA-C) or natalizumab treatment in patients with MS or elfalizumab treatment in patients with psoriasis or other treatments in patients with PML.
  • the T cells of the invention can be used in combination with chemotherapy, radiation, immunosuppressants such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil and FK506, antibodies or other immunotherapeutic agents.
  • the cellular compositions of the invention are administered in conjunction with (eg, before, simultaneously with, or after) bone marrow transplantation, use of a chemotherapeutic agent such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide patient.
  • a subject may undergo standard treatment with high-dose chemotherapy followed by a peripheral blood stem cell transplant.
  • the subject receives an infusion of expanded immune cells of the invention.
  • the expanded cells are administered before or after surgery.
  • the dosage of the above treatments administered to a patient will vary depending on the precise nature of the condition being treated and the recipient of the treatment. Dosage proportions for human administration may be implemented in accordance with art-accepted practice. Typically, each treatment Or for each course of treatment, 1 ⁇ 10 6 to 1 ⁇ 10 10 modified T cells of the present invention (eg, CAR-T cells of the present invention) can be administered to the patient, for example, by intravenous infusion.
  • modified T cells of the present invention eg, CAR-T cells of the present invention
  • fusion protein As used herein, the terms “fusion protein”, “fusion protein of the present invention”, “polypeptide of the present invention” and “logic BiCAR” 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 as shown in any one of SEQ ID NO.: 23-26.
  • fusion protein also includes variant forms of any of the sequences shown in SEQ ID NO.: 23-26 having the above-mentioned activities. These variant forms include (but are not limited to): deletion, insertion and/or substitution of 1-3 (usually 1-2, more preferably 1) amino acids, and addition of or One or several (usually within 3, preferably within 2, and more preferably within 1) amino acids are missing. For example, in the art, substitutions with amino acids with similar or similar properties generally do not alter the function of the protein. For another example, 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. Furthermore, the term also includes monomeric and multimeric forms of the polypeptides of the invention. The term also includes linear as well as non-linear polypeptides (such as cyclic peptides).
  • the present invention also includes active fragments, derivatives and analogs of the above-mentioned fusion proteins.
  • fragment refers to polypeptides that substantially retain the function or activity of the fusion proteins of the invention.
  • the polypeptide fragments, derivatives or analogs of the present invention may be (i) a polypeptide in which one or several conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted, or (ii) in which one or more conservative amino acid residues are substituted.
  • a polypeptide with a substituent group in an amino acid residue or (iii) a polypeptide formed by fusion of a polypeptide with another compound (such as a compound that extends the half-life of the polypeptide, such as polyethylene glycol), or (iv) a fusion of additional amino acid sequences
  • a polypeptide formed from this polypeptide sequence a fusion protein formed by fusion with a leader sequence, secretory sequence or tag sequence such as 6His.
  • Such fragments, derivatives and analogs are within the scope of those skilled in the art in light of the teachings herein.
  • a preferred type of active derivative refers to one in which at most 3, preferably at most 2, and more preferably at most 1 amino acid is replaced by an amino acid with similar or similar properties compared to the amino acid sequence of the present invention to form a polypeptide.
  • These conservative variant polypeptides are preferably produced by amino acid substitutions according to Table 1.
  • the invention also provides analogs of the fusion proteins of the invention.
  • the difference between these analogs and the polypeptides shown in any of SEQ ID NO.: 23-26 can be differences in amino acid sequences, differences in modified forms that do not affect the sequence, or both.
  • Analogues also include analogs with residues that differ from natural L-amino acids (eg, D-amino acids), as well as analogs with non-naturally occurring or synthetic amino acids (eg, beta, gamma-amino acids). It should be understood that the polypeptides of the present invention are not limited to the representative polypeptides exemplified above.
  • Modified forms include chemically derivatized forms of the polypeptide, such as acetylation or carboxylation, either in vivo or in vitro. Modifications also include glycosylation, such as those resulting from glycosylation modifications of the polypeptide during its synthesis and processing 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 having phosphorylated amino acid residues (eg, phosphotyrosine, phosphoserine, phosphothreonine). Also included are polypeptides that have been modified to increase their resistance to proteolysis or to optimize solubility properties.
  • glycosylation such as those resulting from glycosylation modifications of the polypeptide during its synthesis and processing or during further processing steps. This modification can be accomplished by exposing the polypeptide to an enzyme that performs glycosylation, such as a mammalian glycosy
  • the amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
  • the invention also relates to polynucleotides encoding fusion proteins according to the invention.
  • the polynucleotides of the invention may be in DNA form or RNA form.
  • DNA can be a coding strand or a non-coding strand.
  • the sequence of the coding region encoding the mature polypeptide may be identical to the sequence encoding the polypeptide shown in any one of SEQ ID NO.: 23-26 or may be a degenerate variant.
  • degenerate variant in the present invention refers to a nucleic acid sequence encoding a polypeptide shown in any one of SEQ ID NO.: 23-26, but with different sequences in the corresponding coding regions.
  • the full-length nucleotide sequence of the present invention or its fragment can usually be obtained by PCR amplification, recombination or artificial synthesis.
  • the DNA sequence encoding the polypeptide of the present invention (or its fragment, or its derivative) can be obtained entirely through chemical synthesis.
  • the DNA sequence can then be introduced into a variety of existing DNA molecules (or vectors) and cells known in the art.
  • the invention also relates to vectors comprising the polynucleotides of the invention, as well as host cells genetically engineered with the vectors or polypeptide coding sequences of the invention.
  • the polynucleotides, vectors or host cells described above may be isolated.
  • isolated means that a substance has been separated from its original environment (in the case of a natural substance, the original environment is the natural environment).
  • the original environment is the natural environment.
  • polynucleotides and polypeptides in the natural state within living cells are not isolated and purified, but the same polynucleotide or polypeptide is isolated and purified if it is separated from other substances existing in the natural state.
  • the polynucleotides of the invention may be in DNA form or RNA form.
  • Forms of DNA include 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 above-mentioned polynucleotides, which encode protein fragments, analogs and derivatives having the same amino acid sequence as the present invention.
  • Variants of this polynucleotide may 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, which may be the substitution, deletion or insertion of one or more nucleotides, but does not substantially change its encoding of the fusion protein of the invention. function.
  • the full-length nucleotide sequence encoding the fusion protein of the present invention or its fragment can usually be obtained by PCR amplification, recombination or artificial synthesis.
  • primers can be designed based on the relevant published nucleotide sequences, especially the open reading frame sequence, and a commercially available cDNA library or a cDNA library prepared according to conventional methods known to those skilled in the art can be used as the primer.
  • Template amplified to obtain the relevant sequence. When 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 any one of SEQ ID NO.: 27-30.
  • recombination can be used to obtain the relevant sequence in large quantities. This is usually done by cloning it into a vector, transforming it into cells, and then isolating the relevant sequence from the propagated host cells by conventional methods.
  • artificial synthesis methods can also be used to synthesize relevant sequences, especially when the fragment length is short. Often, fragments with long sequences are obtained by first synthesizing multiple small fragments and then ligating them.
  • the method of amplifying DNA/RNA using PCR technology is preferably used to obtain the gene of the present invention.
  • Primers for PCR can be appropriately selected based on 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 using conventional methods such as by gel electrophoresis.
  • the present invention also relates to vectors containing the polynucleotides of the present invention, as well as host cells genetically engineered using the vectors or protein coding sequences of the present invention, and methods for expressing the fusion proteins of the present invention on the NK cells using 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 speaking, it includes the steps of: transducing the first expression cassette and/or the second expression cassette of the present invention into NK cells, thereby obtaining the NK cells.
  • expression vectors containing the DNA sequence encoding the fusion protein of the invention and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA technology, DNA synthesis technology, in vivo recombination technology, etc.
  • the DNA sequence can be effectively linked to an appropriate promoter in an expression vector to direct 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, neomycin resistance, and green color for eukaryotic cell culture.
  • selectable marker genes to provide phenotypic traits for selection of transformed host cells, such as dihydrofolate reductase, neomycin resistance, and green color for eukaryotic cell culture.
  • GFP Fluorescent protein
  • tetracycline or ampicillin resistance in E. coli tetracycline or ampicillin resistance in E. coli.
  • Vectors containing appropriate DNA sequences as described above and appropriate promoter or control sequences can be used to transform appropriate host cells to enable expression of proteins.
  • the host cell can be a prokaryotic cell, such as a bacterial cell; 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 include: bacterial cells of Escherichia coli, Bacillus subtilis, and Streptomyces; fungal cells such as Pichia pastoris and Saccharomyces cerevisiae cells; plant cells; insect cells of Drosophila S2 or Sf9; CHO, NS0, COS7, or 293 cells animal cells etc.
  • NK cells are selected as host cells.
  • Transformation of host cells with recombinant DNA can be performed using conventional techniques well known to those skilled in the art.
  • the host is a prokaryotic organism such as E. coli
  • competent cells capable of absorbing DNA can be harvested after the exponential growth phase and treated with CaCl 2
  • CaCl 2 The procedures used are well known in the art.
  • Another method is to use MgCl 2 .
  • transformation can also be performed by electroporation.
  • 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 using conventional methods to express the protein encoded by the gene of the present invention.
  • the medium used in culture can be selected from various conventional media. Cultivate under conditions suitable for host cell growth. After the host cells grow to an appropriate cell density, the selected promoter is induced using an appropriate method (such as temperature shift or chemical induction), and the cells are cultured for a further period of time.
  • the protein in the above method can be expressed within the cell, on the cell membrane, or secreted outside the cell. If desired, proteins can be isolated and purified by various separation methods using their physical, chemical and other properties. These methods are well known to those skilled in the art. Examples of these methods include, but are not limited to: conventional refolding treatment, treatment with protein precipitating agents (salting out method), centrifugation, osmotic sterilization, ultratreatment, ultracentrifugation, 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 discovered for the first time a first CAR targeting a first tumor cell marker (such as cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47) and a second tumor cell marker (such as EGFR , EpCAM, Her2, Her3), the engineered immune cells of the second CAR can be fully activated, enhance target specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor toxicity and improve cell Safety of treatment.
  • a first tumor cell marker such as cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47
  • a second tumor cell marker such as EGFR , EpCAM, Her2, Her3
  • the present invention discovered for the first time that the immune cells of the present invention can simultaneously recognize two different antigens, EGFR and cMet, and enhance the specificity and selectivity of CAR T cells in targeting tumors, as well as their homing in tumors. In addition, it can inhibit two pathogenic signaling pathways at the same time, thereby inhibiting disease progression and improving survival.
  • Phoenix GP cells in the logarithmic growth phase were seeded into T75 culture flasks at a density of 4E6/flask, and cultured overnight in a 37°C, 5% CO2 incubator for transfection.
  • the culture medium was DMEM containing 10% FBS. culture medium. The next day, the culture medium was replaced with fresh DMEM before transfection.
  • the transfection steps are as follows: Add the target gene plasmid, such as pMSGV-CAR, and the envelope plasmid pMD2.G to the Opti-MEM medium and mix well; then add DNA transfection reagent and mix well; add it dropwise into the culture bottle and culture for 18 hours.
  • PG13 cells in the logarithmic growth phase were seeded into a 6-well plate at a density of 7E4/well and cultured overnight in a 37°C, 5% CO2 incubator for transfection.
  • the culture medium was DMEM containing 10% FBS. base.
  • the culture medium in the 6-well plate was replaced with the aforementioned filtered culture supernatant and the virus infection enhancer Polybrene was added. After 24 h of culture, the culture medium was replaced with fresh DMEM.
  • PG13-CAR retrovirus production cell line After 72 hours of culture, the transduction efficiency of PG13-CAR was detected to construct a PG13-CAR retrovirus production cell line, which was amplified and cultured before cryopreservation.
  • PG13-CAR cells in the logarithmic growth phase were inoculated into T75 culture bottles at a density of 2E7/flask, and cultured in a 37°C, 5% CO2 incubator for 24 hours. The supernatant containing retrovirus was collected, centrifuged, filtered, and infected. cell.
  • Mononuclear cells were isolated from peripheral blood of healthy individuals by Ficoll-Hypaque density gradient centrifugation. Add TransAct activator and recombinant human IL-2 to the lymphocyte culture medium to stimulate, activate and expand T cells; at the same time, use Retronectin to coat a 6-well plate, add the retrovirus supernatant to the 6-well plate and centrifuge; After centrifugation is completed, discard the supernatant, add the activated T cells to the 6-well plate, and centrifuge for transduction; after culturing for 24 hours in a 37°C, 5% CO2 incubator, transfer the T cells from the 6-well plate to the culture chamber. Amplification culture was carried out in bottles.
  • HiBiT tag protein detection method HiBiT Extracellular Detection System, Promega
  • Collect the target cells carrying the HaloTag-HiBiT label (including NCI-H1975-Halotag-HiBiT, EBC-1-HaloTag-HiBiT) in the logarithmic growth phase and the CAR T cells cultured to day 7, and prepare cells with different cell concentrations. Suspension; target cells were seeded in a 96-well plate (3 multiple wells) at 5000 cells/well, then CAR T cell suspension was added according to different effect-to-target ratios, and cultured in a 37°C, 5% CO 2 incubator for 24 hours. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes. Then remove the well plate from the incubator.
  • HaloTag-HiBiT label including NCI-H1975-Halotag-HiBiT, EBC-1-HaloTag-HiBiT
  • Coxicity assay of CAR-T cells was performed using CCK8 kit (Doren Chemical Research Institute). Collect the target cells in the logarithmic growth phase and the CAR T cells cultured to day 7 to prepare cell suspensions with different cell densities. Take 50ul target cells and plate them at 5000 cells/well (3 replicate wells), and then use 3:1, Add 50ul of CAR T cell suspension at the effect-to-target ratio of 1:1, 1:3 and 1:10, and incubate for 24hr in a 5% CO2 incubator at 37°C and saturated humidity, then add 10ul of CCK8 to each well and react for 4hr. Finally, the absorbance at a wavelength of 450 nm was measured using a microplate reader (Thermo Varioskan LUX).
  • NCI-H820 tumor cells in the logarithmic growth phase were collected and inoculated subcutaneously on the backs of immune-deficient mice.
  • CAR T cells were infused into the tail vein, and the mice's status was subsequently observed every week.
  • the size of the tumor mass was measured twice and the mouse tumor growth curve was drawn to evaluate the in vivo anti-tumor activity of CAR T cells.
  • Virus preparation Add retrovirus target gene plasmid pMSGV-CAR and envelope plasmid pMD2.G Mix it into X-tremeGENE 9 DNA Transfection Reagent (Roche), add it to the culture dish cultivating Phoenix GP cells, mix gently, collect the supernatant after 72 hours, centrifuge at 1000g at low speed, filter with 0.45um filter membrane, and filter the supernatant Add it to PG13 cells and add Polybrene transfection enhancer to prepare the retrovirus production cell line PG13-CAR. Inoculate PG13-CAR at 2E7/flask density into T75 cell culture flasks. Collect the supernatant after 24 hours of culture. Centrifuge at 400g at low speed and filter with a 0.45um filter. Use it directly or aliquot and freeze at -80°C.
  • CAR T cell preparation Obtain mononuclear cells from healthy human peripheral blood by Ficoll-Hypaque density gradient centrifugation and inoculate them into lymphocyte culture medium (Gibco), add TransAct (Miltenyi Biotec) and IL-2 (GE Healthcare) for stimulation activation and expansion culture.
  • Retronectin to coat a 6-well plate, add the retrovirus supernatant to the coated 6-well plate, and centrifuge at 2000g for 2 hours; after centrifugation is completed, discard the supernatant, and add the activated T cells to the 6-well plate. Centrifuge at 1000g for 20 minutes for transduction, place in a 37°C, 5% CO2 incubator, and culture.
  • Figure 1A shows the composition of the "AND" logic BiCAR targeting EGFR and cMet, including 2 independent CAR molecules.
  • the CAR1 molecule is an anti-cMet scFv tandem CD3 ⁇ activation signal domain ( As shown in SEQ ID NO.: 2, SEQ ID NO.: 3-4 is a fully functional 2G single CAR against cMet), and the CAR2 molecule is an anti-EGFR scFv tandem CD28 or 4-1BB costimulatory signaling domain (such as Any one of SEQ ID NO.: 7-10 is shown, wherein SEQ ID NO.: 11-14 is a fully functional 2G single CAR against EGFR);
  • Figure 1B shows a schematic diagram of the molecular construction of "AND" logic BiCAR expression, CAR1 The molecule and the CAR2 molecule are linked through P2A (as shown in any of SEQ ID NO.: 23-26
  • Detect the expression of CAR molecules by flow cytometry When the CAR T cells are cultured on the 7th day, take the 1E6 cells, wash them twice with washing buffer, and add 1ug cMet-His (Acrobiosystem) and 1ug EGFR-Fc antigen (Acrobiosystem) , after incubating at 4°C for 30 minutes, wash twice with wash buffer, then add Anti-His-PE (Miltenyi Biotec) and Allophycocyanin (APC) AffiniPure Goat Anti-Human IgG, Fc ⁇ fragment specific (Jackson ImmunoResearch) and incubate at 4°C in the dark for 30 minutes . After washing the cells, flow cytometry (CytoFLEX LX, Beckman Coulter) was used to detect the expression of CAR molecules.
  • flow cytometry CytoFLEX LX, Beckman Coulter
  • HiBiT Extracellular Detection System Promega method to detect cytotoxicity: collect the lung cancer cell line NCI-H1975-HaloTag-HiBiT carrying the HaloTag-HiBiT label in the logarithmic growth phase and CAR T cells cultured to day 7, and prepare different cell concentrations of cell suspension. Inoculate target cells into a 96-well plate at 5000 cells/well (3 multiple wells), then add CAR T cell suspension according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, 37°C, 5 Incubate for 24 hours in a % CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes.
  • Buffer (contains 1:100 diluted LgBiT Protein and 1:50 diluted HiBiT Extracellular Substrate), mix on a shaker, use a microplate reader to detect the luminescence value, and calculate the killing activity of CAR T cells.
  • HiBiT Extracellular Detection System (Promega) method to detect cytotoxicity: collect the lung cancer cell line EBC-1-Halotag-HiBiT carrying the HaloTag-HiBiT label in the logarithmic growth phase and CAR T cells cultured to day 7, and prepare different cell concentrations of cell suspension.
  • the target cells were seeded into a 96-well plate (3 multiple wells) at 5000cells/well, and then the CAR T cell suspension was added according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, at 37°C. Incubate for 24 hours in a 5% CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes.
  • Coxicity assay of CAR-T cells was performed using CCK8 kit (Doren Chemical Research Institute). Collect the human gastric cancer cell line SNU5 in the logarithmic growth phase and the CAR T cells cultured to day 7 to prepare cell suspensions with different cell densities. Take 50ul of target cells and plate them at 5000 cells/well (3 replicate wells), and then use 3 :1, 1:1, 1:3 and 1:10, add 50ul CAR T cell suspension and incubate it in a 5% CO2 incubator at 37°C and saturated humidity for 24hr, then add 10ul CCK8 to each well. , after 4 hours of reaction, use a microplate reader (Thermo Varioskan LUX) to measure the absorbance at a wavelength of 450 nm.
  • a microplate reader Thermo Varioskan LUX
  • HiBiT Extracellular Detection System (Promega) method to detect cytotoxicity: collect stably transfected cell lines 293-Halotag-HiBiT, 293-Halotag-HiBiT-MET, 293-Halotag-HiBiT-EGFR carrying HaloTag-HiBiT tags in logarithmic growth phase , 293-Halotag-HiBiT-MET&EGFR, and CAR T cells cultured to day 7 to prepare cell suspensions with different cell concentrations.
  • the target cells were seeded into a 96-well plate (3 multiple wells) at 5000cells/well, and then the CAR T cell suspension was added according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, at 37°C. Incubate for 24 hours in a 5% CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes.
  • Figure 6 shows the response of BiCAR T cells to the 293T cell line co-expressing EGFR and cMet (6A), the 293T cell line expressing cMet alone (6B), the 293T cell line expressing EGFR alone (6C), and the control 293T cell line.
  • (6D) In vitro cytolytic toxicity after 24 h of co-incubation. The ratios of CAR-T effector cells to target cells are 1:10, 1:3, 1:1 and 3:1 respectively. The results showed that BiCAR T cells exhibited a dose-dependent and specific in vitro killing function against stably transduced cell lines expressing cMet as a single target or cMet/EGFR as both targets.
  • Mouse CDX model was used to detect the anti-tumor activity of CAR T cells: NCI-H820 cells in the logarithmic growth phase were collected and inoculated subcutaneously on the backs of immunodeficient mice. When the tumor volume reaches about 120mm3 , mice with similar tumor burdens are randomly divided into groups, and Mock T, O-28z CAR and BiCAR T cells are infused into the tail vein. The size of subcutaneous tumors was measured twice a week and the status of mice was observed, and tumor growth curves were drawn to evaluate the in vivo anti-tumor activity of CAR T cells.

Abstract

The present invention provides an anti-EGFR and cMet bispecific chimeric antigen receptor and use thereof. Specifically, the present invention provides an immune cell expressing a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the second CAR targets a second tumor cell marker. The first CAR and the second CAR of the present invention recognize the corresponding tumor cell markers at the same time, so that the immune cell can be fully activated to enhance the targeting specificity and exert the anti-tumor effect, and the on-target/off-tumor toxicity is reduced, thereby improving the safety of cell therapy.

Description

抗EGFR和cMet双特异性嵌合抗原受体及其应用Anti-EGFR and cMet bispecific chimeric antigen receptors and their applications 技术领域Technical field
本发明属于生物技术领域。具体地,本发明涉及抗EGFR和cMet双特异性嵌合抗原受体及其应用。The invention belongs to the field of biotechnology. In particular, the present invention relates to anti-EGFR and cMet bispecific chimeric antigen receptors and their uses.
背景技术Background technique
经过基因工程改造表达嵌合抗原受体(CAR)的T细胞已经被证明对某些B细胞白血病或者淋巴瘤亚型的患者具有治疗作用,并且对多发性骨髓瘤患者也显示了潜在的疗效。目前已有3个CAR T药物获批,分别为Yescarta用于治疗大B细胞淋巴瘤和滤泡性淋巴瘤,Tecartus用于治疗套细胞淋巴瘤,以及Kymriah用于治疗大B细胞淋巴瘤和针对小儿和年轻人的急性淋巴细胞白血病。CAR分子包括3个主要的结构成分,分别是细胞外抗原结合功能结构域;跨膜结构域,用以传递抗原识别信号;以及细胞内信号传导结构域。第一代的CAR仅包含CD3ζ激活信号,由于缺乏共刺激信号功能结构域,无法使CAR T细胞充分激活,显示出有限的细胞毒性,不能在体内有效扩增或持续存在。第二代CAR同时引入了CD28或4-1BB的共刺激功能结构域,增强了CAR T的细胞毒性,并提高了其持久性。目前CAR T细胞产品的上市药物均采用了二代的设计。通过添加另一个共刺激结构域,例如OX40、ICOS、CD27等,第三代CAR在二代的基础上进一步扩展,尽管报道其具有更强的持久性,但有可能会诱发细胞因子过量释放,目前在临床上的疗效并没有得到验证。为了扩展CAR T的疗效并将其应用到更广泛的恶性肿瘤,尤其是实体瘤,需要对CAR T进行创新性的工程化构建。T cells genetically engineered to express chimeric antigen receptors (CAR) have been shown to have a therapeutic effect on patients with certain B-cell leukemias or lymphoma subtypes, and have also shown potential efficacy in patients with multiple myeloma. Currently, three CAR T drugs have been approved, namely Yescarta for the treatment of large B-cell lymphoma and follicular lymphoma, Tecartus for the treatment of mantle cell lymphoma, and Kymriah for the treatment of large B-cell lymphoma and Acute lymphoblastic leukemia in children and young adults. CAR molecules include three main structural components, namely the extracellular antigen-binding functional domain; the transmembrane domain to transmit antigen recognition signals; and the intracellular signaling domain. The first generation of CAR only contains CD3ζ activation signal. Due to the lack of costimulatory signal functional domain, it cannot fully activate CAR T cells, shows limited cytotoxicity, and cannot effectively expand or persist in the body. The second-generation CAR also introduced the costimulatory functional domain of CD28 or 4-1BB, which enhanced the cytotoxicity of CAR T and improved its persistence. Currently, all CAR T cell products on the market adopt second-generation designs. By adding another costimulatory domain, such as OX40, ICOS, CD27, etc., the third generation CAR further expands on the second generation. Although it is reported to have stronger persistence, it may induce excessive release of cytokines. At present, its clinical efficacy has not been verified. In order to expand the efficacy of CAR T and apply it to a wider range of malignant tumors, especially solid tumors, innovative engineering construction of CAR T is needed.
在CAR T细胞治疗过程中会出现多种不良反应,如细胞因子释放综合征(CRS),神经系统毒性,在靶/脱肿瘤的靶点毒性(on-target off-tumor toxicity)以及肿瘤溶解综合征。其中CRS是系统性的,可累及全身各个器官,最频发以及症状最突出的急性不良反应,在临床上需要有效地对CRS进行管理和干预。由于肿瘤相关抗原在肿瘤细胞中高表达以外,也会在正常组织或细胞中有非特异性的低水平表达,例如CD19、Her2、ROR1、Muc1等。靶点毒性即CAR T细胞因识别正常组织表达的抗原而被激活,并造成对正常组织的损伤。CAR T治疗CD19阳性的恶性淋巴瘤,由于骨髓中具有表达CD19的B细胞祖细 胞,其同肿瘤细胞一并被清楚,从而导致B细胞发育不良以及血红蛋白降低,临床上通过输注免疫球蛋白来代替B细胞产生的抗体功能。在一项以碳酸酐酶IX为靶点,治疗肾癌的CAR T细胞临床试验中,多名患者出现了肝酶异常,这些不良反应归因于CAR T细胞渗入并且作用于表达碳酸酐酶的胆管上皮。一名结直肠癌患者,在接受靶向Her2的CAR T细胞输注后,由于肺上皮细胞也表达低水平的Her2,患者则出现了致命的肺损伤。另外,在靶向EGFRvIII的CAR T治疗神经胶质瘤的临床试验中,也发生了类似的肺水肿毒性。由于单一靶向CAR T细胞有可能造成靶点毒性,并且目前很难通过IHC或者组织芯片等体外筛选的方法进行准确判断。Various adverse reactions may occur during CAR T cell therapy, such as cytokine release syndrome (CRS), neurological toxicity, on-target off-tumor toxicity, and tumor lysis syndrome. levy. Among them, CRS is systemic and can affect various organs of the body. It is the most frequent and symptomatic acute adverse reaction. Effective clinical management and intervention of CRS is required. As tumor-related antigens are highly expressed in tumor cells, they are also expressed at non-specific low levels in normal tissues or cells, such as CD19, Her2, ROR1, Muc1, etc. Target toxicity means that CAR T cells are activated by recognizing antigens expressed by normal tissues and cause damage to normal tissues. CAR T treats CD19-positive malignant lymphoma because there are CD19-expressing B cell progenitor cells in the bone marrow. cells, which are eliminated together with tumor cells, resulting in B cell dysplasia and reduced hemoglobin. Clinically, immunoglobulin is infused to replace the antibody function produced by B cells. In a clinical trial of CAR T cells targeting carbonic anhydrase IX to treat renal cancer, several patients developed liver enzyme abnormalities. These adverse reactions were attributed to CAR T cell infiltration and action on carbonic anhydrase-expressing cells. Bile duct epithelium. A patient with colorectal cancer developed fatal lung damage after receiving an infusion of Her2-targeting CAR T cells because lung epithelial cells also expressed low levels of Her2. In addition, similar pulmonary edema toxicity also occurred in clinical trials of EGFRvIII-targeted CAR T therapy for glioma. Since single-targeting CAR T cells may cause target toxicity, it is currently difficult to accurately judge through in vitro screening methods such as IHC or tissue chips.
因此本领域迫切需要开发一种新型的嵌合抗原受体T细胞,既可同时识别多个抗原,增强靶向特异性,发挥抗肿瘤功效,并且能够降低在靶/脱肿瘤毒性,提高细胞治疗的安全性。Therefore, there is an urgent need in this field to develop a new type of chimeric antigen receptor T cell, which can recognize multiple antigens at the same time, enhance targeting specificity, exert anti-tumor efficacy, and can reduce on-target/off-tumor toxicity and improve cell therapy. security.
发明内容Contents of the invention
本发明的目的是提供一种新型的嵌合抗原受体T细胞,既可同时识别多个抗原,又可增强靶向特异性,发挥抗肿瘤功效,并且降低在靶/脱肿瘤(on target/off tumor)毒性,提高细胞治疗的安全性。The purpose of the present invention is to provide a new type of chimeric antigen receptor T cell that can recognize multiple antigens at the same time, enhance targeting specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor (on target/off-tumor) off tumor) toxicity and improve the safety of cell therapy.
本发明的第一方面,提供了一种工程化的免疫细胞,所述工程化的免疫细胞表达第一CAR和第二CAR,所述第一CAR靶向第一肿瘤细胞标志物,所述第二CAR靶向第二肿瘤细胞标志物,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。A first aspect of the present invention provides an engineered immune cell, the engineered immune cell expresses a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the third Two CARs target a second tumor cell marker, and the first tumor cell marker is selected from the following group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof; the second tumor cell The marker is selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof.
在另一优选例中,所述免疫细胞含有第一CAR和第二CAR。In another preferred embodiment, the immune cell contains a first CAR and a second CAR.
在另一优选例中,所述免疫细胞为NK细胞、巨噬细胞或T细胞,较佳地为T细胞。In another preferred embodiment, the immune cells are NK cells, macrophages or T cells, preferably T cells.
在另一优选例中,所述第一CAR、第二CAR定位于所述免疫细胞的细胞膜。In another preferred embodiment, the first CAR and the second CAR are located on the cell membrane of the immune cell.
在另一优选例中,所述第一CAR、第二CAR含有靶向肿瘤细胞标志物的抗原结合结构域。In another preferred embodiment, the first CAR and the second CAR contain antigen-binding domains targeting tumor cell markers.
在另一优选例中,所述抗原结合结构域为抗体或抗原结合片段。In another preferred embodiment, the antigen-binding domain is an antibody or antigen-binding fragment.
在另一优选例中,所述抗原结合片段是Fab或scFv或单结构域抗体sdFv。 In another preferred embodiment, the antigen-binding fragment is Fab or scFv or single domain antibody sdFv.
在另一优选例中,所述第一CAR的结构如式I所示:
L1-S1-H1-TM1-C1-CD3ζ   (I)
In another preferred example, the structure of the first CAR is shown in Formula I:
L1-S1-H1-TM1-C1-CD3ζ (I)
式中,所述“-”为连接肽或肽键;In the formula, the "-" is a connecting peptide or peptide bond;
L1为无或第一信号肽序列;L1 is none or the first signal peptide sequence;
S1为靶向第一肿瘤细胞标志物的抗原结合结构域,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;S1 is an antigen-binding domain targeting a first tumor cell marker selected from the group consisting of: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof;
H1为无或第一铰链区;H1 is none or the first hinge area;
TM1为第一跨膜结构域;TM1 is the first transmembrane domain;
C1为无或第一共刺激信号分子;C1 is no or the first costimulatory signal molecule;
CD3ζ为源于CD3ζ的胞浆信号传导序列。CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ.
在另一优选例中,所述靶向第一肿瘤细胞标志物的抗原结合结构域包括靶向肿瘤细胞标志物的抗体单链可变区序列。In another preferred embodiment, the antigen-binding domain targeting the first tumor cell marker includes an antibody single-chain variable region sequence targeting the tumor cell marker.
在另一优选例中,所述的靶向第一肿瘤细胞标志物的抗体单链可变区序列的结构如式A1或A2所示:
VH1-VL1 (A1);或
VL1-VH1 (A2);
In another preferred embodiment, the structure of the single-chain variable region sequence of the antibody targeting the first tumor cell marker is shown in Formula A1 or A2:
V H1 -V L1 (A1); or
V L1 -V H1 (A2);
其中,VL1为抗第一肿瘤细胞标志物抗体的轻链可变区;VH1为抗第一肿瘤细胞标志物抗体的重链可变区;“-”为连接肽(或柔性接头)或肽键。Wherein, V L1 is the light chain variable region of the anti-first tumor cell marker antibody; V H1 is the heavy chain variable region of the anti-first tumor cell marker antibody; "-" is the connecting peptide (or flexible linker) or Peptide bonds.
在另一优选例中,所述的VL1和VH1通过柔性接头相连。In another preferred embodiment, the V L1 and V H1 are connected through a flexible joint.
在另一优选例中,所述的柔性接头为1-5个(较佳地,2-4个)连续的GGGGS所示的序列。In another preferred embodiment, the flexible linker is 1-5 (preferably, 2-4) consecutive sequences represented by GGGGS.
在另一优选例中,所述柔性接头的氨基酸序列如SEQ ID NO.:1的第120-134位所示。In another preferred embodiment, the amino acid sequence of the flexible linker is shown in positions 120-134 of SEQ ID NO.: 1.
在另一优选例中,VL1的氨基酸序列如SEQ ID NO.:1中第135-247位所示,且VH1的氨基酸序列如SEQ ID NO.:1的第1-119位所示。In another preferred embodiment, the amino acid sequence of V L1 is shown at positions 135-247 of SEQ ID NO.: 1, and the amino acid sequence of V H1 is shown at positions 1-119 of SEQ ID NO.: 1.
在另一优选例中,所述的靶向第一肿瘤细胞标志物的抗体单链可变区序列如SEQ ID NO.:1所示。In another preferred embodiment, the single-chain variable region sequence of the antibody targeting the first tumor cell marker is shown in SEQ ID NO.: 1.
在另一优选例中,所述第一CAR的氨基酸序列如SEQ ID NO.:2-4中任一所示。In another preferred embodiment, the amino acid sequence of the first CAR is as shown in any one of SEQ ID NO.: 2-4.
在另一优选例中,所述第二CAR的结构如式II所示:
L2-S2-H2-TM2-C2-Z2  (II)
In another preferred example, the structure of the second CAR is shown in Formula II:
L2-S2-H2-TM2-C2-Z2 (II)
式中,所述“-”为连接肽或肽键; In the formula, the "-" is a connecting peptide or peptide bond;
L2为无或第二信号肽序列;L2 is none or the second signal peptide sequence;
S2为靶向第二肿瘤细胞标志物的抗原结合结构域,所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合;S2 is an antigen-binding domain targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof;
H2为无或第二铰链区;H2 is no or second hinge region;
TM2为第二跨膜结构域;TM2 is the second transmembrane domain;
C2为第二共刺激信号分子;C2 is the second costimulatory signal molecule;
Z2为无或源于CD3ζ的胞浆信号传导序列。Z2 is a cytoplasmic signaling sequence that is absent or derived from CD3ζ.
在另一优选例中,所述靶向第二肿瘤细胞标志物的抗原结合结构域包括靶向肿瘤细胞标志物的抗体单链可变区序列。In another preferred embodiment, the antigen-binding domain targeting the second tumor cell marker includes an antibody single-chain variable region sequence targeting the tumor cell marker.
在另一优选例中,所述的靶向第二肿瘤细胞标志物的抗体单链可变区序列的结构如式A3或A4所示:
VH2-VL2 (A3);或
VL2-VH2  (A4);
In another preferred embodiment, the structure of the single-chain variable region sequence of the antibody targeting the second tumor cell marker is shown in Formula A3 or A4:
V H2 -V L2 (A3); or
V L2 -V H2 (A4);
其中,VL2为抗第二肿瘤细胞标志物抗体的轻链可变区;VH2为抗第二肿瘤细胞标志物抗体的重链可变区;“-”为连接肽(或柔性接头)或肽键。Wherein, V L2 is the light chain variable region of the anti-second tumor cell marker antibody; V H2 is the heavy chain variable region of the anti-second tumor cell marker antibody; "-" is the connecting peptide (or flexible linker) or Peptide bonds.
在另一优选例中,所述的VL2和VH2通过柔性接头相连。In another preferred embodiment, the V L2 and V H2 are connected through a flexible joint.
在另一优选例中,所述的柔性接头为1-5个(较佳地,2-4个)连续的GGGGS所示的序列。In another preferred embodiment, the flexible linker is 1-5 (preferably, 2-4) consecutive sequences represented by GGGGS.
在另一优选例中,所述柔性接头的氨基酸序列如SEQ ID NO.:5中第120-134位所示。In another preferred embodiment, the amino acid sequence of the flexible linker is as shown at positions 120-134 in SEQ ID NO.: 5.
在另一优选例中,所述柔性接头的氨基酸序列如SEQ ID NO.:6中第120-134位所示。In another preferred embodiment, the amino acid sequence of the flexible linker is as shown at positions 120-134 in SEQ ID NO.: 6.
在另一优选例中,VL2的氨基酸序列如SEQ ID NO.:5中第135-241位所示,且VH2的氨基酸序列如SEQ ID NO.:5的第1-119位所示。In another preferred embodiment, the amino acid sequence of V L2 is shown in positions 135-241 of SEQ ID NO.:5, and the amino acid sequence of V H2 is shown in positions 1-119 of SEQ ID NO.:5.
在另一优选例中,VL2的氨基酸序列如SEQ ID NO.:6中第135-241位所示,且VH2的氨基酸序列如SEQ ID NO.:6的第1-119位所示。In another preferred embodiment, the amino acid sequence of V L2 is shown at positions 135-241 of SEQ ID NO.:6, and the amino acid sequence of V H2 is shown at positions 1-119 of SEQ ID NO.:6.
在另一优选例中,所述的靶向第二肿瘤细胞标志物的抗体单链可变区序列如SEQ ID NO.:5或6所示。In another preferred embodiment, the single-chain variable region sequence of the antibody targeting the second tumor cell marker is shown in SEQ ID NO.: 5 or 6.
在另一优选例中,所述第二CAR的氨基酸序列如SEQ ID NO.:7-14中任一所示。In another preferred embodiment, the amino acid sequence of the second CAR is as shown in any one of SEQ ID NO.: 7-14.
在另一优选例中,所述的靶向第一或第二肿瘤细胞标志物的抗体单链可变区 序列为鼠源、人源、人源和鼠源嵌合、或者全人源化的单链抗体可变区片段。In another preferred embodiment, the single-chain variable region of the antibody targeting the first or second tumor cell marker The sequences are murine, human, human and murine chimeric, or fully humanized single-chain antibody variable region fragments.
在另一优选例中,所述L1、L2各自独立地为选自下组的蛋白的信号肽:CD8a、CD8、CD28、GM-CSF、CD4、CD137、或其组合。In another preferred embodiment, each of L1 and L2 is independently a signal peptide selected from the following group of proteins: CD8a, CD8, CD28, GM-CSF, CD4, CD137, or a combination thereof.
在另一优选例中,所述L1为GM-CSF来源的信号肽。In another preferred embodiment, the L1 is a signal peptide derived from GM-CSF.
在另一优选例中,所述L2为CD8a来源的信号肽。In another preferred example, the L2 is a signal peptide derived from CD8a.
在另一优选例中,所述L1、L2的氨基酸序列各自独立地如SEQ ID NO.:15和16所示。In another preferred embodiment, the amino acid sequences of L1 and L2 are independently as shown in SEQ ID NO.: 15 and 16.
在另一优选例中,所述H1、H2各自独立地为选自下组的蛋白的铰链区:CD8、Ig(免疫球蛋白)铰链、或其组合。In another preferred embodiment, the H1 and H2 are each independently a hinge region of a protein selected from the following group: CD8, Ig (immunoglobulin) hinge, or a combination thereof.
在另一优选例中,所述H1、H2各自独立地为CD8来源的铰链区。In another preferred example, H1 and H2 are each independently a hinge region derived from CD8.
在另一优选例中,所述H1、H2的氨基酸序列各自独立地如SEQ ID NO.:17所示。In another preferred embodiment, the amino acid sequences of H1 and H2 are independently as shown in SEQ ID NO.: 17.
在另一优选例中,所述TM1、TM2各自独立地为选自下组的蛋白的跨膜区:CD8、CD28、CD8a、CD33、CD37、CD8α、CD5、CD16、ICOS、CD9、CD22、CD134、CD137、CD154、CD19、CD45、CD4、CD3ε、或其组合。In another preferred example, the TM1 and TM2 are each independently a transmembrane region of a protein selected from the following group: CD8, CD28, CD8a, CD33, CD37, CD8α, CD5, CD16, ICOS, CD9, CD22, CD134 , CD137, CD154, CD19, CD45, CD4, CD3ε, or combinations thereof.
在另一优选例中,所述TM1、TM2为CD8来源的跨膜区。In another preferred example, the TM1 and TM2 are transmembrane regions derived from CD8.
在另一优选例中,所述TM2为CD28来源的跨膜区。In another preferred example, the TM2 is a transmembrane region derived from CD28.
在另一优选例中,所述TM1、TM2的氨基酸序列如SEQ ID NO.:18所示。In another preferred example, the amino acid sequences of TM1 and TM2 are shown in SEQ ID NO.: 18.
在另一优选例中,所述TM2的氨基酸序列如SEQ ID NO.:19所示。In another preferred embodiment, the amino acid sequence of TM2 is shown in SEQ ID NO.: 19.
在另一优选例中,所述C1、C2各自独立地为选自下组的蛋白的共刺激信号分子:CD28、4-1BB(CD137)、CD30、CD40、CD70、CD134、LIGHT、DAP10、CDS、ICAM-1、OX40、或其组合。In another preferred example, the C1 and C2 are each independently a costimulatory signal molecule selected from the following group of proteins: CD28, 4-1BB (CD137), CD30, CD40, CD70, CD134, LIGHT, DAP10, CDS , ICAM-1, OX40, or combinations thereof.
在另一优选例中,所述C1为无;或CD28和/或4-1BB来源的共刺激信号分子。In another preferred example, the C1 is none; or a costimulatory signal molecule derived from CD28 and/or 4-1BB.
在另一优选例中,所述C2为CD28和/或4-1BB来源的共刺激信号分子。In another preferred example, the C2 is a costimulatory signal molecule derived from CD28 and/or 4-1BB.
在另一优选例中,所述C1、C2的氨基酸序列各自独立地如SEQ ID NO.:20所示。In another preferred embodiment, the amino acid sequences of C1 and C2 are independently as shown in SEQ ID NO.: 20.
在另一优选例中,所述C1、C2的氨基酸序列各自独立地如SEQ ID NO.:21所示。In another preferred embodiment, the amino acid sequences of C1 and C2 are independently as shown in SEQ ID NO.: 21.
在另一优选例中,所述CD3ζ的氨基酸序列如SEQ ID NO.:22所示。In another preferred example, the amino acid sequence of CD3ζ is shown in SEQ ID NO.: 22.
本发明第二方面提供了一种制备本发明第一方面所述的工程化的免疫细胞的方法,包括以下步骤: A second aspect of the present invention provides a method for preparing the engineered immune cells described in the first aspect of the present invention, comprising the following steps:
(A)提供一待改造的免疫细胞;和(A) providing an immune cell to be modified; and
(B)对所述的免疫细胞进行改造,从而使得所述的免疫细胞表达第一CAR和第二CAR,所述第一CAR靶向第一肿瘤细胞标志物,所述第二CAR靶向第二肿瘤细胞标志物,从而获得本发明第一方面所述的工程化的免疫细胞。(B) Modify the immune cells so that the immune cells express a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the second CAR targets a first tumor cell marker. Two tumor cell markers, thereby obtaining the engineered immune cells described in the first aspect of the present invention.
在另一优选例中,在步骤(A)中,还包括分离和/或激活待改造的免疫细胞。In another preferred embodiment, step (A) also includes isolating and/or activating immune cells to be modified.
在另一优选例中,在步骤(B)中,包括(B1)将表达靶向所述第一肿瘤细胞标志物的第一CAR的第一表达盒导入所述免疫细胞;和(B2)将表达靶向第二肿瘤细胞标志物的第二CAR的第二表达盒导入所述免疫细胞;其中所述的步骤(B1)可在步骤(B2)之前、之后、同时、或交替进行。In another preferred embodiment, step (B) includes (B1) introducing a first expression cassette expressing a first CAR targeting the first tumor cell marker into the immune cell; and (B2) introducing A second expression cassette expressing a second CAR targeting a second tumor cell marker is introduced into the immune cell; wherein step (B1) can be performed before, after, simultaneously, or alternately with step (B2).
在另一优选例中,在步骤(B)中,将所述第一表达盒和/或第二表达盒导入所述免疫细胞的细胞核中。In another preferred embodiment, in step (B), the first expression cassette and/or the second expression cassette is introduced into the nucleus of the immune cell.
在另一优选例中,当步骤(A)中的待改造的免疫细胞已经表达所述第一CAR和第二CAR时,则步骤(B)可以省略。In another preferred embodiment, when the immune cells to be modified in step (A) already express the first CAR and the second CAR, step (B) can be omitted.
在另一优选例中,所述免疫细胞为NK细胞、巨噬细胞或T细胞。In another preferred embodiment, the immune cells are NK cells, macrophages or T cells.
在另一优选例中,所述的第一表达盒含有编码所述的第一CAR的核酸序列。In another preferred embodiment, the first expression cassette contains a nucleic acid sequence encoding the first CAR.
在另一优选例中,所述的第二表达盒含有编码所述第二CAR的核酸序列。In another preferred embodiment, the second expression cassette contains a nucleic acid sequence encoding the second CAR.
在另一优选例中,所述的第一表达盒、第二表达盒位于相同或不同的载体上。In another preferred embodiment, the first expression cassette and the second expression cassette are located on the same or different vectors.
在另一优选例中,所述的第一表达盒、第二表达盒位于同一载体。In another preferred embodiment, the first expression cassette and the second expression cassette are located in the same vector.
在另一优选例中,所述的第一表达盒、第二表达盒位于不同载体。In another preferred embodiment, the first expression cassette and the second expression cassette are located in different vectors.
在另一优选例中,所述的载体为病毒载体。In another preferred embodiment, the vector is a viral vector.
在另一优选例中,所述的载体选自下组:DNA、RNA、质粒、慢病毒载体、腺病毒载体、逆转录病毒载体、转座子、其他基因转移系统、或其组合。In another preferred embodiment, the vector is selected from the following group: DNA, RNA, plasmid, lentiviral vector, adenoviral vector, retroviral vector, transposon, other gene transfer systems, or combinations thereof.
在另一优选例中,所述的载体为逆转录病毒载体。In another preferred embodiment, the vector is a retroviral vector.
在另一优选例中,所述的方法还包括对获得的工程化免疫细胞进行功能和有效性检测的步骤。In another preferred embodiment, the method further includes the step of testing the function and effectiveness of the obtained engineered immune cells.
本发明第三方面提供了一种制剂,所述制剂含有本发明第一方面所述的工程化的免疫细胞,以及药学上可接受的载体、稀释剂或赋形剂。A third aspect of the present invention provides a preparation, which contains the engineered immune cells described in the first aspect of the present invention, and a pharmaceutically acceptable carrier, diluent or excipient.
在另一优选例中,所述制剂为液态制剂。In another preferred embodiment, the preparation is a liquid preparation.
在另一优选例中,所述制剂的剂型包括注射剂。In another preferred embodiment, the dosage form of the preparation includes injection.
在另一优选例中,所述制剂中所述工程化的免疫细胞的浓度为1×103-1×108个细胞/ml,较佳地1×104-1×107个细胞/ml。 In another preferred embodiment, 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.
在另一优选例中,所述制剂还含有治疗癌症或肿瘤的其他药物(如新兴的抗体药物、其他CAR-T药物或化疗药物)。In another preferred embodiment, the preparation also contains other drugs for treating cancer or tumors (such as emerging antibody drugs, other CAR-T drugs or chemotherapy drugs).
本发明第四方面提供了一种如本发明第一方面所述的工程化的免疫细胞的用途,用于制备选择性杀伤肿瘤的药物或制剂。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 for preparing drugs or preparations that selectively kill tumors.
在另一优选例中,所述肿瘤包括高表达肿瘤细胞标志物(比如EGFR、cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、EpCAM)的肿瘤。In another preferred embodiment, the tumors include tumors that highly express tumor cell markers (such as EGFR, cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, EpCAM).
在另一优选例中,所述肿瘤包括同时表达肿瘤细胞标志物(比如EGFR、cMet、Her2、HER3)的肿瘤。In another preferred embodiment, the tumors include tumors that simultaneously express tumor cell markers (such as EGFR, cMet, Her2, and HER3).
在另一优选例中,所述肿瘤包括同时表达EGFR和cMet的肿瘤。In another preferred embodiment, the tumor includes a tumor that expresses both EGFR and cMet.
在另一优选例中,所述肿瘤选自下组:血液肿瘤、实体瘤、或其组合,优选地,所述肿瘤为实体瘤。In another preferred embodiment, the tumor is selected from the group consisting of hematological tumors, solid tumors, or combinations thereof. Preferably, the tumor is a solid tumor.
在另一优选例中,所述血液肿瘤选自下组:急性骨髓性白血病、急性淋巴细胞白血病、急性单核细胞白血病、急性粒细胞白血病、急性粒-单核细胞白血病、慢性淋巴细胞白血病、慢性粒细胞白血病、慢性骨髓性白血病、淋巴瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、多发性骨髓瘤(MM)、骨髓增生异常综合征、或其组合。In another preferred embodiment, the blood tumor is selected from the following group: acute myelogenous leukemia, acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodysplastic syndrome, or combinations thereof.
在另一优选例中,所述肿瘤包括实体瘤。In another preferred embodiment, the tumor includes a solid tumor.
在另一优选例中,所述实体瘤选自下组:前列腺癌、肝癌、头颈癌、黑色素瘤、非霍奇金淋巴瘤,膀胱癌、胶质母细胞瘤、宫颈癌、肺癌、软骨肉瘤、甲状腺癌、肾癌、间皮瘤、骨肉瘤、胆管癌、卵巢癌、胃癌、膀胱癌、脑膜瘤、胰腺癌、多发性鳞状细胞瘤、食管癌、肺小细胞癌、结直肠癌、乳腺癌、成神经管细胞瘤、乳腺癌、鼻咽癌、胸腺癌、或其组合。In another preferred embodiment, the solid tumor is selected from the group consisting of prostate cancer, liver cancer, head and neck cancer, melanoma, non-Hodgkin lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, and chondrosarcoma. , Thyroid cancer, kidney cancer, mesothelioma, osteosarcoma, cholangiocarcinoma, ovarian cancer, gastric cancer, bladder cancer, meningioma, pancreatic cancer, multiple squamous cell tumor, esophageal cancer, small cell lung cancer, colorectal cancer, Breast cancer, medulloblastoma, breast cancer, nasopharyngeal cancer, thymic cancer, or combinations thereof.
本发明第五方面提供了一种用于选择性杀伤肿瘤的试剂盒,所述试剂盒含有容器,以及位于容器内的:The fifth aspect of the present invention provides a kit for selectively killing tumors. The kit contains a container, and located in the container:
(1)第一核酸序列,所述第一核酸序列含有用于表达靶向第一肿瘤细胞标志物的第一CAR的第一表达盒,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;和(1) A first nucleic acid sequence containing a first expression cassette for expressing a first CAR targeting a first tumor cell marker selected from the group consisting of: cMet , Her2, Her3, Muc1, ROR1, PD-L1, CD47, or combinations thereof; and
(2)第二核酸序列,所述第二核酸序列含有靶向第二肿瘤细胞标志物的第二CAR的第二表达盒,所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。(2) A second nucleic acid sequence containing a second expression cassette of a second CAR targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or combinations thereof.
在另一优选例中,所述的第一、第二核酸序列为独立的或相连的。In another preferred embodiment, the first and second nucleic acid sequences are independent or connected.
在另一优选例中,所述的第一、第二核酸序列位于相同或不同的容器内。 In another preferred embodiment, the first and second nucleic acid sequences are located in the same or different containers.
在另一优选例中,所述的第一、第二核酸序列位于相同或不同的载体上。In another preferred embodiment, the first and second nucleic acid sequences are located on the same or different vectors.
在另一优选例中,所述的第一、第二核酸序列位于同一载体。In another preferred embodiment, the first and second nucleic acid sequences are located in the same vector.
本发明第六方面提供了一种选择性杀伤肿瘤的方法,包括:The sixth aspect of the present invention provides a method for selectively killing tumors, including:
给需要治疗的对象施用安全有效量的本发明第一方面所述的工程化免疫细胞、或本发明第三方面所述的制剂。A safe and effective amount of the engineered immune cells described in the first aspect of the present invention or the preparation described in the third aspect of the present invention is administered to the subject in need of treatment.
在另一优选例中,所述对象包括人或非人哺乳动物。In another preferred embodiment, the subject includes humans or non-human mammals.
在另一优选例中,所述非人哺乳动物包括啮齿动物(如小鼠、大鼠、兔)、灵长类动物(如猴)。In another preferred embodiment, the non-human mammals include rodents (such as mice, rats, rabbits) and primates (such as monkeys).
在另一优选例中,所述方法为非治疗性和非诊断性的。In another preferred embodiment, the method is non-therapeutic and non-diagnostic.
本发明第七方面提供了一种治疗疾病的方法,包括给需要治疗的对象施用安全有效量的本发明第一方面所述的工程化免疫细胞、或本发明第三方面所述的制剂。The seventh aspect of the present invention provides a method for treating diseases, which includes administering a safe and effective amount of the engineered immune cells described in the first aspect of the present invention or the preparation described in the third aspect of the present invention to a subject in need of treatment.
在另一优选例中,所述方法还包括给需要治疗的对象施用治疗癌症或肿瘤的其他药物。In another preferred embodiment, the method further includes administering other drugs for treating cancer or tumors to the subject in need of treatment.
在另一优选例中,所述其他药物包括CAR-T药物。In another preferred embodiment, the other drugs include CAR-T drugs.
在另一优选例中,所述疾病为癌症或肿瘤。In another preferred embodiment, the disease is cancer or tumor.
在另一优选例中,所述肿瘤包括高表达肿瘤细胞标志物(比如EGFR、cMet、HER2、HER3、MUC1、ROR1、PD-L1、CD47)的肿瘤。In another preferred embodiment, the tumors include tumors that highly express tumor cell markers (such as EGFR, cMet, HER2, HER3, MUCl, ROR1, PD-L1, CD47).
在另一优选例中,所述肿瘤包括同时表达肿瘤细胞标志物(比如EGFR、cMet、EpCAM、HER2、HER3)的肿瘤。In another preferred embodiment, the tumors include tumors that simultaneously express tumor cell markers (such as EGFR, cMet, EpCAM, HER2, HER3).
在另一优选例中,所述肿瘤包括同时表达EGFR和cMet的肿瘤。In another preferred embodiment, the tumor includes a tumor that expresses both EGFR and cMet.
在另一优选例中,所述肿瘤选自下组:血液肿瘤、实体瘤、或其组合,优选地,所述肿瘤为实体瘤。In another preferred embodiment, the tumor is selected from the following group: hematological tumors, solid tumors, or combinations thereof. Preferably, the tumor is a solid tumor.
在另一优选例中,所述血液肿瘤选自下组:急性骨髓性白血病、急性淋巴细胞白血病、急性单核细胞白血病、急性粒细胞白血病、急性粒-单核细胞白血病、慢性淋巴细胞白血病、慢性粒细胞白血病、慢性骨髓性白血病、淋巴瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、多发性骨髓瘤(MM)、骨髓增生异常综合征、或其组合。In another preferred embodiment, the blood tumor is selected from the following group: acute myelogenous leukemia, acute lymphoblastic leukemia, acute monocytic leukemia, acute myeloid leukemia, acute myeloid-monocytic leukemia, chronic lymphocytic leukemia, Chronic myeloid leukemia, chronic myelogenous leukemia, lymphoma, Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma (MM), myelodysplastic syndrome, or combinations thereof.
在另一优选例中,所述肿瘤包括实体瘤。In another preferred embodiment, the tumor includes a solid tumor.
在另一优选例中,所述实体瘤选自下组:前列腺癌、肝癌、头颈癌、黑色素瘤、非霍奇金淋巴瘤,膀胱癌、胶质母细胞瘤、宫颈癌、肺癌、软骨肉瘤、甲状腺癌、肾癌、间皮瘤、骨肉瘤、胆管癌、卵巢癌、胃癌、膀胱癌、脑膜瘤、胰腺癌、多发 性鳞状细胞瘤、食管癌、肺小细胞癌、结直肠癌、乳腺癌、成神经管细胞瘤、乳腺癌、鼻咽癌、胸腺癌、或其组合。In another preferred embodiment, the solid tumor is selected from the group consisting of prostate cancer, liver cancer, head and neck cancer, melanoma, non-Hodgkin lymphoma, bladder cancer, glioblastoma, cervical cancer, lung cancer, and 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, nasopharyngeal cancer, thymic cancer, or combinations thereof.
本发明第八方面提供了一种融合蛋白,所述融合蛋白包含靶向第一肿瘤细胞标志物的第一CAR和靶向第二肿瘤细胞标志物的第二CAR,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。An eighth aspect of the present invention provides a fusion protein, the fusion protein comprising a first CAR targeting a first tumor cell marker and a second CAR targeting a second tumor cell marker, the first tumor cell marker The material is selected from the following group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof; the second tumor cell marker is selected from the following group: EGFR, EpCAM, Her2, Her3, or a combination thereof .
在另一优选例中,所述第一CAR和第二CAR通过连接肽连接。In another preferred embodiment, the first CAR and the second CAR are connected through a connecting peptide.
在另一优选例中,所述连接肽包括自剪切蛋白。In another preferred embodiment, the connecting peptide includes a self-cleaving protein.
在另一优选例中,所述自剪切蛋白选自下组:T2A、P2A、E2A、F2A、或其组合。In another preferred embodiment, the self-cleaving protein is selected from the following group: T2A, P2A, E2A, F2A, or a combination thereof.
在另一优选例中,所述自剪切蛋白包括P2A。In another preferred embodiment, the self-cleaving protein includes P2A.
在另一优选例中,所述融合蛋白的结构如下式III所示:
L1-S1-H1-TM1-C1-CD3ζ-Z3-L2-S2-H2-TM2-C2-Z2 (III)
In another preferred example, the structure of the fusion protein is shown in the following formula III:
L1-S1-H1-TM1-C1-CD3ζ-Z3-L2-S2-H2-TM2-C2-Z2 (III)
式中,In the formula,
各“-”独立地为连接肽或肽键;Each "-" is independently a connecting peptide or peptide bond;
L1为无或第一信号肽序列;L1 is none or the first signal peptide sequence;
L2为无或第二信号肽序列;L2 is none or the second signal peptide sequence;
S1为靶向第一肿瘤细胞标志物的抗原结合结构域,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;S1 is an antigen-binding domain targeting a first tumor cell marker selected from the group consisting of: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof;
S2为靶向第二肿瘤细胞标志物的抗原结合结构域,所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合;S2 is an antigen-binding domain targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof;
H1为无或第一铰链区;H1 is none or the first hinge area;
H2为无或第二绞链区;H2 is no or second hinge region;
TM1为第一跨膜结构域;TM1 is the first transmembrane domain;
TM2为第二跨膜结构域;TM2 is the second transmembrane domain;
C1为无或第一共刺激信号分子;C1 is no or the first costimulatory signal molecule;
C2为第二共刺激信号分子;C2 is the second costimulatory signal molecule;
CD3ζ为源于CD3ζ的胞浆信号传导序列;CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ;
Z2为无或源于CD3ζ的胞浆信号传导序列;Z2 is a cytoplasmic signaling sequence that is absent or derived from CD3ζ;
Z3为连接肽。Z3 is the connecting peptide.
在另一优选例中,所述融合蛋白的氨基酸序列如SEQ ID NO.:23-26中任一所示。 In another preferred embodiment, the amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
本发明第九方面提供了一种多核苷酸,所述多核苷酸编码本发明第八方面所述的融合蛋白。The ninth aspect of the present invention provides a polynucleotide encoding the fusion protein according to the eighth aspect of the present invention.
在另一优选例中,所述多核苷酸选自下组:In another preferred embodiment, the polynucleotide is selected from the following group:
(a)编码如SEQ ID NO.:23-26任一所示融合蛋白的多核苷酸;(a) A polynucleotide encoding a fusion protein as shown in any one of SEQ ID NO.: 23-26;
(b)序列如SEQ ID NO.:27-30任一所示的多核苷酸;(b) A polynucleotide whose sequence is shown in any one of SEQ ID NO.: 27-30;
(c)核苷酸序列与(b)所示序列的同源性≥75%(较佳地≥80%)的多核苷酸;(c) A polynucleotide whose nucleotide sequence has a homology of ≥75% (preferably ≥80%) to the sequence shown in (b);
(d)如(b)所示多核苷酸的5’端和/或3’端截短或添加1-60个(较佳地1-30,更佳地1-10个)核苷酸的多核苷酸;(d) The 5' end and/or 3' end of the polynucleotide shown in (b) is truncated or 1-60 (preferably 1-30, more preferably 1-10) nucleotides are added polynucleotide;
(e)与(a)-(d)任一所述的多核苷酸互补的多核苷酸。(e) A polynucleotide complementary to the polynucleotide described in any one of (a) to (d).
在另一优选例中,所述的多核苷酸序列如SEQ ID NO.:27-30任一所示。In another preferred embodiment, the polynucleotide sequence is as shown in any one of SEQ ID NO.: 27-30.
本发明第十方面提供了一种载体,所述载体包括本发明第九方面所述的多核苷酸。A tenth aspect of the present invention provides a vector, which includes the polynucleotide described in the ninth aspect of the present invention.
在另一优选例中,所述的载体包括DNA、RNA。In another preferred embodiment, the vector includes DNA and RNA.
在另一优选例中,所述的载体选自下组:质粒、病毒载体、转座子、或其组合。In another preferred embodiment, the vector is selected from the following group: plasmids, viral vectors, transposons, or combinations thereof.
在另一优选例中,所述的载体包括DNA病毒、逆转录病毒载体。In another preferred embodiment, the vector includes DNA virus and retroviral vector.
在另一优选例中,所述的载体选自下组:慢病毒载体、逆转录病毒载体、腺病毒载体、腺相关病毒载体、或其组合。In another preferred embodiment, the vector is selected from the following group: lentiviral vectors, retroviral vectors, adenoviral vectors, adeno-associated virus vectors, or combinations thereof.
在另一优选例中,所述载体为逆转录病毒载体。In another preferred embodiment, the vector is a retroviral vector.
在另一优选例中,所述载体包含一个或多个启动子,所述启动子可操作地与所述核酸序列、增强子、内含子、转录终止信号、多腺苷酸化序列、复制起点、选择性标记、核酸限制性位点、和/或同源重组位点连接。In another preferred embodiment, the vector contains one or more promoters operably linked to the nucleic acid sequence, enhancer, intron, transcription termination signal, polyadenylation sequence, origin of replication , selectable markers, nucleic acid restriction sites, and/or homologous recombination site ligation.
在另一优选例中,所述载体为含有或插入有本发明第九方面所述的多核苷酸的载体。In another preferred embodiment, the vector is a vector containing or inserted with the polynucleotide described in the ninth aspect of the present invention.
在另一优选例中,所述载体用于表达本发明第八方面所述的融合蛋白。In another preferred embodiment, the vector is used to express the fusion protein according to the eighth aspect of the present invention.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described below (such as embodiments) can be combined with each other to form new or preferred technical solutions. Due to space limitations, they will not be described one by one here.
附图说明Description of the drawings
图1显示了靶向EGFR和cMet双特异性“和”逻辑CAR(And logic BiCAR) 的组成,包括2个独立的CAR分子,CAR1分子为抗cMet的scFv串联CD3ζ激活信号结构域,CAR2分子为抗EGFR的scFv串联CD28或者4-1BB共刺激信号结构域;图1B示出“和”逻辑BiCAR表达的分子构建示意图,CAR1分子和CAR2分子通过P2A连接;图1C示出了靶向cMet的第二代全功能CAR的构建图,抗cMET的scFv串联CD28或者4-1BB共刺激信号结构域及CD3ζ激活信号结构域。Figure 1 shows the And logic BiCAR targeting EGFR and cMet. The composition includes two independent CAR molecules, the CAR1 molecule is an anti-cMet scFv tandem CD3ζ activation signal domain, and the CAR2 molecule is an anti-EGFR scFv tandem CD28 or 4-1BB costimulatory signal domain; Figure 1B shows " and "Schematic diagram of the molecular construction of logical BiCAR expression, CAR1 molecules and CAR2 molecules are connected through P2A; Figure 1C shows the construction diagram of the second generation fully functional CAR targeting cMet, anti-cMET scFv tandem CD28 or 4-1BB costimulatory signal domain and CD3ζ activation signaling domain.
图2显示了Mock T(2A)、O-28z(2B)CAR及“And”logic BiCAR T(2C)细胞的CAR转导效率流式细胞分析图。Figure 2 shows the flow cytometry analysis of CAR transduction efficiency of Mock T (2A), O-28z (2B) CAR and "And" logic BiCAR T (2C) cells.
图3显示了由不同健康donor#1(3A)和#2(3B)的外周血单核细胞(PBMC)所制备的Mock T、O-28z CAR及BiCAR T细胞,在体外以不同的效靶比分别与EGFR/cMet共表达的NCI-H1975人肺腺癌细胞株共孵育24h后,对靶细胞的裂解毒性。Figure 3 shows that Mock T, O-28z CAR and BiCAR T cells prepared from peripheral blood mononuclear cells (PBMC) of different healthy donors #1 (3A) and #2 (3B) have different effects on targets in vitro. Compared with the lytic toxicity to target cells after incubation with NCI-H1975 human lung adenocarcinoma cell line co-expressing EGFR/cMet for 24 hours.
图4显示了由不同健康donor#1(4A)和#2(4B)的外周血单核细胞(PBMC)所制备的Mock T,O-28z CAR及BiCAR T细胞以不同的效靶比分别与EGFR/cMet共表达的EBC-1人肺鳞癌细胞株体外共培养24h后,对靶细胞的裂解毒性。Figure 4 shows that Mock T, O-28z CAR and BiCAR T cells prepared from peripheral blood mononuclear cells (PBMC) of different healthy donors #1 (4A) and #2 (4B) were compared with each other at different effect-target ratios. The lytic toxicity of the EBC-1 human lung squamous cell carcinoma cell line co-expressing EGFR/cMet to target cells after 24 hours of co-culture in vitro.
图5显示了Mock T和BiCAR-T细胞在不同效靶比条件下与EGFR/cMET共表达的人胃癌细胞系SNU5细胞共孵育24h后,对靶细胞的裂解毒性。Figure 5 shows the lytic toxicity of Mock T and BiCAR-T cells to target cells after incubation for 24 hours with the human gastric cancer cell line SNU5 cells co-expressing EGFR/cMET under different effective-target ratio conditions.
图6显示了Mock T和BiCAR-T细胞在不同效靶比条件下,与对照293T-细胞,单表达cMET的293T稳转株,单表达EGFR的293T稳转株和共表达EGFR/cMet的293T稳转株共孵育24h后,对靶细胞的裂解毒性。Figure 6 shows the comparison of Mock T and BiCAR-T cells under different effective target ratio conditions with control 293T-cells, 293T stable transfection strain expressing cMET alone, 293T stably transducing strain expressing EGFR alone and 293T co-expressing EGFR/cMet. Lytic toxicity of the stably transformed strains to target cells after co-incubation for 24 hours.
图7显示了尾静脉注射CAR T细胞后,荷瘤免疫缺陷小鼠体内肿瘤生长大小的折线图,结果表明O-28z CAR及BiCAR T细胞显著杀伤甚至消灭了小鼠体内的肿瘤细胞,并且小鼠并未观察到任何异常。Figure 7 shows the line graph of tumor growth size in tumor-bearing immunodeficient mice after tail vein injection of CAR T cells. The results show that O-28z CAR and BiCAR T cells significantly killed or even eliminated tumor cells in the mice, and were small. No abnormalities were observed in the rats.
具体实施方式Detailed ways
本发明人经过广泛而深入的研究,首次意外地发现含有靶向第一肿瘤细胞标志物(比如cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47)的第一CAR和靶向第二肿瘤细胞标志物(比如,EGFR、EpCAM、Her2、Her3)的第二CAR的免疫细胞可被充分激活,增强靶向特异性,发挥抗肿瘤功效,并且降低在靶/脱肿瘤毒性,提高细胞治疗的安全性。在此基础上,发明人完成了本发明。After extensive and in-depth research, the inventor unexpectedly discovered for the first time a first CAR targeting a first tumor cell marker (such as cMet, Her2, Her3, Muc1, ROR1, PD-L1, CD47) and a second targeting tumor cell marker. The immune cells of the second CAR of tumor cell markers (such as EGFR, EpCAM, Her2, Her3) can be fully activated, enhance targeting specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor toxicity, improving cell therapy. security. On this basis, the inventor completed the present invention.
术语说明 Terminology
除非另外定义,否则本文中所用的全部技术与科学术语均具有如本发明所属领域的普通技术人员通常理解的相同含义。Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
如本文所用,在提到具体列举的数值中使用时,术语“约”意指该值可以从列举的值变动不多于1%。例如,如本文所用,表述“约100”包括99和101和之间的全部值(例如,99.1、99.2、99.3、99.4等)。As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes all values between 99 and 101 and between (eg, 99.1, 99.2, 99.3, 99.4, etc.).
如本文所用,术语“含有”或“包括(包含)”可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”。As used herein, the term "contains" or "includes" can be open, semi-closed and closed. In other words, the term also includes "consisting essentially of," or "consisting of."
如本文所用,“嵌合抗原受体(CAR)”是一种融合蛋白,其包含能够结合抗原的胞外结构域,与胞外结构域衍生自不同多肽的跨膜结构域,以及至少一个胞内结构域。“嵌合抗原受体(CAR)”也称为“嵌合受体”、“T-body”或“嵌合免疫受体(CIR)”。所述的“能够结合抗原的胞外结构域”是指能够结合某一抗原的任何寡肽或多肽。“胞内结构域”是指已知的作为传递信号以激活或抑制细胞内生物过程的结构域的任何寡肽或多肽。As used herein, a "chimeric antigen receptor (CAR)" is a fusion protein comprising an extracellular domain capable of binding an antigen, a transmembrane domain derived from a different polypeptide, and at least one cellular domain. inner domain. "Chimeric antigen receptor (CAR)" is also known as "chimeric receptor", "T-body" or "chimeric immune receptor (CIR)". The "extracellular domain capable of binding antigen" refers to any oligopeptide or polypeptide capable of binding a certain antigen. "Intracellular domain" refers to any oligopeptide or polypeptide known as a domain that transmits signals to activate or inhibit biological processes within a cell.
如本文所用,“结构域”是指多肽中独立于其它区域且折叠成特异结构的区域。As used herein, "domain" refers to a region of a polypeptide that is independent of other regions and folds into a specific structure.
如本文所用,术语“给予”和“处理”是指外源性药物、治疗剂、诊断剂或组合物应用于动物、人、受试者、细胞、组织、器官或生物流体。“给予”和“处理”可以指治疗、药物代谢动力学、诊断、研究和实验方法。细胞的处理包括试剂与细胞的接触、以及试剂与流体的接触、流体与细胞的接触。“给予”和“处理”还意指通过试剂、诊断、结合组合物或通过另一种细胞体外和离体处理。“处理”当应用于人、动物或研究受试者时,是指治疗处理、预防或预防性措施,研究和诊断。As used herein, the terms "administration" and "treatment" refer to the application of an exogenous drug, therapeutic agent, diagnostic agent or composition to an animal, human, subject, cell, tissue, organ or biological fluid. "Administration" and "treatment" may refer to therapeutic, pharmacokinetic, diagnostic, research and experimental methods. Treatment of cells includes contact of reagents with cells, contact of reagents with fluids, and contact of fluids with cells. "Administration" and "treatment" also mean in vitro and ex vivo treatment of cells by a reagent, a diagnostic, a binding composition or by another cell. "Treatment" when applied to humans, animals or research subjects, means therapeutic treatment, prophylactic or prophylactic measures, research and diagnostics.
如本文所用,术语“治疗”指给予患者内用或外用治疗剂,包含本发明的任何一种CAR及其组合物,所述患者具有一种或多种疾病症状,而已知所述治疗剂对这些症状具有治疗作用。通常,以有效缓解一种或多种疾病症状的治疗剂的量(治疗有效量)给予患者。As used herein, the term "treatment" refers to the administration of an internal or external therapeutic agent, including any CAR of the invention and compositions thereof, to a patient with one or more symptoms of a disease for which the therapeutic agent is known to be effective. These symptoms are therapeutic. Typically, a therapeutic agent is administered to a patient in an amount effective to alleviate one or more symptoms of the disease (a therapeutically effective amount).
如本文所用,术语“任选”或“任选地”意味着随后所描述的事件或情况可以发生但不是必须发生。例如,“任选包含1-3个抗体重链可变区”是指特定序列的抗体重链可变区可以有但不是必须有,可以是1个、2个或3个。As used herein, the terms "optionally" or "optionally" mean that the subsequently described event or circumstance may occur but does not necessarily occur. For example, "optionally including 1-3 antibody heavy chain variable regions" means that the antibody heavy chain variable regions of a specific sequence may be present but are not required to be present, and may be 1, 2 or 3.
本发明所述的“序列同一性”表示当具有适当的替换、插入或缺失等突变的情况下最佳比对和比较时,两个核酸或两个氨基酸序列之间的同一性程度。 本发明中所述的序列和其具有同一性的序列之间的序列同一性可以至少为85%、90%或95%,优选至少为95%。非限制性实施例包括85%,86%,87%,88%,89%,90%,91%,92%,93%,94%,95%,96%,97%,98%,99%,100%。"Sequence identity" as used herein means the degree of identity between two nucleic acids or two amino acid sequences when optimally aligned and compared with appropriate substitutions, insertions, deletions and other mutations. The 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%.
EGFREGFR
表皮因子生长受体(EGFR)属于酪氨酸激酶家族,家族包括其它三个成员ERBB2/HER2,ERBB3/HER3以及ERBB4/HER4。其锚定在细胞质膜中,包括胞外配体结合功能结构域,疏水跨膜区及胞内酪氨酸激酶结构域。EGFR的配体(例如EGF,TGF-a等)与其结合后,会形成同源二聚体或与其它家族成员形成异源二聚体,导致酪氨酸残基的自磷酸化,从而激活多个下游信号传导途径,调节细胞的增殖,存活和凋亡。EGFR信号网络在上皮组织的维持和生长中起着重要的作用,其异常激活、活跃与多种癌症的发生、进展和不良预后有关。Epidermal factor growth receptor (EGFR) belongs to the tyrosine kinase family, which includes three other members: ERBB2/HER2, ERBB3/HER3 and ERBB4/HER4. It is anchored in the cytoplasmic membrane and includes an extracellular ligand-binding functional domain, a hydrophobic transmembrane region and an intracellular tyrosine kinase domain. After ligands of EGFR (such as EGF, TGF-a, etc.) bind to it, they will form homodimers or heterodimers with other family members, leading to autophosphorylation of tyrosine residues, thereby activating multiple A downstream signaling pathway that regulates cell proliferation, survival and apoptosis. The EGFR signaling network plays an important role in the maintenance and growth of epithelial tissue, and its abnormal activation and activity are related to the occurrence, progression and poor prognosis of various cancers.
EGFR在多种肿瘤中过表达,包括25-77%的结直肠癌,80-100%的头颈癌,40-80%的非小细胞肺癌(NSCLC),50-90%的肾癌,30-50%的胰腺癌,14-91%的乳腺癌,40-63%的胶质母细胞瘤,35-70%的卵巢癌等。EGFR的基因突变和过度表达均异常激活下游通路,与致癌和癌症进展密切相关。目前针对EGFR靶点的药物,包括酪氨酸激酶抑制剂(TKI)(例如厄洛替尼,吉非替尼,奥西替尼等)和单克隆抗体(西妥昔单抗及帕尼单抗),已批准分别用于非小细胞肺癌,结直肠癌和头颈癌的治疗。EGFR也广泛地表达于人的正常器官和组织中,包括皮肤、肺、肝、肾脏等。获批的单抗药物在真实世界中的不良反应以及针对EGFR开发的CAR T细胞治疗药物临床上的在靶点毒性均主要表现为皮肤毒性。EGFR原癌基因驱动突变是NSCLC致癌、疾病进展的重要因素,在50-60%的亚洲患者中被检测到,临床上作为预后和疾病预测的生物标志物。EGFR-TKI药物(吉非替尼、厄洛替尼等)对于EGFR敏感突变的晚期NSCLC患者有很好的疗效,然而经过中位时间6-12个月的治疗后,大部分患者对EGFR-TKI产生耐药。耐药机制主要包括EGFR的获得性突变,约50%的耐药病人出现T790M突变,而三代TKI药物如奥西替尼的耐药是由于C797S突变;以及旁路途径cMet、Her2信号通路的激活。接受奥西替尼治疗的病人,约30%会出现获得性cMet基因扩增,从而避开靶点EGFR,导致TKI耐药。NSCLC是典型的多驱动基因突变,基因组不稳定性、肿瘤异质性以及疾病进展过程中产生的原发或获得性耐药对于晚期NSCLC的靶向治疗的应用产生了瓶颈,因此多靶点的靶向治疗将有 机会取得更好的临床疗效。EGFR is overexpressed in a variety of tumors, including 25-77% of colorectal cancers, 80-100% of head and neck cancers, 40-80% of non-small cell lung cancer (NSCLC), 50-90% of renal cancers, 30- 50% of pancreatic cancers, 14-91% of breast cancers, 40-63% of glioblastomas, 35-70% of ovarian cancers, etc. Gene mutations and overexpression of EGFR abnormally activate downstream pathways and are closely related to carcinogenesis and cancer progression. Current drugs targeting EGFR include tyrosine kinase inhibitors (TKIs) (such as erlotinib, gefitinib, osimertinib, etc.) and monoclonal antibodies (cetuximab and panitumumab). ), has been approved for the treatment of non-small cell lung cancer, colorectal cancer and head and neck cancer respectively. EGFR is also widely expressed in normal human organs and tissues, including skin, lungs, liver, kidneys, etc. The real-world adverse reactions of approved monoclonal antibody drugs and the clinical on-target toxicity of CAR T cell therapy drugs developed for EGFR are mainly manifested as skin toxicity. EGFR proto-oncogene driver mutations are an important factor in NSCLC carcinogenesis and disease progression. They are detected in 50-60% of Asian patients and are clinically used as biomarkers for prognosis and disease prediction. EGFR-TKI drugs (gefitinib, erlotinib, etc.) have good efficacy in patients with advanced NSCLC with EGFR-sensitive mutations. However, after a median treatment time of 6-12 months, most patients have no response to EGFR-TKI. TKI resistance develops. The resistance mechanism mainly includes acquired mutations of EGFR. About 50% of drug-resistant patients have the T790M mutation. The resistance to third-generation TKI drugs such as osimertinib is due to the C797S mutation; and the activation of the alternative pathway cMet and Her2 signaling pathways. About 30% of patients treated with osimertinib will develop acquired cMet gene amplification, thus avoiding the target EGFR, leading to TKI resistance. NSCLC is a typical case of multiple driver gene mutations. Genomic instability, tumor heterogeneity, and primary or acquired drug resistance generated during disease progression have created bottlenecks in the application of targeted therapy for advanced NSCLC. Therefore, multi-target Targeted therapy will have opportunity to achieve better clinical outcomes.
cMetcMet
间质表皮转化因子(cMet)是受体酪氨酸激酶的一种,与配体肝细胞因子(HGF)结合后,导致受体的二聚及磷酸化,激活多种不同的细胞信号通路,参与细胞的增殖、运动、迁移、侵袭和血管生成。cMet正常表达于人的多种组织和器官中,包括肝、食管、胃、结直肠、皮肤、卵巢及子宫内膜。cMet对于正常生理条件下控制细胞稳态非常重要,而当cMet被过度激活,则会启动正常细胞向肿瘤细胞转化,出现上皮-间质化。cMet的基因突变、基因扩增或过表达可导致其信号通路异常激活,促进肿瘤进展。Mesenchymal epidermal transformation factor (cMet) is a type of receptor tyrosine kinase. After binding to the ligand hepatocyte factor (HGF), it leads to dimerization and phosphorylation of the receptor, activating a variety of different cell signaling pathways. Involved in cell proliferation, movement, migration, invasion and angiogenesis. cMet is normally expressed in a variety of human tissues and organs, including liver, esophagus, stomach, colorectum, skin, ovary and endometrium. cMet is very important for controlling cell homeostasis under normal physiological conditions. When cMet is overactivated, it will initiate the transformation of normal cells into tumor cells, resulting in epithelial-mesenchymal transition. Gene mutation, gene amplification or overexpression of cMet can lead to abnormal activation of its signaling pathway and promote tumor progression.
cMet在多种癌症中过表达,包括肺癌,乳腺癌,卵巢癌,肾癌,结直肠癌,甲状腺癌,肝癌和胃癌。cMet的过度表达与较差的存活期直接相关,在一些癌症如肺癌、结直肠、卵巢癌等,cMet和/或其配体HGF作为临床预后指标。cMet基因异常的癌症中,典型的是胃癌和结直肠癌,其中胃癌约10-20%cMet基因拷贝数扩增,约25%cMet基因突变;结直肠癌病人中cMet突变率为约15%。cMet过表达于25%-75%的NSCLC。EGFR-TKI耐药的NSCLC中,15%-30%出现cMet基因获得性扩增,cMet也作为EGFR突变的NSCLC病人中TKI耐药的检测标志物。cMet is overexpressed in a variety of cancers, including lung, breast, ovarian, renal, colorectal, thyroid, liver, and gastric cancers. Overexpression of cMet is directly related to poor survival. In some cancers, such as lung cancer, colorectal cancer, ovarian cancer, etc., cMet and/or its ligand HGF are used as clinical prognostic indicators. Among cancers with abnormal cMet genes, gastric cancer and colorectal cancer are typical. In gastric cancer, about 10-20% of cMet gene copy number amplifications and about 25% of cMet gene mutations; in colorectal cancer patients, the cMet mutation rate is about 15%. cMet is overexpressed in 25%-75% of NSCLC. In EGFR-TKI-resistant NSCLC, 15%-30% have acquired amplification of the cMet gene, and cMet is also used as a detection marker for TKI resistance in NSCLC patients with EGFR mutations.
针对cMet的靶向药物都是抑制HGF/cMet信号通路,处于临床上试验的主要包括受体酶抑制剂(例如Tivantinib、Capmatinib等),单克隆抗体(例如Onartuzumab),以及抗体药物偶联物(例如Telisotuzumab vedotin)。由于许多类型的癌症中cMet突变导致其过表达,NSCLC中EGFR-TKI耐药引发的获得性基因扩增以及cMet抑制剂开发中的耐药性等,针对特定适应症及病人群体的多靶点开发或者联合治疗将有机会提高临床疗效。Targeted drugs for cMet all inhibit the HGF/cMet signaling pathway. Those in clinical trials mainly include receptor enzyme inhibitors (such as Tivantinib, Capmatinib, etc.), monoclonal antibodies (such as Onartuzumab), and antibody drug conjugates ( For example Telisotuzumab vedotin). Due to cMet mutations in many types of cancer leading to its overexpression, acquired gene amplification caused by EGFR-TKI resistance in NSCLC, and resistance in the development of cMet inhibitors, multiple targets for specific indications and patient groups Developing or combining treatments will have the opportunity to improve clinical efficacy.
抗原结合结构域antigen binding domain
在本发明中,嵌合抗原受体CAR的抗原结合结构域特异性结合于肿瘤细胞标志物,比如EGFR、cMet、Her2、Her3、Muc1、ROR1、或PD-L1等。CAR的抗原结合结构域是由单克隆抗体的重链和轻链通过不同长度的柔性接头连接形成的单链可变片段(scFv)。svFv序列通常衍生来自于小鼠、人源化或者全人源单克隆抗体。另外,抗原结合结构域也包括由于缺少轻链、较小的,来自于羊驼的重链纳米抗体 (VH/nanobody)。scFv或VH对靶点抗原的亲和力对于调节CAR T细胞功能至关重要,但过高的亲和力有可能致使CAR T细胞过度激活而导致细胞死亡。除了亲和力之外,靶抗原的密度和表位也是影响抗原识别以及功效的重要因素。另外,抗原非依赖性的scFv的以及CAR分子在细胞膜上的高表达会诱导CAR分子的聚集,产生抗原非依赖性信号转导(tonic signalling)和脱靶激活,最终可能导致CAR T细胞的早期耗竭。In the present invention, the antigen-binding domain of the chimeric antigen receptor CAR specifically binds to tumor cell markers, such as EGFR, cMet, Her2, Her3, Mucl, ROR1, or PD-L1, etc. The antigen-binding domain of CAR is a single-chain variable fragment (scFv) formed by connecting the heavy chain and light chain of a monoclonal antibody through flexible linkers of different lengths. svFv sequences are usually derived from mouse, humanized or fully human monoclonal antibodies. In addition, the antigen-binding domain also includes smaller, alpaca-derived heavy chain Nanobodies due to the lack of light chains. (VH/nanobody). The affinity of scFv or VH to the target antigen is crucial for regulating CAR T cell function, but too high affinity may cause overactivation of CAR T cells and lead to cell death. In addition to affinity, the density and epitope of the target antigen are also important factors affecting antigen recognition and efficacy. In addition, the high expression of antigen-independent scFv and CAR molecules on the cell membrane will induce the aggregation of CAR molecules, resulting in antigen-independent signal transduction (tonic signaling) and off-target activation, which may ultimately lead to early exhaustion of CAR T cells. .
铰链区和跨膜区hinge region and transmembrane region
对于铰链区和跨膜区(跨膜结构域),CAR可被设计以包括融合至CAR的胞外结构域的跨膜结构域。在一个实施方式中,使用天然与CAR中的结构域之一相关联的跨膜结构域。在一些例子中,可选择跨膜结构域,或通过氨基酸置换进行修饰,以避免将这样的结构域结合至相同或不同的表面膜蛋白的跨膜结构域,从而最小化与受体复合物的其他成员的相互作用。For the hinge region and the transmembrane region (transmembrane domain), the CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR. In one embodiment, a transmembrane domain naturally associated with one of the domains in the CAR is used. In some examples, transmembrane domains may 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, thus minimizing interaction with the receptor complex. Interactions with other members.
跨膜结构域可源于天然来源或合成来源。在天然来源中,该结构域可源于任何膜结合蛋白或跨膜蛋白。优选地,本发明的CAR中的铰链区为CD8的铰链区,本发明的跨膜区为CD8或CD28的跨膜区。The transmembrane domain may be derived from natural or synthetic sources. In natural sources, this domain can be derived from any membrane-bound or transmembrane protein. Preferably, 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 CD8 or CD28.
胞内结构域intracellular domain
本发明的CAR的胞内结构域或另外的细胞内信号传导结构域是造成其中已放置CAR的免疫细胞的至少一种正常效应子功能的活化的原因。术语“效应子功能”指的是细胞的专有功能。例如,T细胞的效应子功能可为包括细胞因子分泌的细胞溶解活性或辅助活性。因此术语“细胞内信号传导结构域”指的是转导效应子功能信号并指导细胞实施专有功能的蛋白部分。尽管通常可使用整个细胞内信号传导结构域,但在很多例子中,不必使用整个链。就使用细胞内信号传导结构域的截短部分而言,这种截短部分可用于代替完整的链,只要它转导效应子功能信号。术语“细胞内信号传导结构域”通常指包括足以转导效应子功能信号的细胞内信号传导结构域的任何截短部分。The intracellular domain or additional intracellular signaling domain of the CAR of the invention is responsible for the activation of at least one normal effector function of the immune cell in which the CAR has been placed. The term "effector function" refers to a cell's specialized function. For example, the effector function of a T cell may be cytolytic activity or auxiliary activity including cytokine secretion. The term "intracellular signaling domain" therefore refers to the portion of a protein that transduces effector function signals and directs the cell to perform specialized functions. Although typically the entire intracellular signaling domain can be used, in many instances it is not necessary to use the entire chain. To the extent that a truncated portion of an intracellular signaling domain is used, such a truncated portion can be used in place of the intact chain as long as it transduces an effector function signal. The term "intracellular signaling domain" generally refers to any truncated portion of an intracellular signaling domain that is sufficient to transduce an effector function signal.
用于本发明的CAR的细胞内信号传导结构域的优选例子包括T细胞受体(TCR)的胞浆序列和协同行动以在抗原受体结合后开始信号转导的共受体,以及这些序列的任何衍生物或变体和具有相同的功能能力的任何合成序列。Preferred examples of intracellular signaling domains for use in CARs of the present invention include cytoplasmic sequences of T cell receptors (TCRs) and co-receptors that act cooperatively to initiate signal transduction upon antigen receptor binding, as well as these sequences any derivative or variant and any synthetic sequence having the same functional capabilities.
在优选的实施方式中,CAR的胞浆结构域可被设计以本身包括CD3ζ信号传导 结构域,或可与在本发明的CAR的内容中有用的任何其他期望的胞浆结构域(一个或多个)联合。例如,CAR的胞浆结构域可包括CD3ζ链部分和共刺激信号传导区。共刺激信号传导区指的是包括共刺激分子的细胞内结构域的一部分CAR。共刺激分子是淋巴细胞对抗原的有效应答所需的细胞表面分子,而不是抗原受体或它们的配体。优选地,包括4-1BB(CD137)、CD28等。In preferred embodiments, the cytoplasmic domain of the CAR can be designed to include CD3ζ signaling itself domain, or may be combined with any other desired cytoplasmic domain(s) useful in the context of the CAR of the invention. For example, the cytoplasmic domain of a CAR may include a CD3 zeta chain portion and a costimulatory signaling region. The costimulatory signaling region refers to the portion of the CAR that includes the intracellular domain of the costimulatory molecule. Costimulatory molecules are cell surface molecules required for an effective lymphocyte response to antigen, rather than antigen receptors or their ligands. Preferably, 4-1BB (CD137), CD28, etc. are included.
本发明的CAR的胞浆信号传导部分内的胞浆信号传导序列可以随机或以规定的顺序相互连接。任选地,短的寡肽或多肽连接体,优选长度在2和10个氨基酸,可形成该连接。甘氨酸-丝氨酸双联体提供了特别合适的连接体。The cytoplasmic signaling sequences within the cytoplasmic signaling portion of the CAR of the present invention can be connected to each other randomly or in a prescribed order. Optionally, short oligopeptide or polypeptide linkers, preferably between 2 and 10 amino acids in length, can form the linkage. Glycine-serine doublets provide particularly suitable linkers.
在一个实施方式中,本发明的CAR中的胞浆结构域被设计以包括4-1BB、和/或CD28的信号传导结构域(共刺激分子)以及CD3ζ的信号传导结构域。In one embodiment, the cytoplasmic domain in the CAR of the invention is designed to include 4-1BB, and/or the signaling domain of CD28 (costimulatory molecule) and the signaling domain of CD3ζ.
嵌合抗原受体(CAR)Chimeric Antigen Receptor (CAR)
嵌合免疫抗原受体(Chimeric antigen receptors,CARs)由胞外抗原识别区域,通常是scFv(single-chain variable fragment),跨膜区以及胞内共刺激信号区域组成。CARs的设计经历了以下过程:第一代CAR只有一个胞内信号组份CD3ζ或者FcγRI分子,由于胞内只有一个活化结构域,因此它只能引起短暂的T细胞增殖和较少的细胞因子分泌,而并不能提供长时间的T细胞增殖信号和持续的体内抗肿瘤效应,所以并没有取得很好的临床疗效。第二代CARs在原有结构基础上引入一个共刺激分子,如CD28、4-1BB、OX40、ICOS,与一代CARs相比功能有很大提高,进一步加强CAR-T细胞的持续性和对肿瘤细胞的杀伤能力。在二代CARs基础上串联一些新的免疫共刺激分子如CD27、CD134,发展成为三代和四代CARs。Chimeric antigen receptors (CARs) are composed of an extracellular antigen recognition region, usually scFv (single-chain variable fragment), a transmembrane region, and an intracellular costimulatory signal region. The design of CARs has gone through the following process: the first-generation CAR has only one intracellular signaling component, CD3ζ or FcγRI molecule. Since there is only one activation domain in the cell, it can only cause short-term T cell proliferation and less cytokine secretion. , but cannot provide long-term T cell proliferation signals and sustained anti-tumor effects in vivo, so it has not achieved good clinical efficacy. The second-generation CARs introduce a co-stimulatory molecule, such as CD28, 4-1BB, OX40, and ICOS, based on the original structure. Compared with the first-generation CARs, their functions are greatly improved, further enhancing the persistence of CAR-T cells and their ability to target tumor cells. of lethality. On the basis of second-generation CARs, some new immune costimulatory molecules such as CD27 and CD134 are connected in series to develop into third- and fourth-generation CARs.
CARs的胞外段可识别一个特异的抗原,随后通过胞内结构域转导该信号,引起细胞的活化增殖、细胞溶解毒性和分泌细胞因子,进而清除靶细胞。首先分离病人自体细胞(或者异源供体),激活并进行基因改造产生CAR的免疫细胞,随后注入同一病人体内。这种方式患移植物抗宿主病概率极低,抗原被免疫细胞以非MHC限制方式识别。The extracellular segment of CARs can recognize a specific antigen and then transduce the signal through the intracellular domain, causing cell activation and proliferation, cytolytic toxicity and secretion of cytokines, thereby eliminating target cells. First, the patient's autologous cells (or allogeneic donors) are isolated, activated and genetically modified to produce CAR immune cells, and then injected into the same patient. In this way, the probability of developing graft-versus-host disease is extremely low, and the antigen is recognized by immune cells in a non-MHC-restricted manner.
CAR-免疫细胞治疗在血液恶性肿瘤治疗中取得了非常高的临床反应率,这样的高反应率是以往任何一种治疗手段都无法达到的,在世界各国引发了临床研究的热潮。CAR-immune cell therapy has achieved a very high clinical response rate in the treatment of hematological malignancies. Such a high response rate has been unachievable by any previous treatment method, triggering an upsurge in clinical research in countries around the world.
具体地,本发明的嵌合抗原受体(CAR)包括细胞外结构域、跨膜结构域、和细胞内结构域。胞外结构域包括靶-特异性结合元件(也称为抗原结合结构域)。细胞 内结构域包括共刺激信号传导区和/或ζ链部分。共刺激信号传导区指包括共刺激分子的细胞内结构域的一部分。共刺激分子为淋巴细胞对抗原的有效应答所需要的细胞表面分子,而不是抗原受体或它们的配体。Specifically, 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). cell The endodomain includes costimulatory signaling regions and/or zeta chain portions. A costimulatory signaling domain refers to the portion of the intracellular domain that includes costimulatory molecules. Costimulatory molecules are cell surface molecules that are required for effective lymphocyte response to antigen, rather than antigen receptors or their ligands.
在CAR的胞外结构域和跨膜结构域之间,或在CAR的胞浆结构域和跨膜结构域之间,可并入接头。如本文所用的,术语“接头”通常指起到将跨膜结构域连接至多肽链的胞外结构域或胞浆结构域作用的任何寡肽或多肽。接头可包括0-300个氨基酸,优选地2至100个氨基酸和最优选地3至50个氨基酸。Linkers can be incorporated between the extracellular and transmembrane domains of the CAR, or between the cytoplasmic and transmembrane domains of the CAR. As used herein, the term "linker" generally refers to any oligopeptide or polypeptide that serves to connect a 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.
本发明的CAR当在T细胞中表达时,能够基于抗原结合特异性进行抗原识别。当其结合其关联抗原时,影响肿瘤细胞,导致肿瘤细胞不生长、被促使死亡或以其他方式被影响,并导致患者的肿瘤负荷缩小或消除。抗原结合结构域优选与来自共刺激分子和/或ζ链中的一个或多个的细胞内结构域融合。优选地,抗原结合结构域与4-1BB信号传导结构域和/或CD3ζ信号结构域组合的细胞内结构域融合。When expressed in T cells, the CAR of the present invention is capable of antigen recognition based on antigen-binding specificity. When it binds to its cognate antigen, it affects tumor cells, causing the tumor cells to fail to grow, be driven to death, or otherwise affected, and cause the patient's tumor burden to shrink or be eliminated. The antigen binding domain is preferably fused to an intracellular domain from one or more of the costimulatory molecules and/or zeta chains. Preferably, the antigen binding domain is fused to an intracellular domain in combination with a 4-1BB signaling domain and/or a CD3ζ signaling domain.
如本文所用,“抗原结合结构域”“单链抗体片段”均指具有抗原结合活性的Fab片段,Fab’片段,F(ab’)2片段,或单一Fv片段。Fv抗体含有抗体重链可变区、轻链可变区,但没有恒定区,并具有全部抗原结合位点的最小抗体片段。一般的,Fv抗体还包含VH和VL结构域之间的多肽接头,且能够形成抗原结合所需的结构。抗原结合结构域通常是scFv(single-chain variable fragment)。scFv的大小一般是一个完整抗体的1/6。单链抗体优选是由一条核苷酸链编码的一条氨基酸链序列。作为本发明的优选方式,所述scFv包含特异性识别肿瘤高表达肿瘤细胞标志物(比如PSMA、GPC3、GD2、HER2、Mesothelin(MSLN)、CEA、EGFR/EGFRvIII、Claudin18.2、Mucin 1(MUC1)、NKG2D配体、CD19、CD20、BCMA、CD22、CD30、IL3RA、CD38、CD138)的抗体,较佳地为单链抗体。As used herein, "antigen-binding domain" and "single-chain antibody fragment" all refer to a Fab fragment, a Fab' fragment, an F(ab')2 fragment, or a single Fv fragment with antigen-binding activity. Fv antibodies are the smallest antibody fragments that contain an antibody heavy chain variable region, a light chain variable region, but no constant region, and have all antigen-binding sites. Typically, Fv antibodies also contain a polypeptide linker between the VH and VL domains and are capable of forming 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 a complete antibody. Single chain antibodies are preferably one amino acid chain sequence encoded by one nucleotide chain. As a preferred mode of the present invention, the scFv includes specific recognition of tumor cell markers with high expression (such as PSMA, GPC3, GD2, HER2, Mesothelin (MSLN), CEA, EGFR/EGFRvIII, Claudin18.2, Mucin 1 (MUC1 ), NKG2D ligand, CD19, CD20, BCMA, CD22, CD30, IL3RA, CD38, CD138), preferably single chain antibodies.
在一优选实施方式中,本发明CAR的抗原结合部分靶向第一肿瘤细胞标志物和第二肿瘤细胞标志物。在一优选实施方式中,本发明的CAR的抗原结合部分是靶向cMet的第一scFV和靶向EGFR的第二scFv。In a preferred embodiment, the antigen-binding portion of the CAR of the present invention targets the first tumor cell marker and the second tumor cell marker. In a preferred embodiment, the antigen-binding portion of the CAR of the present invention is a first scFv targeting cMet and a second scFv targeting EGFR.
在一优选实施方式中,所述的第一scFv的结构如式A1或A2所示:
VH1-VL1  (A1);或
VL1-VH1  (A2);
In a preferred embodiment, the structure of the first scFv is shown in Formula A1 or A2:
V H1 -V L1 (A1); or
V L1 -V H1 (A2);
其中,VL1为抗第一肿瘤细胞标志物抗体的轻链可变区;VH1为抗第一肿瘤细胞标志物抗体的重链可变区;“-”为连接肽(或柔性接头)或肽键。Wherein, V L1 is the light chain variable region of the anti-first tumor cell marker antibody; V H1 is the heavy chain variable region of the anti-first tumor cell marker antibody; "-" is the connecting peptide (or flexible linker) or Peptide bonds.
在一优选实施方式中,VL1的氨基酸序列如SEQ ID NO.:1中第135-247位 所示,且VH1的氨基酸序列如SEQ ID NO.:1的第1-119位所示。In a preferred embodiment, the amino acid sequence of V L1 is such as positions 135-247 in SEQ ID NO.:1 is shown, and the amino acid sequence of V H1 is shown in positions 1-119 of SEQ ID NO.: 1.
在一优选实施方式中,所述的第二scFv的结构如式A3或A4所示:
VH2-VL2  (A3);或
VL2-VH2  (A4);
In a preferred embodiment, the structure of the second scFv is shown in formula A3 or A4:
V H2 -V L2 (A3); or
V L2 -V H2 (A4);
其中,VL2为抗第二肿瘤细胞标志物抗体的轻链可变区;VH2为抗第二肿瘤细胞标志物抗体的重链可变区;“-”为连接肽(或柔性接头)或肽键。Wherein, V L2 is the light chain variable region of the anti-second tumor cell marker antibody; V H2 is the heavy chain variable region of the anti-second tumor cell marker antibody; "-" is the connecting peptide (or flexible linker) or Peptide bonds.
在一优选实施方式中,VL2的氨基酸序列如SEQ ID NO.:5中第135-241位所示,且VH2的氨基酸序列如SEQ ID NO.:5的第1-119位所示。In a preferred embodiment, the amino acid sequence of V L2 is shown in positions 135-241 of SEQ ID NO.:5, and the amino acid sequence of V H2 is shown in positions 1-119 of SEQ ID NO.:5.
在一优选实施方式中,第一scFV、第二scFv包含变体形式,所述变体与其野生型的第一scFV、第二scFV序列分别具有≥80%、≥85%、≥90%、≥95%、≥98%或≥99%的同源性。In a preferred embodiment, the first scFV and the second scFv comprise variant forms, and the variants have ≥80%, ≥85%, ≥90%, ≥ 95%, ≥98% or ≥99% homology.
在本发明中,本发明的第一scFV、第二scFV还包括其保守性变异体,指分别与本发明的第一scFV、第二scFV的氨基酸序列相比,有至多10个,较佳地至多8个,更佳地至多5个,最佳地至多3个氨基酸被性质相似或相近的氨基酸所替换而形成多肽。In the present invention, the first scFV and the second scFV of the invention also include conservative variants thereof, which means that compared with the amino acid sequences of the first scFV and the second scFV of the invention respectively, there are at most 10, preferably At most 8, more preferably at most 5, most preferably at most 3 amino acids are replaced by amino acids with similar or similar properties to form a polypeptide.
在本发明中,所述添加、缺失、修饰和/或取代的氨基酸数量,优选为不超过初始氨基酸序列总氨基酸数量的40%,更优选为不超过35%,更优选为1-33%,更优选为5-30%,更优选为10-25%,更优选为15-20%。In the present invention, 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%, and more preferably 1-33%. More preferably, it is 5-30%, more preferably, it is 10-25%, and even more preferably, it is 15-20%.
在本发明中,所述添加、缺失、修饰和/或取代的氨基酸数量通常是1、2、3、4或5个,较佳地为1-3个,更佳地为1-2个,最佳地为1个。In the present invention, 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, Optimally 1.
对于铰链区和跨膜区(跨膜结构域),CAR可被设计以包括融合至CAR的胞外结构域的跨膜结构域。在一个实施方式中,使用天然与CAR中的结构域之一相关联的跨膜结构域。在一些例子中,可选择跨膜结构域,或通过氨基酸置换进行修饰,以避免将这样的结构域结合至相同或不同的表面膜蛋白的跨膜结构域,从而最小化与受体复合物的其他成员的相互作用。For the hinge region and the transmembrane region (transmembrane domain), the CAR can be designed to include the transmembrane domain fused to the extracellular domain of the CAR. In one embodiment, a transmembrane domain naturally associated with one of the domains in the CAR is used. In some examples, transmembrane domains may 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 interaction with the receptor complex. Interactions with other members.
在本发明中,本发明的第一CAR中的胞内结构域包括CD8的跨膜区、CD3ζ的信号传导结构域。In the present invention, the intracellular domain in the first CAR of the present invention includes the transmembrane region of CD8 and the signaling domain of CD3ζ.
在本发明中,本发明的第二CAR中的胞内结构域包括CD8或CD28的跨膜区、4-1BB或CD28的共刺激因子。In the present invention, the intracellular domain in the second CAR of the present invention includes the transmembrane region of CD8 or CD28, and the costimulatory factor of 4-1BB or CD28.
在本发明的一优选实施方式中,所述第一CAR的氨基酸序列如SEQ ID NO.:2-4中任一所示。 In a preferred embodiment of the present invention, the amino acid sequence of the first CAR is as shown in any one of SEQ ID NO.: 2-4.
在本发明的一优选实施方式中,所述第二CAR的氨基酸序列SEQ ID NO.:7-14中任一所示。In a preferred embodiment of the present invention, the amino acid sequence of the second CAR is shown in any one of SEQ ID NO.: 7-14.
在本发明的一优选实施方式中,所述含有第一CAR和第二CAR的融合蛋白(即logic BiCAR)的氨基酸序列如SEQ ID NO.:23-26中任一所示。In a preferred embodiment of the present invention, the amino acid sequence of the fusion protein containing the first CAR and the second CAR (ie, logic BiCAR) is as shown in any one of SEQ ID NO.: 23-26.
其中,在SEQ ID NO.:23中第1-22位为第一信号肽;第23-269位为靶向第一肿瘤细胞标志物的抗原结合结构域(比如靶向cMet的抗体单链可变区序列);第270-314位为铰链区;第315-338位为跨膜区(如CD8的跨膜区);第339-450位为CD3ζ,第451-472位是连接肽(如自剪切蛋白),第473-493位为第二信号肽,第494-734位为靶向第二肿瘤细胞标志物的抗原结合结构域(比如靶向EGFR的抗体单链可变区序列);第735-779位为铰链区;第780-806位为跨膜区(如CD28的跨膜区);第807-847位为共刺激元件(如CD28)。Among them, in SEQ ID NO.: 23, positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3ζ, and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembrane region of CD28); positions 807-847 are costimulatory elements (such as CD28).
其中,在SEQ ID NO.:24中第1-22位为第一信号肽;第23-269位为靶向第一肿瘤细胞标志物的抗原结合结构域(比如靶向cMet的抗体单链可变区序列);第270-314位为铰链区;第315-338位为跨膜区(如CD8的跨膜区);第339-450位为CD3ζ,第451-472位是连接肽(如自剪切蛋白),第473-493位为第二信号肽,第494-734位为靶向第二肿瘤细胞标志物的抗原结合结构域(比如靶向EGFR的抗体单链可变区序列);第735-779位为铰链区;第780-806位为跨膜区(如CD28的跨膜区);第807-847位为共刺激元件(如CD28)。Among them, in SEQ ID NO.: 24, positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3ζ, and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; positions 780-806 are the transmembrane region (such as the transmembrane region of CD28); positions 807-847 are costimulatory elements (such as CD28).
其中,在SEQ ID NO.:25中第1-22位为第一信号肽;第23-269位为靶向第一肿瘤细胞标志物的抗原结合结构域(比如靶向cMet的抗体单链可变区序列);第270-314位为铰链区;第315-338位为跨膜区(如CD8的跨膜区);第339-450位为CD3ζ,第451-472位是连接肽(如自剪切蛋白),第473-493位为第二信号肽,第494-734位为靶向第二肿瘤细胞标志物的抗原结合结构域(比如靶向EGFR的抗体单链可变区序列);第735-779位为铰链区;第780-803位为跨膜区(如CD8的跨膜区);第804-845位为共刺激元件(如4-1BB)。Among them, in SEQ ID NO.: 25, positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3ζ, and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; positions 780-803 are the transmembrane region (such as the transmembrane region of CD8); positions 804-845 are costimulatory elements (such as 4-1BB).
其中,在SEQ ID NO.:26中第1-22位为第一信号肽;第23-269位为靶向第一肿瘤细胞标志物的抗原结合结构域(比如靶向cMet的抗体单链可变区序列);第270-314位为铰链区;第315-338位为跨膜区(如CD8的跨膜区);第339-450位为CD3ζ,第451-472位是连接肽(如自剪切蛋白),第473-493位为第二信号肽,第494-734位为靶向第二肿瘤细胞标志物的抗原结合结构域(比如靶向EGFR的抗体单链可变区序列);第735-779位为铰链区;第780-803位为跨膜 区(如CD8的跨膜区);第804-845位为共刺激元件(如4-1BB)。Among them, in SEQ ID NO.: 26, positions 1-22 are the first signal peptide; positions 23-269 are the antigen-binding domains targeting the first tumor cell marker (for example, the antibody single chain targeting cMet can variable region sequence); positions 270-314 are the hinge region; positions 315-338 are the transmembrane region (such as the transmembrane region of CD8); positions 339-450 are CD3ζ, and positions 451-472 are the connecting peptide (such as self-cleaving protein), positions 473-493 are the second signal peptide, and positions 494-734 are the antigen-binding domain targeting the second tumor cell marker (such as the single-chain variable region sequence of an antibody targeting EGFR) ; Positions 735-779 are the hinge region; Positions 780-803 are the transmembrane region region (such as the transmembrane region of CD8); positions 804-845 are costimulatory elements (such as 4-1BB).
嵌合抗原受体T细胞(CAR-T细胞)Chimeric Antigen Receptor T Cells (CAR-T Cells)
如本文所用,术语“CAR-T细胞”、“CAR-T”、“本发明CAR-T细胞”均指本发明所述的CAR-T细胞,本发明CAR-T细胞可靶向肿瘤表面抗原(如PSMA),用来治疗PSMA高表达或阳性的肿瘤,尤其是实体瘤。As used herein, the terms "CAR-T cells", "CAR-T" and "CAR-T cells of the present invention" all refer to the CAR-T cells of the present invention. The CAR-T cells of the present invention can target tumor surface antigens. (such as PSMA), used to treat tumors with high expression or positive PSMA, especially solid tumors.
CAR-T细胞较其它基于T细胞的治疗方式存在以下优势:(1)CAR-T细胞的作用过程不受MHC的限制;(2)鉴于很多肿瘤细胞表达相同的肿瘤抗原,针对某一种肿瘤抗原的CAR基因构建一旦完成,便可以被广泛利用;(3)CAR既可以利用肿瘤蛋白质抗原,又可利用糖脂类非蛋白质抗原,扩大了肿瘤抗原的靶点范围;(4)使用患者自体细胞降低了排异反应的风险;(5)CAR-T细胞具有免疫记忆功能,可以长期在体内存活。CAR-T cells have the following advantages over other T cell-based treatments: (1) The action process of CAR-T cells is not restricted by MHC; (2) Since many tumor cells express the same tumor antigen, targeting a certain 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) Use patients' autologous The cells reduce the risk of rejection; (5) CAR-T cells have immune memory function and can survive in the body for a long time.
在本发明中,本发明的含有第一CAR和第二CAR的融合蛋白(即logic BiCAR)包含(i)胞外结构域,其包含靶向第一肿瘤细胞表面抗原的抗原;(ii)第一绞链区;(iii)第一跨膜域;(iv)任选的第一共刺激因子;和(iv)CD3ζ的信号传导结构域;以及;(v)连接肽(如自剪切蛋白);(vi)胞外结构域,其包含靶向第二肿瘤细胞表面抗原的抗原;(ii)第二绞链区;(iii)第二跨膜域;和(iv)第二共刺激因子。In the present invention, the fusion protein containing the first CAR and the second CAR (i.e., logic BiCAR) of the present invention includes (i) an extracellular domain, which includes an antigen targeting a first tumor cell surface antigen; (ii) a third a hinge region; (iii) a first transmembrane domain; (iv) an optional first costimulator; and (iv) a signaling domain of CD3ζ; and; (v) a linking peptide (e.g., a self-cleaving protein ); (vi) an extracellular domain comprising an antigen targeting a second tumor cell surface antigen; (ii) a second hinge region; (iii) a second transmembrane domain; and (iv) a second costimulatory factor .
嵌合抗原受体巨噬细胞(CAR-M细胞)Chimeric Antigen Receptor Macrophages (CAR-M cells)
如本文所用,术语“CAR-M细胞”、“CAR-M”、“本发明CAR-M细胞”均指本发明所述的CAR-M细胞,本发明CAR-M细胞可靶向肿瘤表面抗原(如cMet和EGFR),用来治疗肿瘤抗原(比如cMet和EGFR)高表达或阳性的肿瘤,尤其是实体瘤。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. The CAR-M cells of the present invention can target tumor surface antigens. (such as cMet and EGFR), used to treat tumors with high expression or positivity of tumor antigens (such as cMet and EGFR), especially solid tumors.
巨噬细胞是先天免疫系统的主要效应器和调节者,具有吞噬能力,能够分泌促炎因子,将抗原呈递给T细胞激活免疫系统。Macrophages are the main effectors and regulators of the innate immune system. They have phagocytic ability, can secrete pro-inflammatory factors, and present antigens to T cells to activate the immune system.
CAR-M相比于CART,其本身就可以体外直接杀死抗原特异性肿瘤细胞,体内抑制肿瘤生长,重塑肿瘤微环境,具有良好的抗肿瘤活性,此外,CAR-M还具有抗原呈递能力,将肿瘤细胞抗原呈递并激活内源T细胞。Compared with CART, CAR-M can directly kill antigen-specific tumor cells in vitro, inhibit tumor growth in vivo, reshape the tumor microenvironment, and has good anti-tumor activity. In addition, CAR-M also has the ability to present antigens. , present tumor cell antigens and activate endogenous T cells.
嵌合抗原受体NK细胞(CAR-NK细胞) Chimeric Antigen Receptor NK Cells (CAR-NK Cells)
如本文所用,术语“CAR-NK细胞”、“CAR-NK”、“本发明CAR-NK细胞”均指本发明所述的CAR-NK细胞。本发明CAR-NK细胞可靶向肿瘤表面抗原(如cMet和EGFR),用于治疗cMet和EGFR高表达或阳性的肿瘤,尤其是实体瘤。As used herein, the terms "CAR-NK cells", "CAR-NK" and "CAR-NK cells of the present invention" all refer to the CAR-NK cells of the present invention. The CAR-NK cells of the present invention can target tumor surface antigens (such as cMet and EGFR) and are used to treat tumors with high cMet and EGFR expression or positivity, especially solid tumors.
自然杀伤(NK)细胞是一类主要的免疫效应细胞,通过非抗原特异性途径去保护机体免受病毒感染和肿瘤细胞的侵袭。通过工程化(基因修饰)的NK细胞可能获得新的功能,包括特异性识别肿瘤抗原的能力及具有增强的抗肿瘤细胞毒作用。Natural killer (NK) cells are a major type of immune effector cells that protect the body from viral infection and tumor cell invasion through non-antigen-specific pathways. Engineered (genetically modified) NK cells may acquire new functions, including the ability to specifically recognize tumor antigens and enhanced anti-tumor cytotoxicity.
与自体CAR-T细胞相比,CAR-NK细胞还具有一下优点,例如:(1)通过释放穿孔素和颗粒酶直接杀伤肿瘤细胞,而对机体正常的细胞没有杀伤作用;(2)它们释放很少量的细胞因子从而降低了细胞因子风暴的危险;(3)体外极易扩增及发展为“现成的”产品。除此之外,与CAR-T细胞治疗类似。Compared with autologous CAR-T cells, CAR-NK cells also have the following advantages, such as: (1) they directly kill tumor cells by releasing perforin and granzyme, but have no killing effect on normal cells of the body; (2) they release A very small amount of cytokines thus reduces the risk of cytokine storm; (3) It is easy to amplify in vitro and develop into "off-the-shelf" products. Otherwise, it is similar to CAR-T cell therapy.
外源T细胞抗原受体exogenous T cell antigen receptor
如本文所用,外源T细胞抗原受体(T cell receptor,TCR)为通过基因转移技术从肿瘤反应性T细胞中克隆出TCR的α链和β链,通过基因工程的手段,以慢病毒或逆转录病毒为载体,外源性转入到T细胞内的TCR。As used in this article, exogenous T cell antigen receptor (TCR) refers to the α chain and β chain of TCR cloned from tumor reactive T cells through gene transfer technology, and through genetic engineering means, lentivirus or Retrovirus is used as a vector to transfer exogenous TCR into T cells.
外源TCR修饰的T细胞能够特异性识别和杀伤肿瘤细胞,并通过优化TCR与肿瘤性特异性抗原的亲和力,可以提高T细胞与肿瘤的亲和力,提高抗肿瘤效果。Exogenous TCR-modified T cells can specifically recognize and kill tumor cells, and by optimizing the affinity of TCR and tumor-specific antigens, the affinity of T cells with tumors can be increased and the anti-tumor effect can be improved.
载体carrier
编码期望分子的核酸序列可利用在本领域中已知的重组方法获得,诸如例如通过从表达基因的细胞中筛选文库,通过从已知包括该基因的载体中得到该基因,或通过利用标准的技术,从包含该基因的细胞和组织中直接分离。可选地,感兴趣的基因可被合成生产。Nucleic acid sequences encoding the desired molecules can be obtained using recombinant methods known in the art, such as, for example, by screening libraries from cells expressing the gene, by obtaining the gene from a vector known to include the gene, or by using standard technology to isolate directly from cells and tissues containing the gene. Alternatively, the gene of interest can be produced synthetically.
本发明也提供了其中插入本发明的表达盒的载体。源于逆转录病毒诸如慢病毒的载体是实现长期基因转移的合适工具,因为它们允许转基因长期、稳定的整合并且其在子细胞中增殖。慢病毒载体具有超过源自致癌逆转录病毒诸如鼠科白血病病毒的载体的优点,因为它们可转导非增殖的细胞,诸如肝细胞。它们也具有低免疫原性的优点。The invention also provides vectors into which the expression cassette of the invention is inserted. Vectors derived from retroviruses such as lentiviruses are suitable tools to achieve long-term gene transfer because they allow long-term, stable integration of the transgene and its propagation in daughter cells. Lentiviral vectors have advantages over vectors derived from oncogenic retroviruses, such as murine leukemia virus, in that they can transduce non-proliferating cells, such as hepatocytes. They also have the advantage of low immunogenicity.
简单概括,通常可操作地连接本发明的表达盒或核酸序列至启动子,并将其并入表达载体。该载体适合于复制和整合真核细胞。典型的克隆载体包含可用于调节期望核酸序列表达的转录和翻译终止子、初始序列和启动子。 Briefly summarized, the expression cassette or nucleic acid sequence of the invention is typically operably linked to a promoter and incorporated into an expression vector. This vector is suitable for replication and integration into eukaryotic cells. Typical cloning vectors contain transcriptional and translational terminators, initial sequences, and promoters that can be used to regulate expression of the desired nucleic acid sequence.
本发明的表达构建体也可利用标准的基因传递方案,用于核酸免疫和基因疗法。基因传递的方法在本领域中是已知的。见例如美国专利号5,399,346、5,580,859、5,589,466,在此通过引用全文并入。在另一个实施方式中,本发明提供了基因疗法载体。The expression constructs of the present invention can also be used for nucleic acid immunization and gene therapy using standard gene delivery protocols. 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 incorporated by reference in their entirety. In another embodiment, the present invention provides gene therapy vectors.
该核酸可被克隆入许多类型的载体。例如,该核酸可被克隆入此载体,其包括但不限于质粒、噬菌粒、噬菌体衍生物、动物病毒和粘粒。特定的感兴趣载体包括表达载体、复制载体、探针产生载体和测序载体。The nucleic acid can be cloned into many types of vectors. For example, the nucleic acid can be cloned into such vectors, which include, but are not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. Specific vectors of interest include expression vectors, replication vectors, probe generation vectors and sequencing vectors.
进一步地,表达载体可以以病毒载体形式提供给细胞。病毒载体技术在本领域中是公知的并在例如Sambrook等(2001,Molecular Cloning:A Laboratory Manual,Cold Spring Harbor Laboratory,New York)和其他病毒学和分子生物学手册中进行了描述。可用作载体的病毒包括但不限于逆转录病毒、腺病毒、腺伴随病毒、疱疹病毒和慢病毒。通常,合适的载体包含在至少一种有机体中起作用的复制起点、启动子序列、方便的限制酶位点和一个或多个可选择的标记(例如,WO01/96584;WO01/29058;和美国专利号6,326,193)。Further, 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, for example, in 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, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses. Typically, a suitable vector will contain an origin of replication functional in at least one organism, a promoter sequence, convenient restriction enzyme sites, and one or more selectable markers (eg, WO01/96584; WO01/29058; and U.S. Patent No. 6,326,193).
已经开发许多基于病毒的系统,用于将基因转移入哺乳动物细胞。例如,逆转录病毒提供了用于基因传递系统的方便的平台。可利用在本领域中已知的技术将选择的基因插入载体并包装入逆转录病毒颗粒。该重组病毒可随后被分离和传递至体内或离体的对象细胞。许多逆转录病毒系统在本领域中是已知的。在一些实施方式中,使用腺病毒载体。许多腺病毒载体在本领域中是已知的。在一个实施方式中,使用慢病毒载体。A number of virus-based systems have been developed for gene transfer into mammalian cells. For example, retroviruses provide a convenient platform for gene delivery systems. The selected genes can be inserted into the vector and packaged into retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to cells of the subject in vivo or ex vivo. Many retroviral systems are known in the art. In some embodiments, adenoviral vectors are used. Many adenoviral vectors are known in the art. In one embodiment, lentiviral vectors are used.
额外的启动子元件,例如增强子,可以调节转录开始的频率。通常地,这些位于起始位点上游的30-110bp区域中,尽管最近已经显示许多启动子也包含起始位点下游的功能元件。启动子元件之间的间隔经常是柔性的,以便当元件相对于另一个被倒置或移动时,保持启动子功能。在胸苷激酶(tk)启动子中,启动子元件之间的间隔可被增加隔开50bp,活性才开始下降。取决于启动子,表现出单个元件可合作或独立地起作用,以启动转录。Additional promoter elements, such as enhancers, can modulate the frequency with which transcription is initiated. Typically, these are located in a region of 30-110 bp 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 so that promoter function is maintained 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 activity begins to decrease. Depending on the promoter, individual elements are shown to act cooperatively or independently to initiate transcription.
合适的启动子的一个例子为即时早期巨细胞病毒(CMV)启动子序列。该启动子序列为能够驱动可操作地连接至其上的任何多核苷酸序列高水平表达的强组成型启动子序列。合适的启动子的另一个例子为延伸生长因子-1α(EF-1α)。然而,也可使用其他组成型启动子序列,包括但不限于类人猿病毒40(SV40)早期启动子、小鼠乳癌病毒(MMTV)、人免疫缺陷病毒(HIV)长末端重复(LTR)启动子、MoMuLV启 动子、鸟类白血病病毒启动子、艾伯斯坦-巴尔(Epstein-Barr)病毒即时早期启动子、鲁斯氏肉瘤病毒启动子、以及人基因启动子,诸如但不限于肌动蛋白启动子、肌球蛋白启动子、血红素启动子和肌酸激酶启动子。进一步地,本发明不应被限于组成型启动子的应用。诱导型启动子也被考虑为本发明的一部分。诱导型启动子的使用提供了分子开关,其能够当这样的表达是期望的时,打开可操作地连接诱导型启动子的多核苷酸序列的表达,或当表达是不期望的时关闭表达。诱导型启动子的例子包括但不限于金属硫蛋白启动子、糖皮质激素启动子、孕酮启动子和四环素启动子。An example of 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 thereto. Another example of a suitable promoter is elongation growth factor-1α (EF-1α). However, other constitutive promoter sequences may also be used, including, but not limited to, simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLVqi kinesin, avian leukemia virus promoter, Epstein-Barr virus immediate early promoter, Ruth's sarcoma virus promoter, and human gene promoters, such as, but not limited to, actin promoter, Myosin promoter, heme promoter, and creatine kinase promoter. Further, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also considered part of the invention. The use of an inducible promoter provides a molecular switch capable of turning on expression of a polynucleotide sequence operably linked to the inducible promoter when such expression is desired, or turning off expression when expression is undesirable. Examples of inducible promoters include, but are not limited to, metallothionein promoters, glucocorticoid promoters, progesterone promoters, and tetracycline promoters.
为了评估CAR多肽或其部分的表达,被引入细胞的表达载体也可包含可选择的标记基因或报道基因中的任一个或两者,以便于从通过病毒载体寻求被转染或感染的细胞群中鉴定和选择表达细胞。在其他方面,可选择的标记可被携带在单独一段DNA上并用于共转染程序。可选择的标记和报道基因两者的侧翼都可具有适当的调节序列,以便能够在宿主细胞中表达。有用的可选择标记包括例如抗生素抗性基因,诸如neo等等。To assess expression of a CAR polypeptide or portion thereof, the expression vector introduced into the cell may also contain either or both a selectable marker gene or a reporter gene to facilitate the identification of populations of cells that are transfected or infected by the viral vector. Identify and select expressing cells. In other aspects, the selectable marker can be carried on a separate stretch of DNA and used in co-transfection procedures. Both the selectable marker and the reporter gene can be flanked by appropriate regulatory sequences to enable expression in the host cell. Useful selectable markers include, for example, antibiotic resistance genes such as neo and the like.
报道基因用于鉴定潜在转染的细胞并用于评价调节序列的功能性。通常地,报道基因为以下基因:其不存在于受体有机体或组织或由受体有机体或组织进行表达,并且其编码多肽,该多肽的表达由一些可容易检测的性质例如酶活性清楚表示。在DNA已经被引入受体细胞后,报道基因的表达在合适的时间下进行测定。合适的报道基因可包括编码荧光素酶、β-半乳糖苷酶、氯霉素乙酰转移酶、分泌型碱性磷酸酶或绿色萤光蛋白的基因(例如,Ui-Tei等,2000FEBS Letters479:79-82)。合适的表达系统是公知的并可利用已知技术制备或从商业上获得。通常,显示最高水平的报道基因表达的具有最少5个侧翼区的构建体被鉴定为启动子。这样的启动子区可被连接至报道基因并用于评价试剂调节启动子-驱动转录的能力。Reporter genes are used to identify potentially transfected cells and to evaluate the functionality of regulatory sequences. Typically, a reporter gene is a gene that is not present in or expressed by the recipient organism or tissue and that encodes a polypeptide whose expression is clearly indicated by some readily detectable property, such as enzymatic activity. Expression of the reporter gene is measured at appropriate times after the DNA has been introduced into the recipient cell. Suitable reporter genes may include genes encoding luciferase, β-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. Typically, the construct with a minimum of 5 flanking regions that shows the highest level of reporter gene expression is identified as the promoter. Such promoter regions can be ligated to a reporter gene and used to evaluate the ability of an agent to regulate promoter-driven transcription.
将基因引入细胞和将基因表达入细胞的方法在本领域中是已知的。在表达载体的内容中,载体可通过在本领域中的任何方法容易地引入宿主细胞,例如,哺乳动物、细菌、酵母或昆虫细胞。例如,表达载体可通过物理、化学或生物学手段转移入宿主细胞。Methods of introducing genes into cells and expressing genes into cells are known in the art. In the context of an expression vector, the vector can be readily introduced into a host cell, eg, a mammalian, bacterial, yeast or insect cell, by any method known in the art. For example, expression vectors can be transferred into host cells by physical, chemical or biological means.
将多核苷酸引入宿主细胞的物理方法包括磷酸钙沉淀、脂质转染法、粒子轰击、微注射、电穿孔等等。生产包括载体和/或外源核酸的细胞的方法在本领域中是公知的。见例如Sambrook等(2001,Molecular Cloning:A Laboratory Manual,Cold Spring Harbor Laboratory,New York)。将多核苷酸引入宿主细胞的优选方 法为磷酸钙转染。Physical methods for introducing polynucleotides into host cells include calcium phosphate precipitation, lipofection, particle bombardment, microinjection, electroporation, and the like. 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). Preferred methods for introducing polynucleotides into host cells The method is calcium phosphate transfection.
将感兴趣的多核苷酸引入宿主细胞的生物学方法包括使用DNA和RNA载体。病毒载体,特别是逆转录病毒载体,已经成为最广泛使用的将基因插入哺乳动物例如人细胞的方法。其他病毒载体可源自慢病毒、痘病毒、单纯疱疹病毒I、腺病毒和腺伴随病毒等等。见例如美国专利号5,350,674和5,585,362。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 lentiviruses, poxviruses, herpes simplex virus I, adenovirus and adeno-associated virus, among others. See, for example, US Patent Nos. 5,350,674 and 5,585,362.
将多核苷酸引入宿主细胞的化学手段包括胶体分散系统,诸如大分子复合物、纳米胶囊、微球、珠;和基于脂质的系统,包括水包油乳剂、胶束、混合胶束和脂质体。用作体外和体内传递工具(delivery vehicle)的示例性胶体系统为脂质体(例如,人造膜囊)。Chemical means of introducing polynucleotides into host cells include colloidal dispersion systems, such as macromolecular complexes, nanocapsules, microspheres, beads; and lipid-based systems, including oil-in-water emulsions, micelles, mixed micelles, and liposomes. plastid. Exemplary colloidal systems useful as delivery vehicles in vitro and in vivo are liposomes (eg, artificial membrane vesicles).
在使用非病毒传递系统的情况下,示例性传递工具为脂质体。考虑使用脂质制剂,以将核酸引入宿主细胞(体外、离体(ex vivo)或体内)。在另一方面,该核酸可与脂质相关联。与脂质相关联的核酸可被封装入脂质体的水性内部中,散布在脂质体的脂双层内,经与脂质体和寡核苷酸两者都相关联的连接分子附接至脂质体,陷入脂质体,与脂质体复合,分散在包含脂质的溶液中,与脂质混合,与脂质联合,作为悬浮液包含在脂质中,包含在胶束中或与胶束复合,或以其他方式与脂质相关联。与组合物相关联的脂质、脂质/DNA或脂质/表达载体不限于溶液中的任何具体结构。例如,它们可存在于双分子层结构中,作为胶束或具有“坍缩的(collapsed)”结构。它们也可简单地被散布在溶液中,可能形成大小或形状不均一的聚集体。脂质为脂肪物质,其可为天然发生或合成的脂质。例如,脂质包括脂肪小滴,其天然发生在细胞质以及包含长链脂肪族烃和它们的衍生物诸如脂肪酸、醇类、胺类、氨基醇类和醛类的该类化合物中。Where non-viral delivery systems are used, an exemplary delivery vehicle is liposomes. Consider the use of lipid formulations to introduce nucleic acids into host cells (in vitro, ex vivo, or in vivo). In another aspect, the nucleic acid can be associated with a lipid. Nucleic acids associated with lipids can be encapsulated into the aqueous interior of the liposomes, dispersed within the lipid bilayer of the liposomes, attached via linker molecules associated with both the liposomes and the oligonucleotides to liposomes, entrapped in liposomes, complexed with liposomes, dispersed in a solution containing lipids, mixed with lipids, associated with lipids, contained in lipids as a suspension, contained in micelles or Complexed with micelles, or otherwise associated with lipids. The lipid, lipid/DNA or lipid/expression vector associated with the composition is not limited to any specific structure in solution. For example, they may exist in bilayer structures, as micelles or have a "collapsed" structure. They can also simply be dispersed in solution, possibly forming aggregates that are not uniform in size or shape. Lipids are fatty substances, which may be naturally occurring or synthetic lipids. For example, lipids include lipid droplets that occur naturally in the cytoplasm as well as compounds containing long-chain aliphatic hydrocarbons and their derivatives such as fatty acids, alcohols, amines, aminoalcohols, and aldehydes.
在本发明的一个优选地实施方式中,所述载体为逆转录病毒载体。In a preferred embodiment of the invention, the vector is a retroviral vector.
制剂preparation
本发明提供了一种本发明第一方面所述的工程化的免疫细胞、以及药学上可接受的载体、稀释剂或赋形剂。在一个实施方式中,所述制剂为液态制剂。优选地,所述制剂为注射剂。优选地,所述制剂中所述CAR-T细胞的浓度为1×103-1×108个细胞/Kg体重,更优地1×104-1×107个细胞/Kg体重。The invention provides an engineered immune cell according to the first aspect of the invention, and a pharmaceutically acceptable carrier, diluent or excipient. In one embodiment, the formulation is a liquid formulation. Preferably, the preparation is an injection. Preferably, 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.
在一个实施方式中,所述制剂可包括缓冲液诸如中性缓冲盐水、硫酸盐缓冲盐水等等;碳水化合物诸如葡萄糖、甘露糖、蔗糖或葡聚糖、甘露醇;蛋白质;多肽或氨基酸诸如甘氨酸;抗氧化剂;螯合剂诸如EDTA或谷胱甘肽;佐 剂(例如,氢氧化铝);和防腐剂。本发明的制剂优选配制用于静脉内施用。In one embodiment, the formulation may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine ; antioxidant; chelating agent such as EDTA or glutathione; adjuvant agents (e.g., aluminum hydroxide); and preservatives. The formulations of the present invention are preferably formulated for intravenous administration.
治疗性应用Therapeutic applications
本发明包括用编码本发明表达盒的逆转录病毒载体(RVV)转导的细胞(例如,T细胞)进行的治疗性应用。转导的T细胞可靶向肿瘤细胞的标志物(比如cMet、EGFR)蛋白,协同激活T细胞,引起细胞免疫应答,从而选择性杀伤肿瘤细胞,比如cMet和EGFR高表达的肿瘤细胞。The present invention encompasses the therapeutic use of cells (eg, T cells) transduced with a retroviral vector (RVV) encoding an expression cassette of the invention. The transduced T cells can target tumor cell marker proteins (such as cMet and EGFR), cooperatively activate T cells, trigger cellular immune responses, and thereby selectively kill tumor cells, such as tumor cells with high expression of cMet and EGFR.
因此,本发明也提供了刺激对哺乳动物的靶细胞群或组织的T细胞-介导的免疫应答的方法,其包括以下步骤:给哺乳动物施用本发明的CAR-T细胞。Therefore, the present invention also provides a method of stimulating a T cell-mediated immune response to a target cell population or tissue of a mammal, comprising the steps of administering a CAR-T cell of the present invention to the mammal.
在一个实施方式中,本发明包括一类细胞疗法,分离病人自体T细胞(或者异源供体),激活并进行基因改造产生CAR-T细胞,随后注入同一病人体内。这种方式患移植物抗宿主病概率极低,抗原被T细胞以无MHC限制方式识别。此外,一种CAR-T就可以治疗表达该抗原的所有癌症。不像抗体疗法,CAR-T细胞能够体内复制,产生可导致持续肿瘤控制的长期持久性。In one embodiment, the present invention includes a type of cell therapy in which a patient's autologous T cells (or allogeneic donors) are isolated, activated and genetically modified to produce CAR-T cells, and then injected into the same patient. This method has a very low probability of suffering from graft-versus-host disease, and the antigen is recognized by T cells in an MHC-free manner. In addition, one CAR-T can treat all cancers that express this antigen. Unlike antibody therapies, CAR-T cells are able to replicate in vivo, producing long-term persistence that can lead to sustained tumor control.
在一个实施方式中,本发明的CAR-T细胞可经历稳固的体内T细胞扩展并可持续延长的时间量。另外,CAR介导的免疫应答可为过继免疫疗法步骤的一部分,其中CAR-修饰T细胞诱导对CAR中的抗原结合结构域特异性的免疫应答。例如,肿瘤细胞的标志物(比如cMet、EGFR)的CAR-T细胞引起抗表达肿瘤细胞的标志物(比如cMet、EGFR)的细胞的特异性免疫应答。In one embodiment, CAR-T cells of the invention can undergo robust in vivo T cell expansion for an extended amount of time. Additionally, CAR-mediated immune responses can be part of an adoptive immunotherapy step, in which CAR-modified T cells induce an immune response specific for the antigen-binding domain in the CAR. For example, CAR-T cells expressing tumor cell markers (such as cMet, EGFR) elicit a specific immune response against cells expressing tumor cell markers (such as cMet, EGFR).
尽管本文公开的数据具体公开了包括靶向第一肿瘤细胞表面抗原的抗原结合域、铰链和跨膜区、和CD3ζ信号传导结构域、P2A、靶向第二肿瘤细胞表面抗原的抗原结合域、铰链和跨膜区、和4-1BB/CD28的逆转录病毒载体,但本发明应被解释为包括对构建体组成每一部分中的任意数量的变化。Although the data disclosed herein specifically disclose including an antigen-binding domain targeting a first tumor cell surface antigen, the hinge and transmembrane regions, and the CD3ζ signaling domain, P2A, an antigen-binding domain targeting a second tumor cell surface antigen, hinge and transmembrane regions, and 4-1BB/CD28 retroviral vectors, but the invention should be construed to include any number of changes in each part of the construct composition.
可治疗的癌症包括没有被血管化或基本上还没有被血管化的肿瘤,以及血管化的肿瘤。癌症可包括非实体瘤(诸如血液学肿瘤,例如白血病和淋巴瘤)或实体瘤。用本发明的CAR治疗的癌症类型包括但不限于癌、胚细胞瘤和肉瘤,和某些白血病或淋巴恶性肿瘤、良性和恶性肿瘤、恶性瘤,例如肉瘤、癌和黑素瘤。也包括成人肿瘤/癌症和儿童肿瘤/癌症。Treatable cancers include tumors that are not vascularized or substantially unvascularized, as well as tumors that are vascularized. Cancer may include non-solid tumors (such as hematological tumors, such as leukemias and lymphomas) or solid tumors. Cancer types treated with the CARs of the invention include, but are not limited to, carcinomas, blastomas, and sarcomas, and certain leukemias or lymphoid malignancies, benign and malignant tumors, and malignancies, such as sarcomas, carcinomas, and melanomas. Also includes adult neoplasms/cancers and pediatric neoplasms/cancers.
血液学癌症为血液或骨髓的癌症。血液学(或血原性)癌症的例子包括白血病,包括急性白血病(诸如急性淋巴细胞白血病、急性髓细胞白血病、急性骨髓性白血病和成髓细胞性、前髓细胞性、粒-单核细胞型、单核细胞性和红白 血病)、慢性白血病(诸如慢性髓细胞(粒细胞性)白血病、慢性骨髓性白血病和慢性淋巴细胞白血病)、真性红细胞增多症、淋巴瘤、霍奇金氏疾病、非霍奇金氏淋巴瘤(无痛和高等级形式)、多发性骨髓瘤、瓦尔登斯特伦氏巨球蛋白血症、重链疾病、骨髓增生异常综合征、多毛细胞白血病和脊髓发育不良。Hematologic cancers are cancers of the blood or bone marrow. Examples of hematological (or hematogenous) cancers include leukemias, including acute leukemias such as acute lymphoblastic leukemia, acute myeloid leukemia, acute myeloid leukemia, and myeloblastoid, promyelocytic, myelomonocytic types , monocytic and red and white blood diseases), chronic leukemias (such as chronic myeloid (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, lymphoma, Hodgkin's disease, non-Hodgkin's lymphoma (indolent and high-grade forms), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disorders, myelodysplastic syndromes, hairy cell leukemia, and myelodysplasia.
实体瘤为通常不包含囊肿或液体区的组织的异常肿块。实体瘤可为良性或恶性的。不同类型的实体瘤以形成它们的细胞类型命名(诸如肉瘤、癌和淋巴瘤)。实体瘤诸如肉瘤和癌的例子包括前列腺癌、肝癌、纤维肉瘤、粘液肉瘤、脂肪肉瘤间皮瘤、淋巴恶性肿瘤、胰腺癌、卵巢癌。Solid tumors are abnormal masses of tissue that usually do not contain cysts or areas of fluid. Solid tumors can be benign or malignant. Different types of solid tumors are named after the cell types that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors such as sarcomas and carcinomas include prostate cancer, liver cancer, fibrosarcoma, myxosarcoma, liposarcoma mesothelioma, lymphoid malignancies, pancreatic cancer, ovarian cancer.
本发明的CAR-修饰T细胞也可用作对哺乳动物离体免疫和/或体内疗法的疫苗类型。优选地,哺乳动物为人。The CAR-modified T cells of the present invention may also be used as a type of vaccine for ex vivo immunization and/or in vivo therapy of mammals. Preferably, the mammal is human.
对于离体免疫,以下中的至少一项在将细胞施用进入哺乳动物前在体外发生:i)扩增细胞,ii)将编码CAR的核酸引入细胞,和/或iii)冷冻保存细胞。For ex vivo immunization, at least one of the following occurs in vitro prior to administration of the cells into the mammal: i) expanding the cells, ii) introducing the CAR-encoding nucleic acid into the cells, and/or iii) cryopreserving the cells.
离体程序在本领域中是公知的,并在以下更完全地进行讨论。简单地说,细胞从哺乳动物(优选人)中分离并用表达本文公开的CAR的载体进行基因修饰(即,体外转导或转染)。CAR-修饰的细胞可被施用给哺乳动物接受者,以提供治疗益处。哺乳动物接受者可为人,和CAR-修饰的细胞可相对于接受者为自体的。可选地,细胞可相对于接受者为同种异基因的、同基因的(syngeneic)或异种的。Ex vivo procedures are well known in the art and are discussed more fully below. Briefly, cells are isolated from a mammal (preferably human) and genetically modified (i.e., transduced or transfected in vitro) with a vector expressing a CAR disclosed herein. CAR-modified cells can be administered to mammalian recipients to provide therapeutic benefit. The mammalian recipient can be human, and the CAR-modified cells can be autologous to the recipient. Alternatively, the cells may be allogeneic, syngeneic, or xenogeneic relative to the recipient.
除了就离体免疫而言使用基于细胞的疫苗之外,本发明也提供了体内免疫以引起针对患者中抗原的免疫应答的组合物和方法。In addition to the use of cell-based vaccines for ex vivo immunization, the present invention also provides compositions and methods for in vivo immunization to elicit an immune response against an antigen in a patient.
本发明提供了治疗肿瘤的方法,其包括施用给需要其的对象治疗有效量的本发明的CAR-修饰的T细胞。The invention provides methods of treating tumors comprising administering to a subject in need thereof a therapeutically effective amount of a CAR-modified T cell of the invention.
本发明的CAR-修饰的T细胞可被单独施用或作为药物组合物与稀释剂和/或与其他组分或其他细胞因子或细胞群结合施用。简单地说,本发明的药物组合物可包括如本文所述的靶细胞群,与一种或多种药学或生理学上可接受载体、稀释剂或赋形剂结合。这样的组合物可包括缓冲液诸如中性缓冲盐水、硫酸盐缓冲盐水等等;碳水化合物诸如葡萄糖、甘露糖、蔗糖或葡聚糖、甘露醇;蛋白质;多肽或氨基酸诸如甘氨酸;抗氧化剂;螯合剂诸如EDTA或谷胱甘肽;佐剂(例如,氢氧化铝);和防腐剂。本发明的组合物优选配制用于静脉内施用。The CAR-modified T cells of the invention can be administered alone or as a pharmaceutical composition in combination with diluents and/or with other components or other cytokines or cell populations. Briefly, a pharmaceutical composition of the present invention may comprise a target cell population as described herein, in combination with one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions may include buffers such as neutral buffered saline, sulfate buffered saline, and the like; carbohydrates such as glucose, mannose, sucrose or dextran, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelates Adjuvants such as EDTA or glutathione; adjuvants (eg, aluminum hydroxide); and preservatives. The compositions of the present invention are preferably formulated for intravenous administration.
本发明的药物组合物可以以适于待治疗(或预防)的疾病的方式施用。施用的数量和频率将由这样的因素确定,如患者的病症、和患者疾病的类型和严重 度——尽管适当的剂量可由临床试验确定。The pharmaceutical compositions of the present invention may be administered in a manner suitable for the disease to be treated (or prevented). The amount and frequency of administration will be determined by factors such as the patient's condition, and the type and severity of the patient's condition. Degree—Although appropriate dosage can be determined by clinical trials.
当指出“免疫学上有效量”、“抗肿瘤有效量”、“肿瘤-抑制有效量”或“治疗量”时,待施用的本发明组合物的精确量可由医师确定,其考虑患者(对象)的年龄、重量、肿瘤大小、感染或转移程度和病症的个体差异。可通常指出:包括本文描述的T细胞的药物组合物可以以104至109个细胞/kg体重的剂量,优选105至106个细胞/kg体重的剂量(包括那些范围内的所有整数值)施用。T细胞组合物也可以以这些剂量多次施用。细胞可通过使用免疫疗法中公知的注入技术(见例如Rosenberg等,NewEng.J.of Med.319:1676,1988)施用。对于具体患者的最佳剂量和治疗方案可通过监测患者的疾病迹象并因此调节治疗由医学领域技术人员容易地确定。When an "immunologically effective amount", "anti-tumor effective amount", "tumor-suppressive effective amount" or "therapeutic amount" is indicated, the precise amount of the composition of the invention to be administered can be determined by the physician, who takes into account the patient (subject) ) age, weight, tumor size, degree of infection or metastasis, and individual differences in disease. It may generally be stated that pharmaceutical compositions comprising T cells described herein may be administered at a dose of 10 4 to 10 9 cells/kg body weight, preferably 10 5 to 10 6 cells/kg body weight (including all integers within those ranges). value) application. T cell compositions can also be administered multiple times at these dosages. Cells can be administered using infusion techniques well known in immunotherapy (see, eg, Rosenberg et al., New Eng. J. of Med. 319:1676, 1988). The optimal dosage and treatment regimen for a particular patient can be readily determined by one skilled in the medical field by monitoring the patient for signs of disease and adjusting treatment accordingly.
对象组合物的施用可以以任何方便的方式进行,包括通过喷雾法、注射、吞咽、输液、植入或移植。本文描述的组合物可被皮下、皮内、瘤内、结内、脊髓内、肌肉内、通过静脉内(i.v.)注射或腹膜内施用给患者。在一个实施方式中,本发明的T细胞组合物通过皮内或皮下注射被施用给患者。在另一个实施方式中,本发明的T细胞组合物优选通过i.v.注射施用。T细胞的组合物可被直接注入肿瘤,淋巴结或感染位置。Administration of subject compositions may be in any convenient manner, including by spraying, injection, swallowing, infusion, implantation or transplantation. The compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intraspinally, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one embodiment, the T cell composition of the invention is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the T cell composition of the invention is preferably administered by i.v. injection. The composition of T cells can be injected directly into the tumor, lymph node or site of infection.
在本发明的某些实施方式中,利用本文描述的方法或本领域已知的其他将T细胞扩展至治疗性水平的方法活化和扩展的细胞,与任何数量的有关治疗形式结合(例如,之前、同时或之后)施用给患者,所述治疗形式包括但不限于用以下试剂进行治疗:所述试剂诸如抗病毒疗法、西多福韦和白细胞介素-2、阿糖胞苷(也已知为ARA-C)或对MS患者的那他珠单抗治疗或对牛皮癣患者的厄法珠单抗治疗或对PML患者的其他治疗。在进一步的实施方式中,本发明的T细胞可与以下结合使用:化疗、辐射、免疫抑制剂,诸如,环孢菌素、硫唑嘌呤、甲氨喋呤、麦考酚酯和FK506,抗体或其他免疫治疗剂。在进一步的实施方式中,本发明的细胞组合物与骨髓移植、利用化疗剂诸如氟达拉滨、外部光束放射疗法(XRT)、环磷酰胺结合(例如,之前、同时或之后)而施用给患者。例如,在一个实施方式中,对象可经历高剂量化疗的标准治疗,之后进行外周血干细胞移植。在一些实施方式中,在移植后,对象接受本发明的扩展的免疫细胞的注入。在一个额外的实施方式中,扩展的细胞在外科手术前或外科手术后施用。In certain embodiments of the invention, 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., before , simultaneously or subsequently) administered to a patient, such forms of treatment include, but are not limited to, treatment with agents such as antiviral therapy, cidofovir and interleukin-2, cytarabine (also known as for ARA-C) or natalizumab treatment in patients with MS or elfalizumab treatment in patients with psoriasis or other treatments in patients with PML. In further embodiments, the T cells of the invention can be used in combination with chemotherapy, radiation, immunosuppressants such as cyclosporine, azathioprine, methotrexate, mycophenolate mofetil and FK506, antibodies or other immunotherapeutic agents. In further embodiments, the cellular compositions of the invention are administered in conjunction with (eg, before, simultaneously with, or after) bone marrow transplantation, use of a chemotherapeutic agent such as fludarabine, external beam radiation therapy (XRT), cyclophosphamide patient. For example, in one embodiment, a subject may undergo standard treatment with high-dose chemotherapy followed by a peripheral blood stem cell transplant. In some embodiments, following transplantation, the subject receives an infusion of expanded immune cells of the invention. In an additional embodiment, the expanded cells are administered before or after surgery.
施用给患者的以上治疗的剂量将随着治疗病症的精确属性和治疗的接受者而变化。人施用的剂量比例可根据本领域接受的实践实施。通常,每次治疗 或每个疗程,可将1×106个至1×1010个本发明经修饰的T细胞(如,本发明的CAR-T细胞),通过例如静脉回输的方式,施用于患者。The dosage of the above treatments administered to a patient will vary depending on the precise nature of the condition being treated and the recipient of the treatment. Dosage proportions for human administration may be implemented in accordance with art-accepted practice. Typically, each treatment Or for each course of treatment, 1×10 6 to 1×10 10 modified T cells of the present invention (eg, CAR-T cells of the present invention) can be administered to the patient, for example, by intravenous infusion.
融合蛋白fusion protein
如本文所用,术语“融合蛋白”、“本发明融合蛋白”、和“本发明的多肽”、“logic BiCAR”具有相同的含义,均具有本发明第八方面所述的结构。As used herein, the terms "fusion protein", "fusion protein of the present invention", "polypeptide of the present invention" and "logic BiCAR" have the same meaning, and all have the structure described in the eighth aspect of the present invention.
在另一优选例中,所述融合蛋白的氨基酸序列如SEQ ID NO.:23-26中任一所示。In another preferred embodiment, the amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
如本文所用,术语“融合蛋白”还包括具有上述活性的、SEQ ID NO.:23-26中任一所示序列的变异形式。这些变异形式包括(但并不限于):1-3个(通常为1-2个,更佳地1个)氨基酸的缺失、插入和/或取代,以及在C末端和/或N末端添加或缺失一个或数个(通常为3个以内,较佳地为2个以内,更佳地为1个以内)氨基酸。例如,在本领域中,用性能相近或相似的氨基酸进行取代时,通常不会改变蛋白质的功能。又比如,在C末端和/或N末端添加或缺失一个或数个氨基酸通常也不会改变蛋白质的结构和功能。此外,所述术语还包括单体和多聚体形式的本发明多肽。该术语还包括线性以及非线性的多肽(如环肽)。As used herein, the term "fusion protein" also includes variant forms of any of the sequences shown in SEQ ID NO.: 23-26 having the above-mentioned activities. These variant forms include (but are not limited to): deletion, insertion and/or substitution of 1-3 (usually 1-2, more preferably 1) amino acids, and addition of or One or several (usually within 3, preferably within 2, and more preferably within 1) amino acids are missing. For example, in the art, substitutions with amino acids with similar or similar properties generally do not alter the function of the protein. For another example, 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. Furthermore, the term also includes monomeric and multimeric forms of the polypeptides of the invention. The term also includes linear as well as non-linear polypeptides (such as cyclic peptides).
本发明还包括上述融合蛋白的活性片段、衍生物和类似物。如本文所用,术语“片段”、“衍生物”和“类似物”是指基本上保持本发明融合蛋白的功能或活性的多肽。本发明的多肽片段、衍生物或类似物可以是(i)有一个或几个保守或非保守性氨基酸残基(优选保守性氨基酸残基)被取代的多肽,或(ii)在一个或多个氨基酸残基中具有取代基团的多肽,或(iii)多肽与另一个化合物(比如延长多肽半衰期的化合物,例如聚乙二醇)融合所形成的多肽,或(iv)附加的氨基酸序列融合于此多肽序列而形成的多肽(与前导序列、分泌序列或6His等标签序列融合而形成的融合蛋白)。根据本文的教导,这些片段、衍生物和类似物属于本领域熟练技术人员公知的范围。The present invention also includes active fragments, derivatives and analogs of the above-mentioned fusion proteins. As used herein, the terms "fragment," "derivative" and "analogue" refer to polypeptides that substantially retain the function or activity of the fusion proteins of the invention. The polypeptide fragments, derivatives or analogs of the present invention may be (i) a polypeptide in which one or several conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted, or (ii) in which one or more conservative amino acid residues are substituted. A polypeptide with a substituent group in an amino acid residue, or (iii) a polypeptide formed by fusion of a polypeptide with another compound (such as a compound that extends the half-life of the polypeptide, such as polyethylene glycol), or (iv) a fusion of additional amino acid sequences A polypeptide formed from this polypeptide sequence (a fusion protein formed by fusion with a leader sequence, secretory sequence or tag sequence such as 6His). Such fragments, derivatives and analogs are within the scope of those skilled in the art in light of the teachings herein.
一类优选的活性衍生物指与本发明的氨基酸序列相比,有至多3个,较佳地至多2个,更佳地至多1个氨基酸被性质相似或相近的氨基酸所替换而形成多肽。这些保守性变异多肽最好根据表1进行氨基酸替换而产生。A preferred type of active derivative refers to one in which at most 3, preferably at most 2, and more preferably at most 1 amino acid is replaced by an amino acid with similar or similar properties compared to the amino acid sequence of the present invention to form a polypeptide. These conservative variant polypeptides are preferably produced by amino acid substitutions according to Table 1.
表1

Table 1

本发明还提供本发明融合蛋白的类似物。这些类似物与SEQ ID NO.:23-26中任一所示的多肽的差别可以是氨基酸序列上的差异,也可以是不影响序列的修饰形式上的差异,或者兼而有之。类似物还包括具有不同于天然L-氨基酸的残基(如D-氨基酸)的类似物,以及具有非天然存在的或合成的氨基酸(如β、γ-氨基酸)的类似物。应理解,本发明的多肽并不限于上述例举的代表性的多肽。The invention also provides analogs of the fusion proteins of the invention. The difference between these analogs and the polypeptides shown in any of SEQ ID NO.: 23-26 can be differences in amino acid sequences, differences in modified forms that do not affect the sequence, or both. Analogues also include analogs with residues that differ from natural L-amino acids (eg, D-amino acids), as well as analogs with non-naturally occurring or synthetic amino acids (eg, beta, gamma-amino acids). It should be understood that the polypeptides of the present invention are not limited to the representative polypeptides exemplified above.
修饰(通常不改变一级结构)形式包括:体内或体外的多肽的化学衍生形式如乙酰化或羧基化。修饰还包括糖基化,如那些在多肽的合成和加工中或进一步加工步骤中进行糖基化修饰而产生的多肽。这种修饰可以通过将多肽暴露于进行糖基化的酶(如哺乳动物的糖基化酶或去糖基化酶)而完成。修饰形式还包括具有磷酸化氨基酸残基(如磷酸酪氨酸,磷酸丝氨酸,磷酸苏氨酸)的序列。还包括被修饰从而提高了其抗蛋白水解性能或优化了溶解性能的多肽。Modified forms (which generally do not alter the primary structure) include chemically derivatized forms of the polypeptide, such as acetylation or carboxylation, either in vivo or in vitro. Modifications also include glycosylation, such as those resulting from glycosylation modifications of the polypeptide during its synthesis and processing 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 having phosphorylated amino acid residues (eg, phosphotyrosine, phosphoserine, phosphothreonine). Also included are polypeptides that have been modified to increase their resistance to proteolysis or to optimize solubility properties.
在本发明的一个实施方式中,所述融合蛋白的氨基酸序列如SEQ ID NO.:23-26中任一所示。In one embodiment of the present invention, the amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
编码序列coding sequence
本发明还涉及编码根据本发明的融合蛋白的多核苷酸。The invention also relates to polynucleotides encoding fusion proteins according to the invention.
本发明的多核苷酸可以是DNA形式或RNA形式。DNA可以是编码链或非编码链。 编码成熟多肽的编码区序列可以与编码SEQ ID NO.:23-26中任一所示的多肽的序列相同或者是简并的变异体。如本文所用,“简并的变异体”在本发明中是指编码具有SEQ ID NO.:23-26中任一所示的多肽,但相应编码区序列有差别的核酸序列。The polynucleotides of the invention may be in DNA form or RNA form. DNA can be a coding strand or a non-coding strand. The sequence of the coding region encoding the mature polypeptide may be identical to the sequence encoding the polypeptide shown in any one of SEQ ID NO.: 23-26 or may be a degenerate variant. As used herein, "degenerate variant" in the present invention refers to a nucleic acid sequence encoding a polypeptide shown in any one of SEQ ID NO.: 23-26, but with different sequences in the corresponding coding regions.
本发明的核苷酸全长序列或其片段通常可以用PCR扩增法、重组法或人工合成的方法获得。目前,已经可以完全通过化学合成来得到编码本发明多肽(或其片段,或其衍生物)的DNA序列。然后可将该DNA序列引入本领域中已知的各种现有的DNA分子(或如载体)和细胞中。The full-length nucleotide sequence of the present invention or its fragment can usually be obtained by PCR amplification, recombination or artificial synthesis. At present, the DNA sequence encoding the polypeptide of the present invention (or its fragment, or its derivative) can be obtained entirely through chemical synthesis. The DNA sequence can then be introduced into a variety of existing DNA molecules (or vectors) and cells known in the art.
本发明也涉及包含本发明的多核苷酸的载体,以及用本发明的载体或多肽编码序列经基因工程产生的宿主细胞。上述多核苷酸、载体或宿主细胞可以是分离的。The invention also relates to vectors comprising the polynucleotides of the invention, as well as host cells genetically engineered with the vectors or polypeptide coding sequences of the invention. The polynucleotides, vectors or host cells described above may be isolated.
如本文所用,“分离的”是指物质从其原始环境中分离出来(如果是天然的物质,原始环境即是天然环境)。如活体细胞内的天然状态下的多核苷酸和多肽是没有分离纯化的,但同样的多核苷酸或多肽如从天然状态中同存在的其他物质中分开,则为分离纯化的。As used herein, "isolated" means that a substance has been separated from its original environment (in the case of a natural substance, the original environment is the natural environment). For example, polynucleotides and polypeptides in the natural state within living cells are not isolated and purified, but the same polynucleotide or polypeptide is isolated and purified if it is separated from other substances existing in the natural state.
本发明的多核苷酸可以是DNA形式或RNA形式。DNA形式包括cDNA、基因组DNA或人工合成的DNA。DNA可以是单链的或是双链的。DNA可以是编码链或非编码链。The polynucleotides of the invention may be in DNA form or RNA form. Forms of DNA include 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.
本发明还涉及上述多核苷酸的变异体,其编码与本发明有相同的氨基酸序列的蛋白质片段、类似物和衍生物。此多核苷酸的变异体可以是天然发生的等位变异体或非天然发生的变异体。这些核苷酸变异体包括取代变异体、缺失变异体和插入变异体。如本领域所知的,等位变异体是一个多核苷酸的替换形式,它可能是一个或多个核苷酸的取代、缺失或插入,但不会从实质上改变其编码本发明融合蛋白的功能。The present invention also relates to variants of the above-mentioned polynucleotides, which encode protein fragments, analogs and derivatives having the same amino acid sequence as the present invention. Variants of this polynucleotide may be naturally occurring allelic variants or non-naturally occurring variants. These nucleotide variants include substitution variants, deletion variants, and insertion variants. As known in the art, an allelic variant is an alternative form of a polynucleotide, which may be the substitution, deletion or insertion of one or more nucleotides, but does not substantially change its encoding of the fusion protein of the invention. function.
编码本发明的融合蛋白的核苷酸全长序列或其片段通常可以用PCR扩增法、重组法或人工合成的方法获得。对于PCR扩增法,可根据已公开的有关核苷酸序列,尤其是开放阅读框序列来设计引物,并用市售的cDNA库或按本领域技术人员已知的常规方法所制备的cDNA库作为模板,扩增而得有关序列。当序列较长时,常常需要进行两次或多次PCR扩增,然后再将各次扩增出的片段按正确次序拼接在一起。The full-length nucleotide sequence encoding the fusion protein of the present invention or its fragment can usually be obtained by PCR amplification, recombination or artificial synthesis. For the PCR amplification method, primers can be designed based on the relevant published nucleotide sequences, especially the open reading frame sequence, and a commercially available cDNA library or a cDNA library prepared according to conventional methods known to those skilled in the art can be used as the primer. Template, amplified to obtain the relevant sequence. When 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.
在本发明的一个实施方式中,所述融合蛋白的编码多核苷酸序列如SEQ ID NO.:27-30中任一所示。 In one embodiment of the present invention, the polynucleotide sequence encoding the fusion protein is as shown in any one of SEQ ID NO.: 27-30.
一旦获得了有关的序列,就可以用重组法来大批量地获得有关序列。这通常是将其克隆入载体,再转入细胞,然后通过常规方法从增殖后的宿主细胞中分离得到有关序列。Once the relevant sequence is obtained, recombination can be used to obtain the relevant sequence in large quantities. This is usually done by cloning it into a vector, transforming it into cells, and then isolating the relevant sequence from the propagated host cells by conventional methods.
此外,还可用人工合成的方法来合成有关序列,尤其是片段长度较短时。通常,通过先合成多个小片段,然后再进行连接可获得序列很长的片段。In addition, artificial synthesis methods can also be used to synthesize relevant sequences, especially when the fragment length is short. Often, fragments with long sequences are obtained by first synthesizing multiple small fragments and then ligating them.
应用PCR技术扩增DNA/RNA的方法被优选用于获得本发明的基因。用于PCR的引物可根据本文所公开的本发明的序列信息适当地选择,并可用常规方法合成。可用常规方法如通过凝胶电泳分离和纯化扩增的DNA/RNA片段。The method of amplifying DNA/RNA using PCR technology is preferably used to obtain the gene of the present invention. Primers for PCR can be appropriately selected based on 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 using conventional methods such as by gel electrophoresis.
本发明也涉及包含本发明的多核苷酸的载体,以及用本发明的载体或蛋白编码序列经基因工程产生的宿主细胞,以及经重组技术在所述NK细胞上表达本发明融合蛋白的方法。The present invention also relates to vectors containing the polynucleotides of the present invention, as well as host cells genetically engineered using the vectors or protein coding sequences of the present invention, and methods for expressing the fusion proteins of the present invention on the NK cells using recombinant technology.
通过常规的重组DNA技术,可利用本发明的多核苷酸序列获得表达本发明融合蛋白的NK细胞。一般来说包括步骤:将本发明所述的第一表达盒和/或第二表达盒转导入NK细胞内,从而获得所述NK细胞。Through conventional recombinant DNA technology, the polynucleotide sequence of the present invention can be used to obtain NK cells expressing the fusion protein of the present invention. Generally speaking, it includes the steps of: transducing the first expression cassette and/or the second expression cassette of the present invention into NK cells, thereby obtaining the NK cells.
本领域的技术人员熟知的方法能用于构建含本发明融合蛋白的编码DNA序列和合适的转录/翻译控制信号的表达载体。这些方法包括体外重组DNA技术、DNA合成技术、体内重组技术等。所述的DNA序列可有效连接到表达载体中的适当启动子上,以指导mRNA合成。表达载体还包括翻译起始用的核糖体结合位点和转录终止子。Methods well known to those skilled in the art can be used to construct expression vectors containing the DNA sequence encoding the fusion protein of the invention and appropriate transcription/translation control signals. These methods include in vitro recombinant DNA technology, DNA synthesis technology, in vivo recombination technology, etc. The DNA sequence can be effectively linked to an appropriate promoter in an expression vector to direct mRNA synthesis. The expression vector also includes a ribosome binding site for translation initiation and a transcription terminator.
此外,表达载体优选地包含一个或多个选择性标记基因,以提供用于选择转化的宿主细胞的表型性状,如真核细胞培养用的二氢叶酸还原酶、新霉素抗性以及绿色荧光蛋白(GFP),或用于大肠杆菌的四环素或氨苄青霉素抗性。In addition, 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, neomycin resistance, and green color for eukaryotic cell culture. Fluorescent protein (GFP), or for tetracycline or ampicillin resistance in E. coli.
包含上述的适当DNA序列以及适当启动子或者控制序列的载体,可以用于转化适当的宿主细胞,以使其能够表达蛋白质。Vectors containing appropriate DNA sequences as described above and appropriate promoter or control sequences can be used to transform appropriate host cells to enable expression of proteins.
宿主细胞可以是原核细胞,如细菌细胞;或是低等真核细胞,如酵母细胞;或是高等真核细胞,如哺乳动物细胞。代表性例子有:大肠杆菌,枯草芽胞杆菌,链霉菌属的细菌细胞;真菌细胞如毕赤酵母、酿酒酵母细胞;植物细胞;果蝇S2或Sf9的昆虫细胞;CHO、NS0、COS7、或293细胞的动物细胞等。在本发明的一个优选实施方式中,选择NK细胞为宿主细胞。The host cell can be a prokaryotic cell, such as a bacterial cell; a lower eukaryotic cell, such as a yeast cell; or a higher eukaryotic cell, such as a mammalian cell. Representative examples include: bacterial cells of Escherichia coli, Bacillus subtilis, and Streptomyces; fungal cells such as Pichia pastoris and Saccharomyces cerevisiae cells; plant cells; insect cells of Drosophila S2 or Sf9; CHO, NS0, COS7, or 293 cells animal cells etc. In a preferred embodiment of the present invention, NK cells are selected as host cells.
用重组DNA转化宿主细胞可用本领域技术人员熟知的常规技术进行。当宿主为原核生物如大肠杆菌时,能吸收DNA的感受态细胞可在指数生长期后收获,用CaCl2 法处理,所用的步骤在本领域众所周知。另一种方法是使用MgCl2。如果需要,转化也可用电穿孔的方法进行。当宿主是真核生物,可选用如下的DNA转染方法:磷酸钙共沉淀法,常规机械方法如显微注射、电穿孔、脂质体包装等。Transformation of host cells with recombinant DNA can be performed using conventional techniques well known to those skilled in the art. When the host is a prokaryotic organism such as E. coli, competent cells capable of absorbing DNA can be harvested after the exponential growth phase and treated with CaCl 2 The procedures used are well known in the art. Another method is to use MgCl 2 . If necessary, transformation can also be performed by electroporation. When the host is a eukaryotic organism, the following DNA transfection methods can be used: calcium phosphate co-precipitation method, conventional mechanical methods such as microinjection, electroporation, liposome packaging, etc.
获得的转化子可以用常规方法培养,表达本发明的基因所编码的蛋白质。根据所用的宿主细胞,培养中所用的培养基可选自各种常规培养基。在适于宿主细胞生长的条件下进行培养。当宿主细胞生长到适当的细胞密度后,用合适的方法(如温度转换或化学诱导)诱导选择的启动子,将细胞再培养一段时间。The obtained transformants can be cultured using conventional methods to express the protein encoded by the gene of the present invention. Depending on the host cells used, the medium used in culture can be selected from various conventional media. Cultivate under conditions suitable for host cell growth. After the host cells grow to an appropriate cell density, the selected promoter is induced using an appropriate method (such as temperature shift or chemical induction), and the cells are cultured for a further period of time.
在上面的方法中的蛋白质可在细胞内、或在细胞膜上表达、或分泌到细胞外。如果需要,可利用其物理的、化学的和其它特性通过各种分离方法分离和纯化蛋白。这些方法是本领域技术人员所熟知的。这些方法的例子包括但并不限于:常规的复性处理、用蛋白沉淀剂处理(盐析方法)、离心、渗透破菌、超处理、超离心、分子筛层析(凝胶过滤)、吸附层析、离子交换层析、高效液相层析(HPLC)和其它各种液相层析技术及这些方法的结合。The protein in the above method can be expressed within the cell, on the cell membrane, or secreted outside the cell. If desired, proteins can be isolated and purified by various separation methods using their physical, chemical and other properties. These methods are well known to those skilled in the art. Examples of these methods include, but are not limited to: conventional refolding treatment, treatment with protein precipitating agents (salting out method), centrifugation, osmotic sterilization, ultratreatment, ultracentrifugation, 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.
本发明的主要优点包括:The main advantages of the present invention include:
1.本发明首次发现含有靶向第一肿瘤细胞标志物(比如cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47)的第一CAR和靶向第二肿瘤细胞标志物(比如,EGFR、EpCAM、Her2、Her3)的第二CAR的工程化免疫细胞可被充分激活,增强靶向特异性,发挥抗肿瘤功效,并且降低在靶/脱肿瘤(on target/off tumor)毒性,提高细胞治疗的安全性。1. The present invention discovered for the first time a first CAR targeting a first tumor cell marker (such as cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47) and a second tumor cell marker (such as EGFR , EpCAM, Her2, Her3), the engineered immune cells of the second CAR can be fully activated, enhance target specificity, exert anti-tumor efficacy, and reduce on-target/off-tumor toxicity and improve cell Safety of treatment.
2.本发明首次发现,本发明的免疫细胞能同时识别EGFR和cMet这2个不同的抗原,增强CAR T细胞靶向肿瘤的特异性和选择性,以及在肿瘤中的归巢。另外,可同时抑制两个致病信号通路,从而抑制疾病进展,提高生存期。2. The present invention discovered for the first time that the immune cells of the present invention can simultaneously recognize two different antigens, EGFR and cMet, and enhance the specificity and selectivity of CAR T cells in targeting tumors, as well as their homing in tumors. In addition, it can inhibit two pathogenic signaling pathways at the same time, thereby inhibiting disease progression and improving survival.
下面结合具体实施,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。The present invention will be further described below in conjunction with specific implementations. It should be understood that these examples are only used to illustrate the invention and are not intended to limit the scope of the invention. Experimental methods without specifying specific conditions in the following examples usually follow conventional conditions, such as the conditions described in Sambrook et al., Molecular Cloning: Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989), or according to manufacturing Conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight.
除非特别说明,否则本发明实施例中所用材料和试剂均为市售产品。 Unless otherwise stated, the materials and reagents used in the examples of the present invention are commercially available products.
通用方法general method
逆转录病毒颗粒制备Retroviral particle preparation
将处于对数生长期的Phoenix GP细胞以4E6/flask的密度接种至T75培养瓶中,37℃、5%CO2培养箱中培养过夜后用于转染,培养基为含有10%FBS的DMEM培养基。次日,于转染前更换为新鲜的DMEM培养基。转染步骤如下:将目的基因质粒例如pMSGV-CAR,与包膜质粒pMD2.G,加入Opti-MEM培养基中混匀;再加入DNA转染试剂混匀;逐滴加入培养瓶中,培养18h;更换含有10%FBS的DMEM培养基;72h后收集上清,离心、过滤、分装,-80℃保存备用或者直接感染PG13细胞。将处于对数生长期的PG13细胞以7E4/well的密度接种至6孔板中,37℃、5%CO2培养箱中培养过夜后用于转染,培养基为含有10%FBS的DMEM培养基。次日,将6孔板中的培养基更换为前述过滤的培养基上清同时加入病毒感染增强剂Polybrene。培养24h后更换为新鲜的DMEM培养基。培养72h后,检测PG13-CAR的转导效率构建PG13-CAR逆转录病毒生产细胞系,扩增培养后冻存保藏。将处于对数生长期的PG13-CAR细胞以2E7/flask的密度接种至T75培养瓶中,37℃、5%CO2培养箱培养24h,收集含有逆转录病毒的上清,离心、过滤后感染细胞。Phoenix GP cells in the logarithmic growth phase were seeded into T75 culture flasks at a density of 4E6/flask, and cultured overnight in a 37°C, 5% CO2 incubator for transfection. The culture medium was DMEM containing 10% FBS. culture medium. The next day, the culture medium was replaced with fresh DMEM before transfection. The transfection steps are as follows: Add the target gene plasmid, such as pMSGV-CAR, and the envelope plasmid pMD2.G to the Opti-MEM medium and mix well; then add DNA transfection reagent and mix well; add it dropwise into the culture bottle and culture for 18 hours. ; Replace the DMEM medium containing 10% FBS; collect the supernatant after 72 hours, centrifuge, filter, aliquot, store at -80°C for later use or directly infect PG13 cells. PG13 cells in the logarithmic growth phase were seeded into a 6-well plate at a density of 7E4/well and cultured overnight in a 37°C, 5% CO2 incubator for transfection. The culture medium was DMEM containing 10% FBS. base. The next day, the culture medium in the 6-well plate was replaced with the aforementioned filtered culture supernatant and the virus infection enhancer Polybrene was added. After 24 h of culture, the culture medium was replaced with fresh DMEM. After 72 hours of culture, the transduction efficiency of PG13-CAR was detected to construct a PG13-CAR retrovirus production cell line, which was amplified and cultured before cryopreservation. PG13-CAR cells in the logarithmic growth phase were inoculated into T75 culture bottles at a density of 2E7/flask, and cultured in a 37°C, 5% CO2 incubator for 24 hours. The supernatant containing retrovirus was collected, centrifuged, filtered, and infected. cell.
CAR T细胞制备CAR T cell preparation
通过Ficoll-Hypaque密度梯度离心法由健康人的外周血中分离获得单个核细胞。淋巴细胞培养基中加入TransAct激活剂和重组人IL-2进行T细胞的刺激活化和扩增培养;同时,使用Retronectin包被6孔板,将逆转录病毒上清加入到6孔板中离心;离心完成后,弃去上清,将活化后的T细胞加入到6孔板中离心进行转导;在37℃、5%CO2培养箱中培养24h后将T细胞从6孔板转移到培养瓶中进行扩增培养。Mononuclear cells were isolated from peripheral blood of healthy individuals by Ficoll-Hypaque density gradient centrifugation. Add TransAct activator and recombinant human IL-2 to the lymphocyte culture medium to stimulate, activate and expand T cells; at the same time, use Retronectin to coat a 6-well plate, add the retrovirus supernatant to the 6-well plate and centrifuge; After centrifugation is completed, discard the supernatant, add the activated T cells to the 6-well plate, and centrifuge for transduction; after culturing for 24 hours in a 37°C, 5% CO2 incubator, transfer the T cells from the 6-well plate to the culture chamber. Amplification culture was carried out in bottles.
流式细胞术检测CAR分子表达Detection of CAR molecule expression by flow cytometry
当CAR T细胞培养至第7天时,取1E6细胞,缓冲液洗涤细胞2次;添加1ug MET-His、1ug EGFR-Fc抗原于细胞悬液中4℃孵育30min;缓冲液洗涤细胞2次后添加Anti-His-PE和Allophycocyanin AffiniPure Goat Anti-Human IgG抗体于细胞悬液中4℃避光孵育30min;洗涤细胞2次后使用流式细胞术检 测CAR分子的表达。When the CAR T cells are cultured to the 7th day, take 1E6 cells and wash the cells twice with buffer; add 1ug MET-His and 1ug EGFR-Fc antigen in the cell suspension and incubate for 30 minutes at 4°C; wash the cells twice with buffer and then add Anti-His-PE and Allophycocyanin AffiniPure Goat Anti-Human IgG antibodies were incubated in the cell suspension at 4°C in the dark for 30 minutes; the cells were washed twice and then analyzed by flow cytometry. Measure the expression of CAR molecules.
通过HiBiT标签蛋白检测法(HiBiT Extracellular Detection System,Promega)测定细胞毒性Determination of cytotoxicity by HiBiT tag protein detection method (HiBiT Extracellular Detection System, Promega)
收集处于对数生长期的携带HaloTag-HiBiT标签的靶细胞(包括NCI-H1975-Halotag-HiBiT、EBC-1-HaloTag-HiBiT)和培养至第7天的CAR T细胞,制备不同细胞浓度的细胞悬液;靶细胞按照5000cells/well接种于96孔板中(3复孔),然后按照不同的效靶比加入CAR T细胞悬液,37℃、5%CO2培养箱中培养24h。最大释放的阳性对照组中加入一定终浓度的digitonin孵育30min,随后将孔板从培养箱中取出,待恢复至室温后,加入等体积的预先配制的检测Buffer(含有1:100稀释的LgBiT Protein和1:50稀释的HiBiT Extracellular Substrate),摇床上混匀后,使用酶标仪检测luminescence的数值,计算CAR T细胞的杀伤活力。Collect the target cells carrying the HaloTag-HiBiT label (including NCI-H1975-Halotag-HiBiT, EBC-1-HaloTag-HiBiT) in the logarithmic growth phase and the CAR T cells cultured to day 7, and prepare cells with different cell concentrations. Suspension; target cells were seeded in a 96-well plate (3 multiple wells) at 5000 cells/well, then CAR T cell suspension was added according to different effect-to-target ratios, and cultured in a 37°C, 5% CO 2 incubator for 24 hours. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes. Then remove the well plate from the incubator. After returning to room temperature, add an equal volume of pre-prepared detection Buffer (containing 1:100 dilution of LgBiT Protein). and diluted 1:50 HiBiT Extracellular Substrate), mix on a shaker, use a microplate reader to detect the luminescence value, and calculate the killing activity of CAR T cells.
细胞毒性测定via CCK8Cytotoxicity assay via CCK8
使用CCK8试剂盒(同仁化学研究所)对CAR-T细胞进行毒性测定。收集处于对数生长期的靶细胞及培养至第7天的CAR T细胞制备不同细胞密度的细胞悬液,取50ul靶细胞按照5000cells/well进行铺板(3复孔),然后以3:1、1:1、1:3和1:10的效靶比添加50ul CAR T细胞悬液,并在37℃、饱和湿度的5%CO2培养箱中孵育24hr后,每孔添加10ul CCK8,反应4hr后,用酶标仪(Thermo Varioskan LUX)测定在450nm波长下的吸光度。Toxicity assay of CAR-T cells was performed using CCK8 kit (Doren Chemical Research Institute). Collect the target cells in the logarithmic growth phase and the CAR T cells cultured to day 7 to prepare cell suspensions with different cell densities. Take 50ul target cells and plate them at 5000 cells/well (3 replicate wells), and then use 3:1, Add 50ul of CAR T cell suspension at the effect-to-target ratio of 1:1, 1:3 and 1:10, and incubate for 24hr in a 5% CO2 incubator at 37°C and saturated humidity, then add 10ul of CCK8 to each well and react for 4hr. Finally, the absorbance at a wavelength of 450 nm was measured using a microplate reader (Thermo Varioskan LUX).
小鼠CDX模型检测CAR T细胞的抗肿瘤活性Mouse CDX model to detect anti-tumor activity of CAR T cells
收集处于对数生长期的NCI-H820肿瘤细胞,接种于免疫缺陷小鼠的背部皮下,当瘤块生长至大约120mm3时,尾静脉输注CAR T细胞,随后观察小鼠的状态,每周两次测量瘤块的大小,绘制小鼠肿瘤生长曲线,以评估CAR T细胞的体内抗肿瘤活性。NCI-H820 tumor cells in the logarithmic growth phase were collected and inoculated subcutaneously on the backs of immune-deficient mice. When the tumors grew to approximately 120 mm3 , CAR T cells were infused into the tail vein, and the mice's status was subsequently observed every week. The size of the tumor mass was measured twice and the mouse tumor growth curve was drawn to evaluate the in vivo anti-tumor activity of CAR T cells.
实施例1Example 1
病毒制备:将逆转录病毒目的基因质粒pMSGV-CAR与包膜质粒pMD2.G加 入X-tremeGENE 9 DNA Transfection Reagent(Roche)中混匀,加入培养Phoenix GP细胞的培养皿中,轻轻混匀,72h后收集上清,1000g低速离心、0.45um滤膜过滤后,将上清加入到PG13细胞中同时加入Polybrene转染增强剂制备逆转录病毒生产细胞系PG13-CAR。接种2E7/flask密度的PG13-CAR于T75细胞培养瓶中,培养24h后收集上清,400g低速离心、0.45um滤膜过滤后,直接使用或者分装后冻存于-80℃。Virus preparation: Add retrovirus target gene plasmid pMSGV-CAR and envelope plasmid pMD2.G Mix it into X-tremeGENE 9 DNA Transfection Reagent (Roche), add it to the culture dish cultivating Phoenix GP cells, mix gently, collect the supernatant after 72 hours, centrifuge at 1000g at low speed, filter with 0.45um filter membrane, and filter the supernatant Add it to PG13 cells and add Polybrene transfection enhancer to prepare the retrovirus production cell line PG13-CAR. Inoculate PG13-CAR at 2E7/flask density into T75 cell culture flasks. Collect the supernatant after 24 hours of culture. Centrifuge at 400g at low speed and filter with a 0.45um filter. Use it directly or aliquot and freeze at -80°C.
CAR T细胞制备:通过Ficoll-Hypaque密度梯度离心法从健康人外周血中获取单个核细胞,并将其接种至淋巴细胞培养基中(Gibco),添加TransAct(Miltenyi Biotec)和IL-2(GE Healthcare)进行刺激活化和扩增培养。同时,使用Retronectin包被6孔板,将逆转录病毒上清加入到包被的6孔板中2000g离心2h;离心完成后,弃去上清,将活化后的T细胞加入到6孔板中1000g离心20min进行转导,置于37℃、5%CO2培养箱中培养,24h后将细胞转移至T75培养瓶中培养扩增。结果如图1所示,结果表明,图1A示出了靶向EGFR和cMet“和”逻辑BiCAR的组成,包括2个独立的CAR分子,CAR1分子为抗cMet的scFv串联CD3ζ激活信号结构域(如SEQ ID NO.:2所示,其中SEQ ID NO.:3-4为抗cMet的全功能2G单CAR),CAR2分子为抗EGFR的scFv串联CD28或者4-1BB共刺激信号结构域(如SEQ ID NO.:7-10中任一所示,其中SEQ ID NO.:11-14为抗EGFR的全功能2G单CAR);图1B示出“和”逻辑BiCAR表达的分子构建示意图,CAR1分子和CAR2分子通过P2A连接(如SEQ ID NO.:23-26中任一所示);图1C示出了靶向cMet的第二代全功能CAR的组成,scFv串联CD28或4-1BB共刺激信号结构域以及CD3ζ激活信号结构域(如SEQ ID NO.:3-4中任一所示)。CAR T cell preparation: Obtain mononuclear cells from healthy human peripheral blood by Ficoll-Hypaque density gradient centrifugation and inoculate them into lymphocyte culture medium (Gibco), add TransAct (Miltenyi Biotec) and IL-2 (GE Healthcare) for stimulation activation and expansion culture. At the same time, use Retronectin to coat a 6-well plate, add the retrovirus supernatant to the coated 6-well plate, and centrifuge at 2000g for 2 hours; after centrifugation is completed, discard the supernatant, and add the activated T cells to the 6-well plate. Centrifuge at 1000g for 20 minutes for transduction, place in a 37°C, 5% CO2 incubator, and culture. After 24 hours, transfer the cells to a T75 culture flask for culture and expansion. The results are shown in Figure 1. The results show that Figure 1A shows the composition of the "AND" logic BiCAR targeting EGFR and cMet, including 2 independent CAR molecules. The CAR1 molecule is an anti-cMet scFv tandem CD3ζ activation signal domain ( As shown in SEQ ID NO.: 2, SEQ ID NO.: 3-4 is a fully functional 2G single CAR against cMet), and the CAR2 molecule is an anti-EGFR scFv tandem CD28 or 4-1BB costimulatory signaling domain (such as Any one of SEQ ID NO.: 7-10 is shown, wherein SEQ ID NO.: 11-14 is a fully functional 2G single CAR against EGFR); Figure 1B shows a schematic diagram of the molecular construction of "AND" logic BiCAR expression, CAR1 The molecule and the CAR2 molecule are linked through P2A (as shown in any of SEQ ID NO.: 23-26); Figure 1C shows the composition of a second-generation fully functional CAR targeting cMet, scFv tandem CD28 or 4-1BB Stimulation signal domain and CD3ζ activation signal domain (as shown in any one of SEQ ID NO.: 3-4).
实施例2Example 2
流式细胞术检测CAR分子的表达:CAR T细胞培养至第7天时,取1E6的细胞,使用洗涤缓冲液洗涤2次后,添加1ug cMet-His(Acrobiosystem)和1ug EGFR-Fc抗原(Acrobiosystem),4℃孵育30min后,使用洗涤缓冲液洗涤2次,随后添加Anti-His-PE(Miltenyi Biotec)和Allophycocyanin(APC)AffiniPure Goat Anti-Human IgG,Fcγfragment specific(Jackson ImmunoResearch)4℃避光孵育30min。细胞洗涤后使用流式细胞仪(CytoFLEX LX,Beckman Coulter)检测CAR分子的表达。 Detect the expression of CAR molecules by flow cytometry: When the CAR T cells are cultured on the 7th day, take the 1E6 cells, wash them twice with washing buffer, and add 1ug cMet-His (Acrobiosystem) and 1ug EGFR-Fc antigen (Acrobiosystem) , after incubating at 4°C for 30 minutes, wash twice with wash buffer, then add Anti-His-PE (Miltenyi Biotec) and Allophycocyanin (APC) AffiniPure Goat Anti-Human IgG, Fcγfragment specific (Jackson ImmunoResearch) and incubate at 4°C in the dark for 30 minutes . After washing the cells, flow cytometry (CytoFLEX LX, Beckman Coulter) was used to detect the expression of CAR molecules.
结果如图2所示,通过流式细胞术检测CAR T细胞中CAR分子的表达。如图2A所示,Mock T细胞并未检测出第一CAR(anti-cMet-scFv-CD3ζ/O-z)或第二CAR分子(anti-EGFR-scFv-CD28/C-28)的表达;如图2B所示,靶向cMet靶点的全功能二代CAR T(anti-cMet-scFv-CD28-CD3ζ/O-28z)具有67%的转导率;如图2C所示,“和”逻辑BiCAR T细胞中,第一CAR分子O-z和第二CAR分子C-28共表达的转导率为76%。The results are shown in Figure 2. The expression of CAR molecules in CAR T cells was detected by flow cytometry. As shown in Figure 2A, Mock T cells did not detect the expression of the first CAR (anti-cMet-scFv-CD3ζ/O-z) or the second CAR molecule (anti-EGFR-scFv-CD28/C-28); Figure As shown in Figure 2B, the fully functional second-generation CAR T (anti-cMet-scFv-CD28-CD3ζ/O-28z) targeting the cMet target has a transduction rate of 67%; as shown in Figure 2C, the "AND" logic BiCAR In T cells, the transduction rate of co-expression of the first CAR molecule O-z and the second CAR molecule C-28 was 76%.
实施例3Example 3
HiBiT Extracellular Detection System(Promega)方法检测细胞毒性:收集处于对数生长期的携带HaloTag-HiBiT标签的肺癌细胞株NCI-H1975-HaloTag-HiBiT及培养至第7天的CAR T细胞,制备不同细胞浓度的细胞悬液。按照5000cells/well接种靶细胞于96孔板中(3复孔),然后按照1:10、1:3、1:1和3:1的效靶比添加CAR T细胞悬液,37℃、5%CO2培养箱中培养24h,最大释放的阳性对照组中加入一定终浓度的digitonin孵育30min,随后将96孔板从培养箱中取出,待恢复至室温后,加入等体积的预先配制的检测Buffer(含有1:100稀释的LgBiT Protein和1:50稀释的HiBiT Extracellular Substrate),摇床上混匀后,使用酶标仪检测luminescence的数值,计算CAR T细胞的杀伤活力。 HiBiT Extracellular Detection System (Promega) method to detect cytotoxicity: collect the lung cancer cell line NCI-H1975-HaloTag-HiBiT carrying the HaloTag-HiBiT label in the logarithmic growth phase and CAR T cells cultured to day 7, and prepare different cell concentrations of cell suspension. Inoculate target cells into a 96-well plate at 5000 cells/well (3 multiple wells), then add CAR T cell suspension according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, 37°C, 5 Incubate for 24 hours in a % CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes. Then take out the 96-well plate from the incubator. After returning to room temperature, add an equal volume of pre-prepared test solution. Buffer (contains 1:100 diluted LgBiT Protein and 1:50 diluted HiBiT Extracellular Substrate), mix on a shaker, use a microplate reader to detect the luminescence value, and calculate the killing activity of CAR T cells.
如图3所示,结果表明,来自Donor#1(图3A)和Donor#2(图3B)的BiCAR T细胞在体外可以有效的杀伤NCI-H1975肿瘤细胞,并且这种细胞毒性与细胞剂量呈正相关关系。As shown in Figure 3, the results show that BiCAR T cells from Donor#1 (Figure 3A) and Donor#2 (Figure 3B) can effectively kill NCI-H1975 tumor cells in vitro, and this cytotoxicity is positively correlated with cell dose. relationship.
实施例4Example 4
HiBiT Extracellular Detection System(Promega)方法检测细胞毒性:收集处于对数生长期的携带HaloTag-HiBiT标签的肺癌细胞株EBC-1-Halotag-HiBiT及培养至第7天的CAR T细胞,制备不同细胞浓度的细胞悬液。按照5000cells/well将靶细胞接种于96孔板中(3复孔),然后按照1:10、1:3、1:1和3:1的效靶比添加CAR T细胞悬液,37℃、5%CO2培养箱中培养24h,最大释放的阳性对照组中加入一定终浓度的digitonin孵育30min,随后将96孔板从培养箱中取出,加入等体积的预先配制的检测Buffer(含有1:100稀释的LgBiT Protein和1:50稀释的HiBiT Extracellular  Substrate),摇床上混匀后,使用酶标仪检测luminescence的数值,计算CAR T细胞的杀伤活力。 HiBiT Extracellular Detection System (Promega) method to detect cytotoxicity: collect the lung cancer cell line EBC-1-Halotag-HiBiT carrying the HaloTag-HiBiT label in the logarithmic growth phase and CAR T cells cultured to day 7, and prepare different cell concentrations of cell suspension. The target cells were seeded into a 96-well plate (3 multiple wells) at 5000cells/well, and then the CAR T cell suspension was added according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, at 37°C. Incubate for 24 hours in a 5% CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes. Then take out the 96-well plate from the incubator and add an equal volume of pre-prepared detection Buffer (containing 1: 100 diluted LgBiT Protein and 1:50 diluted HiBiT Extracellular Substrate), mix on a shaker, use a microplate reader to detect the luminescence value, and calculate the killing activity of CAR T cells.
如图4所示,结果表明,来自Donor#1(图4A)和Donor#2(图4B)的BiCAR T细胞在体外可以有效的杀伤EBC-1肿瘤细胞,并且这种细胞毒性与细胞剂量呈正相关关系。As shown in Figure 4, the results show that BiCAR T cells from Donor#1 (Figure 4A) and Donor#2 (Figure 4B) can effectively kill EBC-1 tumor cells in vitro, and this cytotoxicity is positively correlated with cell dose. relationship.
实施例5Example 5
使用CCK8试剂盒(同仁化学研究所)对CAR-T细胞进行毒性测定。收集处于对数生长期的人胃癌细胞株SNU5及培养至第7天的CAR T细胞制备不同细胞密度的细胞悬液,取50ul靶细胞按照5000cells/well进行铺板(3复孔),然后以3:1、1:1、1:3和1:10的效靶比添加50ul CAR T细胞悬液,并在37℃、饱和湿度的5%CO2培养箱中孵育24hr后,每孔添加10ul CCK8,反应4hr后,用酶标仪(Thermo Varioskan LUX)测定在450nm波长下的吸光度。Toxicity assay of CAR-T cells was performed using CCK8 kit (Doren Chemical Research Institute). Collect the human gastric cancer cell line SNU5 in the logarithmic growth phase and the CAR T cells cultured to day 7 to prepare cell suspensions with different cell densities. Take 50ul of target cells and plate them at 5000 cells/well (3 replicate wells), and then use 3 :1, 1:1, 1:3 and 1:10, add 50ul CAR T cell suspension and incubate it in a 5% CO2 incubator at 37°C and saturated humidity for 24hr, then add 10ul CCK8 to each well. , after 4 hours of reaction, use a microplate reader (Thermo Varioskan LUX) to measure the absorbance at a wavelength of 450 nm.
如图5所示,结果表明,BiCAR-T细胞可以显著抑制SNU5肿瘤细胞的生长,且这种细胞毒性与细胞剂量呈正相关关系。As shown in Figure 5, the results show that BiCAR-T cells can significantly inhibit the growth of SNU5 tumor cells, and this cytotoxicity is positively correlated with cell dose.
实施例6Example 6
HiBiT Extracellular Detection System(Promega)方法检测细胞毒性:收集处于对数生长期的携带HaloTag-HiBiT标签的稳转细胞株293-Halotag-HiBiT,293-Halotag-HiBiT-MET,293-Halotag-HiBiT-EGFR,293-Halotag-HiBiT-MET&EGFR,及培养至第7天的CAR T细胞,制备不同细胞浓度的细胞悬液。按照5000cells/well将靶细胞接种于96孔板中(3复孔),然后按照1:10、1:3、1:1和3:1的效靶比添加CAR T细胞悬液,37℃、5%CO2培养箱中培养24h,最大释放的阳性对照组中加入一定终浓度的digitonin孵育30min,随后将96孔板从培养箱中取出,加入等体积的预先配制的检测Buffer(含有1:100稀释的LgBiT Protein和1:50稀释的HiBiT Extracellular Substrate),摇床上混匀后,使用酶标仪检测luminescence的数值,计算CAR T细胞的杀伤活力。 HiBiT Extracellular Detection System (Promega) method to detect cytotoxicity: collect stably transfected cell lines 293-Halotag-HiBiT, 293-Halotag-HiBiT-MET, 293-Halotag-HiBiT-EGFR carrying HaloTag-HiBiT tags in logarithmic growth phase , 293-Halotag-HiBiT-MET&EGFR, and CAR T cells cultured to day 7 to prepare cell suspensions with different cell concentrations. The target cells were seeded into a 96-well plate (3 multiple wells) at 5000cells/well, and then the CAR T cell suspension was added according to the effect-to-target ratio of 1:10, 1:3, 1:1 and 3:1, at 37°C. Incubate for 24 hours in a 5% CO 2 incubator. Add a certain final concentration of digitonin to the maximum-release positive control group and incubate for 30 minutes. Then take out the 96-well plate from the incubator and add an equal volume of pre-prepared detection Buffer (containing 1: 100 diluted LgBiT Protein and 1:50 diluted HiBiT Extracellular Substrate), mix on a shaker, use a microplate reader to detect the luminescence value, and calculate the killing activity of CAR T cells.
图6示出了BiCAR T细胞对共表达EGFR和cMet的293T细胞株(6A)、单表达cMet的293T细胞株(6B),单表达EGFR的293T细胞株(6C)、对照293T细胞株 (6D)共孵育24h后的体外细胞溶解毒性。CAR-T效应细胞与靶细胞的比例分别为1:10,1:3,1:1以及3:1。结果表明BiCAR T细胞对于cMet单一靶点或者cMet/EGFR两个靶点共表达的稳转细胞株呈现了剂量依赖的特异性的体外杀伤功能。Figure 6 shows the response of BiCAR T cells to the 293T cell line co-expressing EGFR and cMet (6A), the 293T cell line expressing cMet alone (6B), the 293T cell line expressing EGFR alone (6C), and the control 293T cell line. (6D) In vitro cytolytic toxicity after 24 h of co-incubation. The ratios of CAR-T effector cells to target cells are 1:10, 1:3, 1:1 and 3:1 respectively. The results showed that BiCAR T cells exhibited a dose-dependent and specific in vitro killing function against stably transduced cell lines expressing cMet as a single target or cMet/EGFR as both targets.
实施例7Example 7
小鼠CDX模型检测CAR T细胞的抗肿瘤活性:收集处于对数生长期的NCI-H820细胞,接种于免疫缺陷鼠的背部皮下。当瘤块的体积达到120mm3左右,将肿瘤负荷大小相当的小鼠随机分组,尾静脉输注Mock T、O-28z CAR及BiCAR T细胞。每周两次测量皮下肿瘤的大小并观察小鼠的状态,绘制肿瘤生长曲线,以评估CAR T细胞的体内抗肿瘤活性。Mouse CDX model was used to detect the anti-tumor activity of CAR T cells: NCI-H820 cells in the logarithmic growth phase were collected and inoculated subcutaneously on the backs of immunodeficient mice. When the tumor volume reaches about 120mm3 , mice with similar tumor burdens are randomly divided into groups, and Mock T, O-28z CAR and BiCAR T cells are infused into the tail vein. The size of subcutaneous tumors was measured twice a week and the status of mice was observed, and tumor growth curves were drawn to evaluate the in vivo anti-tumor activity of CAR T cells.
如图7所示,结果表明,O-28z CAR及BiCAR-T细胞均显著的抑制了肿瘤的生长,并在CAR T细胞输注20天后完全消灭了小鼠体内的肿瘤细胞。As shown in Figure 7, the results showed that both O-28z CAR and BiCAR-T cells significantly inhibited tumor growth and completely eliminated tumor cells in mice 20 days after CAR T cell infusion.
序列表sequence list
SEQ ID NO.1:
SEQ ID NO.1:
SEQ ID NO.2:
SEQ ID NO.2:
SEQ ID NO.3:
SEQ ID NO.3:
SEQ ID NO.4:

SEQ ID NO.4:

SEQ ID NO. 5:
SEQ ID NO. 5:
SEQ ID NO. 6:
SEQ ID NO. 6:
SEQ ID NO. 7:
SEQ ID NO. 7:
SEQ ID NO. 8:
SEQ ID NO. 8:
SEQ ID NO. 9:
SEQ ID NO. 9:
SEQ ID NO. 10:
SEQ ID NO. 10:
SEQ ID NO. 11:
SEQ ID NO. 11:
SEQ ID NO. 12:
SEQ ID NO. 12:
SEQ ID NO. 13:
SEQ ID NO. 13:
SEQ ID NO. 14:
SEQ ID NO. 14:
SEQ ID NO. 15:
SEQ ID NO. 15:
SEQ ID NO. 16:
SEQ ID NO. 16:
SEQ ID NO. 17:
SEQ ID NO. 17:
SEQ ID NO. 18:
SEQ ID NO. 18:
SEQ ID NO. 19:
SEQ ID NO. 19:
SEQ ID NO. 20:
SEQ ID NO. 20:
SEQ ID NO. 21:
SEQ ID NO. 21:
SEQ ID NO. 22:
SEQ ID NO. 22:
SEQ ID NO. 23:
SEQ ID NO. 23:
SEQ ID NO. 24:
SEQ ID NO. 24:
SEQ ID NO. 25:

SEQ ID NO. 25:

SEQ ID NO. 26:
SEQ ID NO. 26:
SEQ ID NO. 27:

SEQ ID NO. 27:

SEQ ID NO. 28:

SEQ ID NO. 28:

SEQ ID NO. 29:
SEQ ID NO. 29:
SEQ ID NO. 30:

SEQ ID NO. 30:

在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in this application are incorporated by reference in this application to the same extent as if each individual document was individually incorporated by reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art can make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of this application.

Claims (12)

  1. 一种工程化的免疫细胞,其特征在于,所述工程化的免疫细胞表达第一CAR和第二CAR,所述第一CAR靶向第一肿瘤细胞标志物,所述第二CAR靶向第二肿瘤细胞标志物,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。An engineered immune cell, characterized in that the engineered immune cell expresses a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the second CAR targets a first tumor cell marker. Two tumor cell markers, the first tumor cell marker is selected from the following group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof; the second tumor cell marker is selected from the following group Group: EGFR, EpCAM, Her2, Her3, or combinations thereof.
  2. 如权利要求1所述的免疫细胞,其特征在于,所述第一CAR的结构如式I所示:
    L1-S1-H1-TM1-C1-CD3ζ    (I)
    The immune cell according to claim 1, wherein the structure of the first CAR is as shown in Formula I:
    L1-S1-H1-TM1-C1-CD3ζ (I)
    式中,所述“-”为连接肽或肽键;In the formula, the "-" is a connecting peptide or peptide bond;
    L1为无或第一信号肽序列;L1 is none or the first signal peptide sequence;
    S1为靶向第一肿瘤细胞标志物的抗原结合结构域,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;S1 is an antigen-binding domain targeting a first tumor cell marker selected from the group consisting of: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof;
    H1为无或第一铰链区;H1 is none or the first hinge area;
    TM1为第一跨膜结构域;TM1 is the first transmembrane domain;
    C1为无或第一共刺激信号分子;C1 is no or the first costimulatory signal molecule;
    CD3ζ为源于CD3ζ的胞浆信号传导序列。CD3ζ is a cytoplasmic signaling sequence derived from CD3ζ.
  3. 如权利要求1所述的免疫细胞,其特征在于,所述第二CAR的结构如式II所示:
    L2-S2-H2-TM2-C2-Z2    (II)
    The immune cell according to claim 1, wherein the structure of the second CAR is shown in Formula II:
    L2-S2-H2-TM2-C2-Z2 (II)
    式中,所述“-”为连接肽或肽键;In the formula, the "-" is a connecting peptide or peptide bond;
    L2为无或第二信号肽序列;L2 is none or the second signal peptide sequence;
    S2为靶向第二肿瘤细胞标志物的抗原结合结构域,所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合;S2 is an antigen-binding domain targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or a combination thereof;
    H2为无或第二铰链区;H2 is no or second hinge region;
    TM2为第二跨膜结构域;TM2 is the second transmembrane domain;
    C2为第二共刺激信号分子;C2 is the second costimulatory signal molecule;
    Z2为无或源于CD3ζ的胞浆信号传导序列。Z2 is a cytoplasmic signaling sequence that is absent or derived from CD3ζ.
  4. 一种制备权利要求1所述的工程化的免疫细胞的方法,其特征在于,包括以下步骤: A method for preparing engineered immune cells according to claim 1, characterized by comprising the following steps:
    (A)提供一待改造的免疫细胞;和(A) providing an immune cell to be modified; and
    (B)对所述的免疫细胞进行改造,从而使得所述的免疫细胞表达第一CAR和第二CAR,所述第一CAR靶向第一肿瘤细胞标志物,所述第二CAR靶向第二肿瘤细胞标志物,从而获得权利要求1所述的工程化的免疫细胞。(B) Modify the immune cells so that the immune cells express a first CAR and a second CAR, the first CAR targets a first tumor cell marker, and the second CAR targets a first tumor cell marker. Two tumor cell markers, thereby obtaining the engineered immune cells described in claim 1.
  5. 一种制剂,其特征在于,所述制剂含有权利要求1所述的工程化的免疫细胞,以及药学上可接受的载体、稀释剂或赋形剂。A preparation, characterized in that the preparation contains the engineered immune cells of claim 1 and a pharmaceutically acceptable carrier, diluent or excipient.
  6. 一种如权利要求1所述的工程化的免疫细胞的用途,其特征在于,用于制备选择性杀伤肿瘤的药物或制剂。The use of engineered immune cells according to claim 1, characterized in that it is used to prepare drugs or preparations that selectively kill tumors.
  7. 如权利要求6所述的用途,其特征在于,所述肿瘤包括同时表达EGFR和cMet的肿瘤。The use according to claim 6, wherein the tumor includes a tumor expressing both EGFR and cMet.
  8. 一种用于选择性杀伤肿瘤的试剂盒,其特征在于,所述试剂盒含有容器,以及位于容器内的:A kit for selectively killing tumors, characterized in that the kit contains a container, and located in the container:
    (1)第一核酸序列,所述第一核酸序列含有用于表达靶向第一肿瘤细胞标志物的第一CAR的第一表达盒,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;和(1) A first nucleic acid sequence containing a first expression cassette for expressing a first CAR targeting a first tumor cell marker selected from the group consisting of: cMet , Her2, Her3, Muc1, ROR1, PD-L1, CD47, or combinations thereof; and
    (2)第二核酸序列,所述第二核酸序列含有靶向第二肿瘤细胞标志物的第二CAR的第二表达盒,所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。(2) A second nucleic acid sequence containing a second expression cassette of a second CAR targeting a second tumor cell marker selected from the group consisting of: EGFR, EpCAM, Her2, Her3, or combinations thereof.
  9. 一种融合蛋白,其特征在于,所述融合蛋白包含靶向第一肿瘤细胞标志物的第一CAR和靶向第二肿瘤细胞标志物的第二CAR,所述第一肿瘤细胞标志物选自下组:cMet、Her2、Her3、Muc1、ROR1、PD-L1、CD47、或其组合;所述第二肿瘤细胞标志物选自下组:EGFR、EpCAM、Her2、Her3、或其组合。A fusion protein, characterized in that the fusion protein includes a first CAR targeting a first tumor cell marker and a second CAR targeting a second tumor cell marker, the first tumor cell marker being selected from the group consisting of The lower group: cMet, Her2, Her3, Mucl, ROR1, PD-L1, CD47, or a combination thereof; the second tumor cell marker is selected from the lower group: EGFR, EpCAM, Her2, Her3, or a combination thereof.
  10. 如权利要求9所述的融合蛋白,其特征在于,所述融合蛋白的氨基酸序列如SEQ ID NO.:23-26中任一所示。The fusion protein of claim 9, wherein the amino acid sequence of the fusion protein is as shown in any one of SEQ ID NO.: 23-26.
  11. 一种多核苷酸,所述多核苷酸编码权利要求9所述的融合蛋白。A polynucleotide encoding the fusion protein of claim 9.
  12. 一种载体,其特征在于,所述载体包括权利要求11所述的多核苷酸。 A vector, characterized in that the vector includes the polynucleotide of claim 11.
PCT/CN2023/116708 2022-09-09 2023-09-04 Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof WO2024051641A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202211104889.9 2022-09-09
CN202211104889.9A CN117305248A (en) 2022-09-09 2022-09-09 Bispecific chimeric antigen receptor for resisting EGFR and cMet and application thereof

Publications (1)

Publication Number Publication Date
WO2024051641A1 true WO2024051641A1 (en) 2024-03-14

Family

ID=89285449

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/116708 WO2024051641A1 (en) 2022-09-09 2023-09-04 Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof

Country Status (2)

Country Link
CN (1) CN117305248A (en)
WO (1) WO2024051641A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109161532A (en) * 2018-05-31 2019-01-08 华东师范大学 The engineering immunocyte of PSMA and PD-L1 is targeted simultaneously
CN109423495A (en) * 2017-08-24 2019-03-05 上海恒润达生生物科技有限公司 A kind of pair of targeting Chimeric antigen receptor and application thereof
WO2020172177A1 (en) * 2019-02-18 2020-08-27 Memorial Sloan-Kettering Cancer Center Combinations of multiple chimeric antigen receptors for immunotherapy

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115109754A (en) * 2021-03-23 2022-09-27 复星凯特生物科技有限公司 anti-EGFR and cMet bispecific chimeric antigen receptor and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109423495A (en) * 2017-08-24 2019-03-05 上海恒润达生生物科技有限公司 A kind of pair of targeting Chimeric antigen receptor and application thereof
CN109161532A (en) * 2018-05-31 2019-01-08 华东师范大学 The engineering immunocyte of PSMA and PD-L1 is targeted simultaneously
WO2020172177A1 (en) * 2019-02-18 2020-08-27 Memorial Sloan-Kettering Cancer Center Combinations of multiple chimeric antigen receptors for immunotherapy

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
MENG, TAO ET AL.: "Parallel CAR-T Cells Targeting HER2 and CLDN18.2 Reduce the Off Target Effect on Gastric Cancer", ACTA UNIVERSITATIS MEDICINALIS ANHUI, 30 August 2022 (2022-08-30) *
ZENG, WANYING ET AL.: "Engineering Novel CD19/CD22 Dual-Target CAR-T Cells for Improved Anti-Tumor Activity", CANCER INVESTIGATION, 25 November 2021 (2021-11-25) *

Also Published As

Publication number Publication date
CN117305248A (en) 2023-12-29

Similar Documents

Publication Publication Date Title
CN110950953B (en) Monoclonal antibody against B7-H3 and application thereof in cell therapy
CN109306016B (en) NKG2D-CAR-T cells co-expressing cytokine IL-7 and uses thereof
TWI771678B (en) Engineered immune cells targeting BCMA and their uses
CN109575143B (en) Bispecific CD20-CD19-CAR and application thereof
CN112778427B (en) Bispecific CS1-BCMA CAR-T cells and uses thereof
CN113087806B (en) Novel CAR-T cells targeting multiple tumors, and preparation and methods thereof
CN111378625A (en) Preparation and application of CXCL13 chemotactic CAR-T cell
CN110054698B (en) Construction and application of novel CD19-CAR vector of anti-CD 19 antibody
WO2021223720A1 (en) Humanized cd19 antibody and use thereof
WO2023217192A1 (en) Preparation of chimeric antigen receptor immune cell constructed based on msln precursor protein and use thereof
WO2023016524A1 (en) Combined her2 and meso dual-target car-t vector, construction method therefor and application thereof in cancer
CN109897114B (en) CD 47-targeted engineered immune cells with suicide gene switch
WO2021208750A1 (en) Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
US20210395362A1 (en) Car-t cells with humanized cd19 scfv with mutation in cdr 1 region
CN115109754A (en) anti-EGFR and cMet bispecific chimeric antigen receptor and application thereof
WO2024051641A1 (en) Anti-egfr and cmet bispecific chimeric antigen receptor and use thereof
CN115873115A (en) Application of anti-4-1 BB monoclonal antibody in tumor immunotherapy
WO2023093888A1 (en) Preparation and use of immune cells of chimeric antigen receptor constructed on the basis of efna1
WO2022151959A1 (en) Car-t cell targeting b7-h3 and application thereof in treatment of acute myeloid leukemia
CN109593137B (en) Construction and application of novel CD20-CAR vector of anti-CD 20 antibody
WO2024041618A1 (en) Engineered immune cell co-expressing cd40l, preparation therefor, and use thereof
CN115491360A (en) Preparation and application of targeted NKG2D ligand and FOLR1 double targeting target point CAR T
CN115572715A (en) Preparation and application of FOLR1 and HER2 targeting double targeting CAR T
CN110577604A (en) Chimeric antigen receptor T cell carrying GITR costimulatory signal targeting EGFR