CN109971724B - CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof - Google Patents

CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof Download PDF

Info

Publication number
CN109971724B
CN109971724B CN201711461266.6A CN201711461266A CN109971724B CN 109971724 B CN109971724 B CN 109971724B CN 201711461266 A CN201711461266 A CN 201711461266A CN 109971724 B CN109971724 B CN 109971724B
Authority
CN
China
Prior art keywords
seq
amino acid
cell
sequence
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201711461266.6A
Other languages
Chinese (zh)
Other versions
CN109971724A (en
Inventor
钱其军
金华君
游术梅
江芏青
刘祥箴
李林芳
王超
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Cell Therapy Research Institute
Shanghai Cell Therapy Group Co Ltd
Original Assignee
Shanghai Cell Therapy Research Institute
Shanghai Cell Therapy Group Co Ltd
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 Shanghai Cell Therapy Research Institute, Shanghai Cell Therapy Group Co Ltd filed Critical Shanghai Cell Therapy Research Institute
Priority to CN201711461266.6A priority Critical patent/CN109971724B/en
Priority to PCT/CN2018/124316 priority patent/WO2019129138A1/en
Publication of CN109971724A publication Critical patent/CN109971724A/en
Application granted granted Critical
Publication of CN109971724B publication Critical patent/CN109971724B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • 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
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Abstract

The present invention provides chimeric antigen receptor T cells that self-express PD1 immune checkpoint inhibitory antibodies and target the ErbB receptor family and uses thereof. T cells of the invention: comprising a coding sequence for expressing a chimeric antigen receptor that targets the ErbB receptor family and a coding sequence for a PD1 antibody; and/or express chimeric antigen receptor and PD1 antibodies that target the ErbB receptor family. Compared with the single chimeric antigen receptor T cells of the targeted ErbB receptor family, the T cells have better proliferation activation capacity, can overcome the inhibition of immune microenvironment, promote the apoptosis of tumor cells, play an anti-tumor immune response and kill the tumor cells.

Description

CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof
Technical Field
The invention belongs to genetic engineering and immunology, and relates to a CAR-T cell which targets an ErbB receptor family and self-expresses a PD-1 antibody and a PD1 antibody thereof and an application of the PD1 antibody.
Background
Cancer is now the first killer of human health, and rapid life rhythm, huge working pressure, unhealthy eating habits and poor environment are all the help of cancer occurrence, so that the high incidence and the younger trend of cancer are more and more obvious. The current commonly used treatment methods have very limited effects, and a more effective treatment method is still needed to be explored to improve the survival rate and the survival quality of cancer patients.
Chimeric antigen receptor T cell (CAR-T) therapy has achieved very good efficacy in hematological malignancies as one of the important branches of tumor immunotherapy, with a complete remission rate of more than 90% for relapsed refractory B-cell leukemia. A chimeric antigen receptor is an artificial synthetic receptor that generally comprises an extracellular antigen binding domain, a transmembrane hinge region, and an intracellular signaling region. Gene recombination is performed in vitro by combining a single-chain variable region (scFv) of an antibody recognizing a tumor-associated antigen (tumor associated antigen, TAA) with an intracellular signaling domain "immunoreceptor tyrosine activation motif (immunoreceptor tyrosine-based activation motifs, ITAM)". The resulting genetically engineered T cells are then introduced into T cells by a viral or other vector system, and are referred to as CAR-T cells. After large-scale expansion in vitro, CAR-T cells are infused back into patients and can exhibit potent anticancer effects in a non-MHC restricted mode.
However, many obstacles have been encountered in replicating this therapy to solid tumors. Firstly, solid tumors lack tumor specific antigens, and perfect therapeutic targets cannot be found. Secondly, the solid tumor has stronger tumor immunosuppression microenvironment. In addition, tumors themselves have a strong heterogeneity. These factors lead to a recurrence of tumor after immunotherapy. Therefore, the preparation of CART cells with multiple targets and stronger functions is a key for improving the tumor treatment effect. For example, treatment of B cell acute lymphoblastic leukemia with CART19 resulted in a negative recurrence of CD19 antigen in about 30% of patients treated, and studies have shown that CART cells with both CART19/123 double targets, constructed with CD19 and IL3 receptor alpha chain CD123 as targets, have better in vivo therapeutic efficacy and lower recurrence rates (J Clin invest.2016 Oct 3;126 (10)). However, most of antigen recognition regions of these CARs are artificially constructed single-chain antibodies, have strong immunogenicity, and in vivo treatment is easily recognized and cleared, resulting in poor therapeutic effects. Therefore, the antigen recognition region with multiple targets is constructed by adopting natural peptide segments, and the method has the advantages of weak immunogenicity and wider targeting range.
The ErbB receptor family comprises four members, the epidermal growth factor receptors ErbB1 (EGFR/Her 2), erbB2 (Her 2), erbB3 (Her 3) and ErbB4 (Her 4), respectively. In normal adults, the expression levels of these four receptors are all at low levels. However, many malignant tumors occur in association with over-expression of ErbB1 and/or ErbB2, including head and neck cancer, breast cancer, lung cancer, gastrointestinal cancer, prostate cancer, pancreatic cancer, and the like. Thus, many of the antitumor drugs in clinical studies are monoclonal antibodies targeting the extracellular regions of different ErbB receptors and small molecule tyrosine kinase inhibitors, such as HER2 humanized antibody Herceptin (Herceptin), which has been FDA approved for clinical treatment of breast cancer.
However, erbB receptors typically function as tightly bound dimers. Her2 is the best dimerization partner, especially when dimerized with EGFR or Her3, that enhances tyrosine kinase signaling activation. Her3 has ligand binding capacity but lacks tyrosine kinase activity, and the heterodimer formed with Her2 is the strongest signal complex, with Her2/Her3 being the most representative heterodimer in breast cancer. Antitumor drugs that target a receptor alone are prone to cause tumor recurrence because they cannot target other ErbB receptor dimers.
T1E is a chimeric polypeptide consisting of seven amino acids at the N-terminus of human transcription growth factor alpha (TGF alpha) and 48 amino acids at the C-terminus of Epidermal Growth Factor (EGF), which has high affinity for both ErbB 1-based homodimers and heterodimers, and T1E is also capable of efficiently binding ErbB2/3 heterodimers. Studies show that the CAR with T1E as scFv transfects T cells, and can effectively treat the mouse tumor xenograft model of human head and neck cancer. However, T1E is unable to bind ErbB2 or ErbB3 alone, and has drawbacks in targeting the ErbB receptor family.
Herstatin is a truncated version of Her2, resulting from selective cleavage of Her2, whose sequence includes 340 amino acids from the extracellular domains I and II of Her2 and 79 amino acids from the eighth intron, and is a soluble Her2 self-inhibitor. Herstatin binds with high affinity to EGFR alone or Her2, with 79 amino acids encoded by the eighth intron (designated Herin) serving as the primary role.
PD1 (Programmed Death 1): reprogrammed cell death receptor 1, a member of the CD28 family of regulatory T cells, belongs to the immunoglobulin superfamily of receptors. PD-1 and its ligand PD-L1/PD-L2 play an important role in co-suppression and failure of T cells, and their interaction inhibits proliferation of T cells and cytokine secretion regulated by co-stimulatory molecules, down regulates expression of anti-apoptotic molecule BCL-xl, weakens the function of tumor-specific T cells, and results in some tumor patients unable to completely eliminate tumors. Thus, inhibition of the immune checkpoint PD-1/PD-L1 pathway is a new direction and target for current treatment of lymphomas. The PD-1 antibody blocking treatment has a certain treatment effect on advanced or refractory melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, colorectal cancer, hodgkin lymphoma, ovarian cancer and the like, but the higher production cost and response rate of the antibody blocking treatment do not limit the clinical application of the antibody blocking treatment.
Disclosure of Invention
Provided herein are T cells that self-express an immune checkpoint inhibitory PD1 antibody and target a chimeric antigen receptor of the ErbB receptor family; preferably, the T cell:
(1) Comprising a coding sequence for a chimeric antigen receptor that targets an ErbB receptor and a coding sequence for a PD1 antibody; and/or
(2) Chimeric antigen receptor and PD1 antibodies are expressed that target ErbB receptors.
In one or more embodiments, the T cell has integrated into its genome an expression cassette for a chimeric antigen receptor that targets an ErbB receptor and an expression cassette for a PD1 antibody.
In one or more embodiments, the PD1 antibody comprises an anti-PD 1 single chain antibody and an IgG4Fc.
In one or more embodiments, the amino acid sequence of the IgG4Fc is shown as amino acid residues 267-495 of SEQ ID NO. 1; preferably, the coding sequence is shown as the base sequence of 799-1485 of SEQ ID NO. 2.
In one or more embodiments, the amino acid sequence of the variable region (VL region) of the light chain of the anti-PD 1 single-chain antibody (scFv) is shown as amino acid residues 21-131 of SEQ ID NO. 1; preferably, the coding sequence is shown as the 64 th to 393 th base sequence of SEQ ID NO. 2; the amino acid sequence of a heavy chain variable region (VH region) in the anti-PD 1 single-chain antibody is shown as 147 th-266 th amino acid sequence of SEQ ID NO. 1; preferably, the coding sequence is shown in the base sequence of 439-798 of SEQ ID NO. 2; preferably, the amino acid sequence of the anti-PD 1 single-chain antibody is shown as amino acid residues 21-266 of SEQ ID NO. 1; preferably, the coding sequence is shown as 61-798 base sequences of SEQ ID NO. 2.
In one or more embodiments, the PD1 antibody comprises, in order from N-terminus to C-terminus, a light chain signal peptide, an anti-PD 1 single chain antibody, and an IgG4Fc; preferably, the amino acid sequence of the light chain signal peptide is shown as amino acid residues 1 to 20 of SEQ ID NO. 1; preferably, the coding sequence of the light chain signal peptide is shown as the 1 st to 60 th base sequence of SEQ ID NO. 2.
In one or more embodiments, the PD1 antibody has an amino acid sequence as set forth in SEQ ID NO. 1 at amino acid positions 21-495 or as set forth in SEQ ID NO. 1; preferably, the coding sequence of the PD1 antibody is shown as amino acid residues 61-1485 of SEQ ID NO. 2, preferably as shown as SEQ ID NO. 2.
In one or more embodiments, the chimeric antigen receptor comprises an optional signal peptide, a T1E, EAAAK linker, a Herin, a hinge region, a transmembrane region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain; preferably, the hinge region is a hinge region that is more than 50 amino acid residues in length.
In one or more embodiments, the signal peptide is a secretory signal peptide and a membrane-bound signal peptide, preferably a CD8 signal peptide; more preferably, the amino acid sequence of the CD8 signal peptide is shown as amino acid residues 1-22 of SEQ ID NO. 5.
In one or more embodiments, the amino acid sequence of T1E is shown as amino acid residues 23-77 of SEQ ID NO. 5; preferably, the coding sequence is shown as the 67 th to 231 th base sequence of SEQ ID NO. 6.
In one or more embodiments, the EAAAK linker has an amino acid sequence as set forth in amino acid residues 78-92 of SEQ ID NO. 5; preferably, the coding sequence is shown as 232-276 base sequences of SEQ ID NO. 6.
In one or more embodiments, the amino acid sequence of the Herin is as set forth in amino acid residues 93-171 of SEQ ID NO. 5; preferably, the coding sequence is shown in the base sequence of 277-513 of SEQ ID NO. 6.
In one or more embodiments, the hinge region is a CD8 hinge region or an IgG4CH2CH3 hinge region; preferably, the hinge region is an IgG4CH2CH3 hinge region, and the amino acid sequence of the hinge region is shown as the 172 th to 399 th amino acid residues of SEQ ID NO. 5; preferably, the coding sequence is shown as the 514 th to 1197 th base sequence of SEQ ID NO. 6.
In one or more embodiments, the transmembrane region is one of a CD28 transmembrane region, a CD8 transmembrane region, a cd3ζ transmembrane region, a CD134 transmembrane region, a CD137 transmembrane region, an ICOS transmembrane region, and a DAP10 transmembrane region; preferably a CD28 transmembrane region, preferably having an amino acid sequence as shown in amino acid residues 400-427 of SEQ ID NO. 5, preferably having a coding sequence as shown in base sequences 1198-1281 of SEQ ID NO. 6.
In one or more embodiments, the intracellular co-stimulatory signaling domain includes an intracellular domain of a co-stimulatory signaling molecule, including an intracellular domain of CD28, CD134/OX40, CD137/4-1BB, lymphocyte-specific protein tyrosine kinase, inducible T cell co-stimulatory factor (ICOS), and DNAX activator protein 10; preferably, the intracellular domain of the costimulatory signal molecule is a CD28 intracellular domain, the amino acid sequence of which is shown as amino acid residues 428-468 of SEQ ID NO. 5, and the coding sequence of which is shown as base sequences 1282-1404 of SEQ ID NO. 6.
In one or more embodiments, the intracellular signaling domain is a cd3ζ intracellular signaling domain or an fcsriy intracellular signaling domain; preferably a CD3 zeta intracellular signal domain, preferably the amino acid sequence of said CD3 zeta intracellular signal domain is shown as SEQ ID NO:5 amino acid residues 469-580, preferably the coding sequence is shown as SEQ ID NO:6 base sequences 1405-1740.
In one or more embodiments, the chimeric antigen receptor has an amino acid sequence as shown in amino acid residues 23-580 of SEQ ID NO. 5, or as shown in SEQ ID NO. 5; preferably, the nucleotide sequence of the chimeric antigen receptor is shown as the 67 th to 1740 th base sequence shown as SEQ ID NO. 6 or as SEQ ID NO. 6.
The present invention also provides a composition comprising:
(1) A vector comprising an expression cassette for a chimeric antigen receptor described herein for integration of the expression cassette into the genome of a host cell; and
(2) A vector comprising an expression cassette for a CD40 activating antibody for integration of said expression cassette into the genome of a host cell.
In one or more embodiments, the chimeric antigen receptor and its coding sequence and the CD40 activating antibody and its coding sequence are as described in any of the embodiments herein.
The invention also provides a kit comprising:
(1) A vector comprising an expression cassette for a chimeric antigen receptor described herein for integration of the expression cassette into the genome of a host cell; and
(2) A vector comprising an expression cassette for a PD1 antibody, said vector being used to integrate said expression cassette into the genome of a host cell.
In one or more embodiments, the chimeric antigen receptor and its coding sequence and the PD1 antibody and its coding sequence are as described in any of the embodiments herein.
The invention also provides a pharmaceutical composition comprising a T cell as described herein or a PD1 antibody expressed by the T cell.
The invention also provides the use of the T cells or the pharmaceutical composition thereof or the PD1 antibodies expressed by the T cells in the preparation of medicaments for treating or preventing malignant tumors.
In one or more embodiments, the cancer is selected from: liver cancer, adenocarcinoma, lung cancer, colon cancer, rectal cancer, carcinoma of large intestine, breast cancer, ovarian cancer, cervical cancer, gastric cancer, head and neck cancer, cholangiocarcinoma, gall bladder cancer, melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, hodgkin's lymphoma, esophageal cancer, pancreatic cancer, or prostate cancer.
Drawings
Fig. 1: gene structural pattern of recombinant plasmids pS328-antiPD1-wt, pS328-antiPD1, pNB328-EHCAR-EK-28 TIZ.
Fig. 2A-2B: positive rate and antibody expression level of chimeric antigen receptor modified T cells constructed by pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 plasmids with different mass ratios.
Fig. 3A-3B: EHCAR-EK-28TIZ T cells and EHCAR-EK-28TIZ-antiPD1T cells were used to measure the positive rate and the antibody expression level.
Fig. 4: proliferation rate comparison of EHCAR-EK-28TIZ and EHCAR-EK-28 TIZ-anti-iPD 1T cells.
Fig. 5A-5D: cell phenotyping analysis of T cells of EHCAR-EK-28TIZ and EHCAR-EK-28TIZ-antiPD1T cells, 5A representing senescence phenotypes PD1,5B and 5C representing activation phenotypes CD69 and CD107 alpha, respectively, 5D representing a memory phenotype.
Fig. 6: killing contrast of EHCAR-EK-28TIZ and EHCAR-EK-28 TIZ-anti-iPD 1T cells, including human hepatoma cell HCCLM3, human hepatoma cell Hep3B and human non-small cell lung cancer H23.
Fig. 7: alterations in IL-2, IL-4, IL-6, IL-10, TNF- α and IFN- γ cytokine secretion by EHCAR-EK-28TIZ and EHCAR-EK-28 TIZ-anti-iPD 1T cells under EGFR antigen stimulation.
Fig. 8: EHCAR-EK-28TIZ T cells, EHCAR-EK-28TIZ-antiPD1-wt T cells, EHCAR-EK-28TIZ-antiPD1T cells, mock-T cells and PBS blank, tumor cell fluorescence values varied on different days after treatment of mice, respectively.
Detailed Description
The following is a description of some of the terms involved in the present invention.
In the present invention, the term "expression cassette" refers to the complete elements required for expression of a gene, including promoters and gene coding sequences.
The term "coding sequence" is defined herein as that portion of a nucleic acid sequence that directly determines the amino acid sequence of its protein product (e.g., CAR, single chain antibody, hinge region, and transmembrane region). The boundaries of the coding sequence are typically determined by a ribosome binding site (for prokaryotic cells) immediately upstream of the open reading frame at the 5 'end of the mRNA and a transcription termination sequence immediately downstream of the open reading frame at the 3' end of the mRNA. Coding sequences may include, but are not limited to, DNA, cDNA, and recombinant nucleic acid sequences.
The term "Fc", i.e., the crystallizable section of an antibody (fragment crystallizable, fc), refers to the peptide section comprising the CH2 and CH3 domains of the heavy chain of an antibody at the end of the stem of the "Y" structure of an antibody molecule, which is the site of interaction of the antibody with an effector molecule or cell.
The term "costimulatory molecule" refers to a molecule that is present on the surface of an antigen presenting cell and that is capable of binding to a costimulatory molecule receptor on a Th cell to produce a costimulatory signal. Proliferation of lymphocytes requires not only antigen binding but also signal of the co-stimulatory molecule. The co-stimulatory signal is transmitted to the T cell primarily through the co-stimulatory molecule CD80, CD86 expressed on the surface of the antigen presenting cell binding to the CD28 molecule on the surface of the T cell. B cells receive costimulatory signals through common pathogen components such as LPS, or through complement components, or through activated antigen-specific CD40L on Th cell surfaces.
The term "linker" or hinge is a polypeptide fragment that connects between different proteins or polypeptides in order to maintain the connected proteins or polypeptides in their respective spatial conformations in order to maintain the function or activity of the protein or polypeptide. Exemplary linkers include linkers comprising G and/or S, and for example Furin 2A peptides.
The term "specific binding" refers to a reaction between an antibody or antigen binding fragment and an antigen against which it is directed. In certain embodiments, an antibody that specifically binds to (or has specificity for) an antigen means that the antibody binds to or has specificity for an antigen in an amount of less than about 10 -5 M, e.g. less than about 10 -6 M、10 -7 M、10 -8 M、10 -9 M or 10 -10 M or less affinity (KD) binds the antigen. "specific recognition" has similar meaning.
The term "pharmaceutically acceptable excipients" refers to carriers and/or excipients that are pharmacologically and/or physiologically compatible with the subject and active ingredient, which are well known in the art (see, e.g., remington's Pharmaceutical sciences. Mediated by Gennaro AR,19th ed.Pennsylvania:Mack Publishing Company,1995), and include, but are not limited to: pH adjusters, surfactants, adjuvants, ionic strength enhancers. For example, pH modifiers include, but are not limited to, phosphate buffers; surfactants include, but are not limited to, cationic, anionic or nonionic surfactants, such as Tween-80; ionic strength enhancers include, but are not limited to, sodium chloride.
The term "effective amount" refers to the amount that achieves treatment, prevention, alleviation and/or relief of a disease or condition of the present invention in a subject.
The term "disease and/or disorder" refers to a physical state of the subject that is associated with the disease and/or disorder of the present invention.
The term "subject" or "patient" may refer to a patient or other animal, particularly a mammal, such as a human, dog, monkey, cow, horse, etc., receiving a pharmaceutical composition of the invention for treating, preventing, alleviating and/or alleviating a disease or condition described herein.
The term "chimeric antigen receptor" (CAR) is an engineered receptor capable of anchoring a specific molecule (e.g., an antibody) that recognizes a tumor cell surface antigen to an immune cell (e.g., a T cell), allowing the immune cell to recognize a tumor antigen or viral antigen and kill a tumor cell or virus-infected cell. The CAR typically comprises, in order, an optional signal peptide, a polypeptide that binds to a tumor cell membrane antigen, such as a single chain antibody, a hinge region, a transmembrane region, and an intracellular signal region. In general, polypeptides that bind tumor cell membrane antigens are capable of binding with moderate affinity to membrane antigens that are widely expressed by tumor cells. The polypeptide that binds to a tumor cell membrane antigen may be a natural polypeptide or an artificial polypeptide; preferably, the synthetic polypeptide is a single chain antibody or Fab fragment.
The term "single chain antibody" (scFv) refers to an antibody fragment having the ability to bind antigen, which is formed by the amino acid sequence of the light chain variable region (VL region) and the amino acid sequence of the heavy chain variable region (VH region) of an antibody, which are joined by a hinge. In certain embodiments, the single chain antibody of interest (scFv) is from an antibody of interest. The antibody of interest may be a human antibody, including a human murine chimeric antibody and a humanized antibody. Antibodies may be secreted or membrane anchored; preferably of the membrane anchor type.
In order to improve the immunotherapy effect of the EHCAR-EK-28TIZ T cells, the PD1 antibody is expressed on the existing CAR-T cells, and the immune check point PD-1/PD-L1 pathway is inhibited while adoptive immune cell therapy is carried out, so that the functions of the residual tumor specific T cells can be activated in situ, the anti-tumor killing effect of endogenous cytotoxic T cells is increased, and the proliferation of the CAR-T cells in vivo is promoted, thereby improving the curative effect of specifically killing tumors.
Studies show that the IgG4Fc fragment of the PD1 antibody is easy to be recognized by mononuclear/macrophages to be phagocytosed, and the PD-1 antibody can well perform and not cause ADCC reaction after the base mutation modification is performed on the PD-1 antibody IgG4Fc fragment so as to meet the requirement of the PD-1 antibody expressed by T cells.
Accordingly, the present invention provides a PD1 antibody comprising an anti-PD 1 single chain antibody and IgG4Fc. In certain embodiments, the amino acid sequence of the IgG4Fc is shown as amino acid residues 267-495 of SEQ ID NO. 1; preferably, the coding sequence is shown as the base sequence of 799-1485 of SEQ ID NO. 2.
In certain embodiments, the anti-PD 1 single chain antibody (scFv) has an antibody light chain variable region (VL region) amino acid sequence as set forth in amino acid residues 21-131 of SEQ ID NO. 1; preferably, the coding sequence is shown as the 64-393 base sequence of SEQ ID NO. 2. In certain embodiments, the heavy chain variable region (VH region) amino acid sequence of the anti-PD 1 single-chain antibody is shown as 147-266 amino acid sequences of SEQ ID NO. 1; preferably, the coding sequence is shown as the base sequence of 439-798 of SEQ ID NO. 2. In certain embodiments, the anti-PD 1 single-chain antibody has an amino acid sequence as set forth in amino acid residues 21-266 of SEQ ID NO. 1; preferably, the coding sequence is shown as 61-798 base sequences of SEQ ID NO. 2.
In certain embodiments, the PD1 antibody further comprises a light chain signal peptide. In certain embodiments, the PD1 antibody comprises, from N-terminus to C-terminus, a light chain signal peptide, an anti-PD 1 single chain antibody, and IgG4Fc, in that order. In certain embodiments, the amino acid sequence of the light chain signal peptide is as shown in amino acid residues 1-20 of SEQ ID NO. 1; preferably, the coding sequence of the light chain signal peptide is shown as the 1 st to 60 th base sequence of SEQ ID NO. 2.
In certain embodiments, the PD1 antibody has an amino acid sequence as set forth in SEQ ID NO. 1 at amino acid positions 21-495 or as set forth in SEQ ID NO. 1; preferably, the coding sequence of the PD1 antibody is shown as SEQ ID NO. 2 at amino acid residues 61-1485, preferably as SEQ ID NO. 2.
The invention also includes the coding sequence of the PD1 antibody or its complement, which comprises at least the coding sequence of IgG4Fc or its complement as described herein. In certain embodiments, the coding sequence of the PD1 antibody comprises the sequence shown in the base sequence at positions 61-1485 of SEQ ID NO. 2, preferably the sequence shown in SEQ ID NO. 2.
The invention also includes a nucleic acid construct comprising the coding sequence of the PD1 antibodies of the invention or the complement thereof. Preferably, the nucleic acid construct is an expression vector or an integration vector for integrating the coding sequence or the complement thereof into a host cell.
The invention also provides a host cell comprising a nucleic acid construct as described herein.
The invention also provides the use of the PD1 antibodies, their coding sequences or complementary sequences, nucleic acid constructs and host cells for the preparation of a medicament for the treatment or prophylaxis of malignancies, particularly those associated with PD1, including but not limited to the various malignancies described herein.
The invention also provides T cells that self-express the immune checkpoint inhibitory PD1 antibodies and target chimeric antigen receptors of the ErbB receptor family. In the Chimeric Antigen Receptor (CAR), natural T1E and Herin are fused and expressed as antigen recognition regions of the CAR, and the chimeric antigen receptor and the Herin can complementarily recognize ErbB receptor families, so that the targeting range is enlarged.
The CARs of the invention generally contain an optional signal peptide sequence, a fusion protein of T1E and Herin, a hinge region, a transmembrane region, an intracellular co-stimulatory signaling domain, and an intracellular signaling domain.
The signal peptide is a short peptide chain (5-30 amino acids in length) that directs the transfer of a newly synthesized protein to the secretory pathway, often referred to as the N-terminal amino acid sequence (sometimes not necessarily at the N-terminus) of the newly synthesized polypeptide chain that directs the transmembrane transfer (localization) of the protein, which is responsible for directing the protein into subcellular organelles of the cell containing different membrane structures. The signal peptide may be a secretory signal peptide or a membrane-bound signal peptide. In certain embodiments of the invention, the signal peptide is a CD8 signal peptide, a CD28 signal peptide, or a CD4 signal peptide; more preferably a CD8 signal peptide. The amino acid sequence of the CD8 signal peptide can be shown as the amino acid residues 1 to 22 of SEQ ID NO. 5; in certain embodiments, the coding sequence is shown as bases 1-66 of SEQ ID NO. 6.
Native T1E and Herin are expressed fusion herein as the antigen recognition region of the CAR. The T1E is a chimeric polypeptide consisting of seven amino acids at the N-terminus of human transcription growth factor alpha (TGF alpha) and 48 amino acids at the C-terminus of Epidermal Growth Factor (EGF). Preferably, the amino acid sequence of T1E is shown as the 23 rd to 77 th amino acid residues of SEQ ID NO. 5; preferably, the coding sequence is shown as the 67 th to 231 th base sequence of SEQ ID NO. 6.
In the present invention, herin refers to 79 amino acids encoded by the eighth intron of Herstatin. Preferably, the amino acid sequence is shown as amino acid residues 93-171 of SEQ ID NO. 5. The present invention optimizes the codon encoding the amino acid of Herin. Thus, the preferred nucleotide sequence of Herin of the present invention is as shown in SEQ ID NO: the base sequence of the 277 th to 513 th positions is shown.
Typically, T1E and Herin may be linked by a rigid linker sequence. An illustrative example of a rigid linker sequence is a 2 or more repeat sequence with EAAAK as a unit, also referred to herein as EAAAK linker. Exemplary rigid linker sequences are shown as amino acid residues 78-92 of SEQ ID NO. 5; exemplary coding sequences are shown in the 232 th to 276 th base sequences of SEQ ID NO. 6.
The hinge region, as used herein, refers to the region between the functional regions of the heavy chains CH1 and CH2 of an immunoglobulin which is rich in proline, does not form an alpha helix, and is subject to stretching and some degree of warping, which facilitates complementary binding between the antigen binding site of the antibody and the epitope. Hinge regions suitable for use herein may be selected from any one or more of the extracellular hinge region of CD8, the IgG1FcCH2CH3 hinge region, the IgD hinge region, the extracellular hinge region of CD28, the IgG4Fc CH2CH3 hinge region, and the extracellular hinge region of CD 4. The hinge region is preferably a hinge region that is more than 50 amino acid residues in length, more preferably more than 80 amino acids in length. In certain embodiments, a CD8 a hinge region or an IgG4FcCH2CH3 hinge region is used herein. The amino acid sequence of an exemplary IgG4FcCH2CH3 hinge region is shown as amino acid residues 172-399 of SEQ ID NO. 5, and the coding sequence of an exemplary IgG4FcCH2CH3 hinge region is shown as 514-1197 of SEQ ID NO. 6.
The transmembrane region may be one of a CD28 transmembrane region, a CD8 transmembrane region, a cd3ζ transmembrane region, a CD134 transmembrane region, a CD137 transmembrane region, an ICOS transmembrane region, and a DAP10 transmembrane region; preferably a CD28 transmembrane region, preferably having an amino acid sequence as shown in amino acid residues 400-427 of SEQ ID NO. 5; in certain embodiments, the coding sequence is as shown in bases 1198-1281 of SEQ ID NO. 6.
Intracellular costimulatory signaling domains the intracellular domain comprising the costimulatory signaling molecule may be selected from the group consisting of the intracellular domains of CD28, CD134/OX40, CD137/4-1BB, lymphocyte-specific protein tyrosine kinase (LCK), inducible T cell costimulatory factor (ICOS) and DNAX activator protein 10 (DAP 10). In certain embodiments, the intracellular domain of the costimulatory signaling molecule is the intracellular domain of CD28, preferably having the amino acid sequence shown as amino acid residues 428-468 of SEQ ID NO. 5, and exemplary coding sequences shown as bases 1282-1404 of SEQ ID NO. 6.
The intracellular signaling domain is preferably an immunoreceptor tyrosine-activating motif, which may be a cd3ζ intracellular signaling domain or an fcsriy intracellular signaling domain; preferably a CD3 zeta intracellular signaling domain, preferably the amino acid sequence of said CD3 zeta intracellular signaling domain is as described in SEQ ID NO. 5, 469-580; in certain embodiments, the coding sequence is as shown in bases 1405-1740 of SEQ ID NO. 6.
In certain embodiments, the chimeric antigen receptor comprises, in order from N-terminus to C-terminus: an optional CD signal peptide, T1E, EAAAK linker, herin, igG4Fc CH2CH3 hinge region, CD28 transmembrane region, intracellular domain of CD28, and cd3ζ intracellular signal domain; preferably, the amino acid sequence of the chimeric antigen receptor is as shown in amino acid residues 23-580 of SEQ ID NO. 5. In certain embodiments, the chimeric antigen receptor further comprises a signal peptide, preferably the amino acid sequence of the chimeric antigen receptor is shown in SEQ ID NO. 5.
It is to be understood that the present invention also includes chimeric antibody receptors described herein and coding sequences thereof.
The above-described portions forming the chimeric antigen receptor herein, such as the signal peptide, T1E, herin, hinge region, transmembrane region, intracellular co-stimulatory signaling domain, and intracellular signaling domain, and the like, may be directly linked to each other or may be linked by a linker sequence. The linker sequences may be linker sequences suitable for antibodies as known in the art, such as G and S containing linker sequences. The length of the linker may be 3 to 25 amino acid residues, for example 3 to 15, 5 to 15, 10 to 20 amino acid residues. In certain embodiments, the linker sequence is a glycine linker sequence. The number of glycine in the linker sequence is not particularly limited, and is usually 2 to 20, for example 2 to 15, 2 to 10, 2 to 8. In addition to glycine and serine, other known amino acid residues may be contained in the linker, such as alanine (A), leucine (L), threonine (T), glutamic acid (E), phenylalanine (F), arginine (R), glutamine (Q), etc.
It will be appreciated that in gene cloning operations, it is often necessary to design suitable cleavage sites, which tend to introduce one or more unrelated residues at the end of the expressed amino acid sequence, without affecting the activity of the sequence of interest. To construct fusion proteins, facilitate expression of recombinant proteins, obtain recombinant proteins that are automatically secreted outside of the host cell, or facilitate purification of recombinant proteins, it is often desirable to add some amino acid to the N-terminus, C-terminus, or other suitable region within the recombinant protein, including, for example, but not limited to, suitable linker peptides, signal peptides, leader peptides, terminal extensions, and the like. Thus, the amino-or carboxy-terminus of a CAR herein can also contain one or more polypeptide fragments as protein tags. Any suitable label may be used herein. For example, the tag may be FLAG, HA, HA1, c-Myc, poly-His, poly-Arg, strep-TagII, AU1, EE, T7,4A6, ε, B, gE, and Ty1. These tags can be used to purify proteins.
Also included herein are polynucleotide sequences encoding the chimeric antigen receptors. The polynucleotide sequences herein may be in the form of DNA or RNA. DNA forms include cDNA, genomic DNA, or synthetic DNA. The DNA may be single-stranded or double-stranded.
The polynucleotide sequences described herein can generally be obtained using PCR amplification methods. Specifically, primers can be designed based on the nucleotide sequences disclosed herein and amplified to obtain the relevant sequences using a commercially available cDNA library or a cDNA library prepared by conventional methods known to those skilled in the art as a template. When the sequence is longer, it is often necessary to perform two or more PCR amplifications, and then splice the amplified fragments together in the correct order. For example, in certain embodiments, the polynucleotide sequence encoding the fusion proteins described herein is shown in SEQ ID NO. 6.
Also included herein are nucleic acid constructs comprising a polynucleotide sequence encoding the chimeric antigen receptor or a polynucleotide sequence encoding the PD1 antibody described herein, and one or more regulatory sequences operably linked to these sequences. In certain embodiments, the nucleic acid construct is an expression cassette.
The regulatory sequence may be a suitable promoter sequence. The promoter sequence is typically operably linked to the coding sequence of the protein to be expressed. The promoter may be any nucleotide sequence that exhibits transcriptional activity in the host cell of choice including mutant, truncated, and hybrid promoters, and may be obtained from genes encoding extracellular or intracellular polypeptides either homologous or heterologous to the host cell.
The regulatory sequence may also be a suitable transcription terminator sequence, a sequence recognized by a host cell to terminate transcription. The terminator sequence is operably linked to the 3' terminus of the nucleotide sequence encoding the polypeptide. Any terminator which is functional in the host cell of choice may be used herein.
In certain embodiments, the nucleic acid construct is a vector. In particular, the coding sequence of the CAR or the coding sequence of the PD1 antibody herein can be cloned into many types of vectors, for example, such types of vectors include, but are not limited to, plasmids, phagemids, phage derivatives, animal viruses, and cosmids. The vector may be an expression vector. The expression vector may be provided to the cell as a viral vector. Viruses that may be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpesviruses, and lentiviruses.
In general, suitable vectors comprise an origin of replication functional in at least one organism, a promoter sequence, a convenient restriction enzyme site and one or more selectable markers. For example, in certain embodiments, the invention uses a retroviral vector comprising a replication initiation site, a 3'LTR, a 5' LTR, the coding sequences for CARs described herein or the coding sequences for PD1 antibodies, and optionally a selectable marker.
Suitable promoters include, but are not limited to, the immediate early Cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operably linked thereto. Another example of a suitable promoter is extended growth factor-1α (EF-1α). However, other constitutive promoter sequences may also be used, including but not limited to the simian virus 40 (SV 40) early promoter, the mouse mammary carcinoma virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the epstein barr virus immediate early promoter, the ruses sarcoma virus promoter, and human gene promoters such as but not limited to the actin promoter, the myosin promoter, the heme promoter, and the creatine kinase promoter. Further, the use of inducible promoters is also contemplated. The use of an inducible promoter provides a molecular switch that is capable of switching on expression of a polynucleotide sequence operably linked to the inducible promoter when expressed for a period of time and switching 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.
In certain embodiments, various promoter sequences published by CN201510021408.1 can be used, including but not limited to the CCEF promoter comprising the mCMV enhancer, the hCMV enhancer and the EF 1. Alpha. Promoter shown in SEQ ID NO. 5 of this application; the TCEF promoter shown in SEQ ID NO. 7 and containing the CD3e enhancer, the mCMV enhancer, the hCMV enhancer and the EF1 alpha promoter; the CCEFI promoter shown in SEQ ID NO. 8 and containing the mCMV enhancer, the hCMV enhancer and the EF1 alpha promoter containing the intron; the TEFI promoter shown in SEQ ID NO. 3 and containing a CD3e enhancer and an EF1 alpha promoter containing an intron; and the TCEFI promoter shown in SEQ ID NO. 3 and containing the CD3e enhancer, the mCMV enhancer, the hCMV enhancer and the EF1 alpha promoter containing the intron. The entire contents of this application are incorporated herein by reference.
Selectable markers include either or both selectable marker genes or reporter genes to facilitate identification and selection of expressing cells from a population of cells infected with the viral vector. Useful selectable marker genes include, for example, antibiotic resistance genes, such as neo and the like. Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyl transferase, secreted alkaline phosphatase, or green fluorescent protein genes.
In certain embodiments, the coding sequences for the chimeric antigen receptor and the coding sequences for the PD1 antibody described herein are separately cloned into vectors (also referred to as integration vectors), particularly transposon vectors, for integration of the nucleic acid sequences of interest into the genome of the host cell. In certain embodiments, the transposon vector is a eukaryotic expression vector containing a transposable element selected from piggybac, sleep reliability, frog priority, tn5, or Ty. Such transposon vectors contain the 5 'inverted terminal repeat (5' LTR) of the corresponding transposon and the 3 'inverted terminal repeat (3' LTR) of the corresponding transposon. The transposase may be a transposase from a piggybac, sleep bearing, frog priority, tn5 or Ty transposase system. When transposases from different transposition systems are used, the sequences of the 5'LTR and 3' LTR in the vector are also changed accordingly to sequences that fit the transposition system, as can be readily determined by one skilled in the art. In certain embodiments, between the 5'ltr and the 3' ltr is an expression cassette for a CAR or antibody of the invention, comprising a corresponding promoter sequence, a coding sequence for the CAR or antibody, and a polyA tailing signal sequence.
In certain embodiments, the transposase is a transposase from the piggybac transposable system. Thus, in these embodiments, the transposon 5 'inverted terminal repeat and 3' inverted terminal repeat are the 5 'inverted terminal repeat and 3' inverted terminal repeat, respectively, of the piggybac transposon. In certain embodiments, the transposon 5' inverted terminal repeat is as shown in CN201510638974.7 (the contents of which are incorporated herein by reference) SEQ ID NO. 1. In certain embodiments, the transposon 3' inverted terminal repeat is as shown in CN 201510638974.7SEQ ID NO:4. In certain embodiments, the piggybac transposase is a transposase comprising a c-myc nuclear localization signal coding sequence. In certain embodiments, the coding sequence of the piggybac transposase is as set forth in CN 201510638974.7SEQ ID NO:5.
Promoters of the transposase coding sequence may be any of the promoters known in the art for controlling the expression of the transposase coding sequence. In certain embodiments, the expression of the transposase coding sequence is controlled using a CMV promoter. The sequence of the CMV promoter may be as shown in CN 201510638974.7SEQ ID NO:6.
In certain embodiments, the vector of the invention comprising the coding sequence for the chimeric antigen receptor is the pNB328 vector disclosed in CN 201510638974.7. The coding sequences for the chimeric antigen receptor of the invention can be prepared by methods conventional in the art and cloned into a suitable vector.
In certain embodiments, the vector for integrating the gene of interest into the genome of the host cell does not contain a transposase coding sequence. For example, such vectors may be obtained by removing the transposase coding sequence from the pNB328 vector. Typically, such vectors are used to integrate the coding sequence of the PD1 antibody and the coding sequence of a signal peptide (e.g., the coding sequence of a light chain signal peptide) into the genome of a host cell. Exemplary light chain signal peptides have the amino acid sequence shown in SEQ ID NO: 1-20 amino acid residues, and the coding sequence of an exemplary light chain signal peptide is shown in SEQ ID NO:2, 1-60 th base.
In certain embodiments, T cells described herein that are modified by a CAR gene and that are capable of expressing a PD1 antibody can be transformed into: a vector comprising a transposase coding sequence for integration into the expression cassette of a chimeric antigen receptor in the T cell genome, and a vector comprising no transposase coding sequence for integration into the expression cassette of a PD1 antibody described herein in the T cell genome.
Preferably, the T cells are transformed with a vector comprising a chimeric antigen receptor expression cassette constructed with the pNB328 vector as a backbone vector and a vector comprising a PD1 antibody expression cassette constructed with the pS328 vector (without the transposase coding sequence as compared to pNB 328) as a backbone vector. In certain embodiments, the chimeric antigen receptor has a coding sequence as set forth in SEQ ID NO. 6; the coding sequence of the PD1 antibody is shown as 61 th-1485 th base sequence of SEQ ID NO. 2. In certain embodiments, the signal peptide of the PD1 antibody is a light chain signal peptide in the vector comprising the expression cassette of the PD1 antibody. An exemplary light chain signal peptide may have an amino acid sequence as shown in amino acid residues 1-60 of SEQ ID NO. 2. More specifically, in certain embodiments, the transposase-containing vector having a chimeric antigen receptor coding sequence incorporated into the T cell genome comprises, in order, a 5'ltr, a promoter, a CD8 signal peptide coding sequence, a T1E, EAAAK linker, a Herin, a coding sequence for an IgG4Fc CH2CH3 hinge region, a coding sequence for a CD28 transmembrane region, a coding sequence for a CD28 intracellular domain, a coding sequence for a CD3 zeta intracellular signal domain, a polyA tailing signal sequence, a coding sequence for a 3' ltr and a transposase, and promoters thereof; the vector without transposase coding sequence, which incorporates the coding sequence of the PD1 antibody described herein in the T cell genome, contains a promoter, a coding sequence for a light chain signal peptide, a coding sequence for the PD1 antibody and a polyA tailing signal sequence in that order between the 5'LTR and the 3' LTR.
Preferably, the mass ratio of the vector containing the chimeric antigen receptor coding sequence to the vector containing the PD1 antibody coding sequence is 1-7 during transfection: 1 to 7, preferably 1:1 to 5, preferably 1:1 to 3, more preferably 1:1 to 2, more preferably 1:1.
methods of transfection are conventional in the art and include, but are not limited to: viral transduction, microinjection, particle bombardment, gene gun transformation, electrotransformation, and the like. In certain embodiments, electrotransfection is used to transfect the vector into a cell of interest.
The cells of interest may be a variety of T cells well known in the art, including but not limited to T cells of mixed cell populations such as peripheral blood T lymphocytes, cytotoxic killer T Cells (CTLs), helper T cells, suppressor/regulatory T cells, γδ T cells, and cytokine-induced killer Cells (CIKs), tumor Infiltrating Lymphocytes (TILs), and the like. In certain embodiments, the T cells may be derived from PBMCs of B cell malignancy patients. In certain embodiments, the T cell is a primary culture T cell.
The invention also provides a composition comprising a vector comprising the chimeric antigen receptor expression cassette described herein and a vector comprising the expression cassette of the PD1 antibody described herein. Suitable agents may also be included in the composition, including but not limited to agents for transfection.
The invention also provides a kit comprising a vector comprising the chimeric antigen receptor expression cassette described herein and a vector comprising the expression cassette of the PD1 antibody described herein, or a composition described herein. The kit may also be provided with reagents or instruments for transferring the vector into cells.
It is to be understood that the expression cassettes described herein contain at least a suitable promoter and polyA tailing signal sequence in addition to the coding sequences for the CARs or antibodies described herein.
The invention also provides a pharmaceutical composition comprising a T cell as described herein or a PD1 antibody expressed by the T cell. The pharmaceutical composition may contain suitable pharmaceutically acceptable carriers or excipients. The pharmaceutical composition contains a therapeutically or prophylactically effective amount of T cells. The therapeutically or prophylactically effective amount of T cells can be determined based on factors such as the patient's condition.
The invention also provides the use of a T cell or a pharmaceutical composition thereof or the T cell and an expressed PD1 antibody thereof as described herein in the manufacture of a medicament for the treatment or prevention of a malignancy (cancer); preferably, the malignancy is a tumor associated with PD1 and/or ErbB receptors; more preferably, the cancer cell surface of the malignancy abnormally expresses at least one EGFR family member protein. More preferably, the malignancy is selected from: liver cancer, adenocarcinoma, lung cancer, colon cancer, rectal cancer, carcinoma of large intestine, breast cancer, ovarian cancer, cervical cancer, gastric cancer, head and neck cancer, cholangiocarcinoma, gall bladder cancer, melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, hodgkin's lymphoma, esophageal cancer, pancreatic cancer, or prostate cancer.
Embodiments of the present invention will be described in detail below with reference to examples. It will be appreciated by those skilled in the art that the following examples are illustrative of the present invention and should not be construed as limiting the scope of the invention. The specific techniques or conditions are not noted in the examples, and are carried out according to techniques or conditions described in the literature in the art (for example, refer to J. Sam Brookfield et al, ind. Molecular cloning Experimental guidelines, third edition, scientific Press) or according to the product specifications. The reagents or apparatus used were conventional products commercially available without the manufacturer's attention.
Example 1: construction of recombinant plasmids pS328-antiPD1, pS328-antiPD1-wt and pNB328-EHCAR-EK-28TIZ
The EHCAR-EK-28TIZ gene, the anti-PD1 gene and the anti-PD1-wt gene are synthesized by Shanghai JieRui biosystems, and the structural modes are shown in figure 1. Each gene was separately loaded into pNB328 and pS328 vectors (the structure and sequence of pNB328 is referred to as CN 201510638974.7; pS328 lacks PB transposon sequences compared to pNB328 and other elements are identical to pNB328 vectors) double digested with EcoR1+SalI, and plasmids were constructed, designated pNB328-EHCAR-EK-28TIZ, pS328-antiPD1 and pS328-antiPD1-wt, respectively.
The nucleotide sequence of the light chain signal peptide in the structure pattern diagram is shown as the 1 st to 60 th base sequence of SEQ ID NO. 2; the coding of the Anti-PD1-wt is shown as SEQ ID NO. 4 (the amino acid sequence is shown as amino acid residues 21-495 of SEQ ID NO. 3); the nucleotide sequence of the Anti-PD1 is shown as the 61 st to 1488 th base sequence of SEQ ID NO. 2; the nucleotide sequence of the CD8 signal peptide is shown as the 1 st to 66 th base sequence of SEQ ID NO. 6; the nucleotide sequence of the T1E is shown as the 67 th to 231 th base sequence of SEQ ID NO. 6; the nucleotide sequence of the Herin is shown as 232 th-276 th base sequence of SEQ ID NO. 6; the nucleotide sequence of EAAAK-linker (EK-linker) is shown as the base sequence of 277-513 th bit of SEQ ID NO. 6; the nucleotide sequence of the membrane-spanning region of the mIgG4Fc CH2CH3 hinge is shown as the 514 th-1197 th base sequence of SEQ ID NO. 6; the nucleotide sequence of the CD28 transmembrane region (CD 28 TM) is shown as the base sequences 1198-1281 at positions 1-66 of SEQ ID NO. 6; the nucleotide sequence of the CD28 intracellular co-stimulatory signal structure region (CD 28 IC) is shown as the 1282-1404 base sequence of SEQ ID NO. 6; the nucleotide sequence of the CD3 zeta intracellular signal domain is shown as the 1405 th to 1740 th base sequence of SEQ ID NO. 6. The promoter sequence and polyA tailing signal sequence are not shown in the structural schematic diagrams, and are located between the 5'LTR and the signal peptide sequence and before the 3' LTR, respectively.
Example 2: positive rate and antibody expression quantity measurement of chimeric antigen receptor modified T cells constructed by pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 plasmids with different mass ratios
The CART cell construction was performed by setting the amounts of pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 plasmids to 7 ratios of 1ug+7ug, 2ug+6ug, 3ug+5ug, 4ug+4ug, 5ug+3ug, 6ug+2ug, 7ug+1ug, respectively. The construction method comprises the following steps:
peripheral Blood Mononuclear Cells (PBMCs) were isolated from Shanghai cell therapy production centers. Culturing PBMC for 2-4h in an adherence way, wherein non-adherence suspension cells are initial T cells, collecting the suspension cells into a 15ml centrifuge tube, centrifuging for 3min at 1200rmp, discarding the supernatant, adding physiological saline, centrifuging for 3min at 1200rmp, discarding the physiological saline, and repeating the steps; eight 1.5ml centrifuge tubes were taken and 5X 10 tubes were added to each tube 6 The individual cells, accession numbers a, b, c, d, e, f, g and h,1200rmp, centrifuge for 3min, discard supernatant, take electrotransfer kit (from Lonza corporation), add electrotransfer reagent 100ul in proportion to each tube, add recombinant plasmids pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 of different mass ratios to a, b, c, d, e, f and g tube respectively, add 6ug control plasmid (pNB 328, for construction of Mock-T cells) to h tube; transferring the mixed solution to an electric rotating cup, putting the electric rotating cup into an electric rotating instrument, selecting a required program, and performing electric shock; transferring the electrotransformed cell suspension to a six-well plate (AIM-V culture solution containing 2% FBS) added with a culture solution by using a micropipette in a kit, uniformly mixing, placing the mixture in a 37 ℃ and 5% CO2 incubator for culture, adding the stimulating factors IL-2 and EGFR/anti-CD28 after six hours, culturing at 37 ℃ and 5% CO2 for 3-4 days, observing the growth condition of T cells, and obtaining the EHCAR-EK-28TIZ T cells expressing PD1 antibodies. And respectively detecting the positive rate and the antibody secretion amount of the CAR T cells constructed under 7 proportions.
1. Flow detection of CAR T cell positive rate
The seven CAR-T and Mock-T cells were collected and divided into two parts, each 1X 10 6 Cells were washed twice with saline, resuspended in 100ul saline, one portion added 1ug EGFR-biotin and the other portion not added, and incubated at 4℃for 30 minutes. The cells were resuspended in 100ul of saline, and 1ul of streptomycin-PE antibody was added and incubated at 4℃for 30 min. Washing twice with normal saline, and checking in a machine, wherein only secondary antibody is added as a control.
As shown in FIG. 2A, the amounts of pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 plasmids gave the highest positive rate of CAR-T cells constructed in the form of 7ug+1ug.
ELISA to detect the expression level of EHCAR-EK-28 TIZ-anti-iPD 1T cell antibody.
(1) PD1 antigen was diluted to 0.5ug/ml (5 ul+1ml coating) with coating solution, and the enzyme-labeled reaction plate was coated at 100 ul/well overnight at 4 ℃.
(2) The cells were washed 5 times with PBST for 3 minutes each, and then dried with absorbent paper at 200 ul/well.
(3) 100ul of blocking solution was added to each well and incubated at 37℃for 1 hour.
(4) The cells were washed 5 times with PBST for 3 minutes each, and then dried with absorbent paper at 200 ul/well.
(5) Samples and standards were added, 100 ul/well, multiplex wells and control wells were set, and incubated for 1 hour at 37 ℃.
(6) The cells were washed 5 times with PBST for 3 minutes each, and then dried with absorbent paper at 200 ul/well.
(7) Blocking solution IgG F4HRP1: diluted at 30000, 100 ul/well, incubated at 37℃for 45 min.
(8) The cells were washed 5 times with PBST for 3 minutes each, and then dried with absorbent paper at 200 ul/well.
(9) Adding the color development liquid TMB,100 ul/hole, and developing for 10-15min at 37 ℃ in dark.
The reaction was stopped by adding a stop solution, 50 ul/well.
OD value is measured at 450nm on an enzyme labeling instrument, a standard curve is drawn, and PD1 antibody concentration is calculated.
As shown in FIG. 2B, the amounts of pNB328-EHCAR-EK-28TIZ and pS328-antiPD1 plasmids were the highest with respect to the amount of antibody secreted by CAR-T cells constructed in the form of 1 ug+7ug.
And 4ug pNB328-EHCAR-EK-28TIZ and 4ug pS 328-anti-iPD 1 are selected to construct EHCAR-EK-28 TIZ-anti-iPD 1T cells with best effect by combining positive rate and antibody secretion amount results.
Example 3: construction of EHCAR-EK-28TIZ T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells and determination of the Positive Rate and antibody expression level
6ug pNB328-EHCAR-EK-28TIZ plasmids were used to construct EHCAR-EK-28TIZ T cells, and 4ug pNB328-EHCAR-EK-28TIZ and 4ug pS328-antiPD1 plasmids were used to construct EHCAR-EK-28TIZ-antiPD1T cells, respectively, in the same manner as in example 2.
The positive rates of EHCAR-EK-28TIZ T cells and EHCAR-EK-28TIZ-antiPD1T cells were flow-tested as described in example 2. The results are shown in fig. 3A, and the self-expression of PD1 antibody did not decrease the positive rate of CART cells.
ELISA was used to detect the amount of EHCAR-EK-28TIZ-antiPD1T cell antibody expression, and the results are shown in FIG. 3B as described in example 2.
Example 4: comparison of EHCAR-EK-28TIZ and EHCAR-EK-28 TIZ-anti-iPD 1T cell proliferation rates
Each taking 3×10 5 Individual cells example 3 EHCAR-EK-28TIZ T cells, EHCAR-EK-28 TIZ-anti-iPD 1T cells and Mock-T cells from example 2 were cultured to day 8 and placed in 12 well plates for culture in 1ml volumes. A96-well white opaque plate was prepared, and 80. Mu.L of each cell-containing culture solution was added to each well from the three groups of cells, and 80. Mu.L of the nutrient solution was simultaneously fed to the original 12-well plate. Then 80. Mu.L CellTiter-Glo reagent was added to the 96-well plate, mixed well on a shaker for 2min, and incubated at room temperature for 10min, and the Luc fluorescence value was read by an ELISA reader. The CellTiter-Glo Luminescent Cell Viability Assay kit used was purchased from Promega corporation. Cells cultured in 12-well plates were sampled daily on days 9, 10, 11, 12, and 13 of culture, and cell proliferation curves were plotted according to fluorescence values as measured by the procedure described above.
The results are shown in FIG. 4, where the proliferation rate of EHCAR-EK-28 TIZ-anti-iPD 1T cells is significantly higher than that of EHCAR-EK-28TIZ T cells, indicating that expression of PD1 antibodies can promote proliferation of CAR-T cells.
Example 5: cell phenotyping assays of EHCAR-EK-28TIZ and EHCAR-EK-28TIZ-antiPD1T cells
Two EHCAR-EK-28TIZ T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells obtained in example 3 were collected and counted 1X 10 6 The individual cells/tubes were added to 6 1.5ml EP tubes, washed twice with PBS and centrifuged at 1200rpm for 5min, discarding the supernatant; wherein, 2 tubes are respectively added with the streaming antibodies anti-CD107 alpha-PE and anti-CD69-PE for detecting the phenotype of activated T cells, 1 tube is added with the streaming antibodies anti-CD45RO-PECy5+anti-CD197-FITC+anti-CD62L-PE for detecting the phenotype of memory T cells, 1 tube is respectively added with the streaming antibodies anti-PD1-PE for detecting the phenotype of inhibitory T cells, and the other 2 tubes are respectively added with isotype control streaming antibodies IgG1-PE and IgG1-PE+IgG2a-PECy5+IgG2a-PE, each antibody 2 μl (all purchased from Jackson ImmunoResearch company) and are subjected to flick precipitation to be uniformly mixed; after incubation for 30min at room temperature in the absence of light, PBS was washed once, centrifuged at 1200rpm for 5min, 400. Mu.l of physiological saline was added to the supernatant, and the cells were transferred to a flow tube and detected on-machine.
Experimental results show that the expression level of the senescent phenotype PD1 of the EHCAR-EK-28TIZ-antiPD1T cells is significantly lower than that of the EHCAR-EK-28TIZ (figure 5A); the expression level of activated phenotypes CD69 and CD107 a of EHCAR-EK-28TIZ-antiPD1T cells was higher than that of EHCAR-EK-28TIZ cells (FIGS. 5B and 5C); meanwhile, CD62L (L-selectin) is a marker of central memory T cells, CD197 is a marker of effector memory T cells, and the proportion of effector T cells in EHCAR-EK-28TIZ-antiPD1T cells is significantly higher than that in EHCAR-EK-28TIZ cells and Mock-T cells (FIG. 5D). These results demonstrate that expressing PD1 antibodies can inhibit the depletion of CAR-T cells, promote activation of CAR-T cells, and enhance their immune killing function.
Example 6: comparison of EHCAR-EK-28TIZ and EHCAR-EK-28 TIZ-anti-iPD 1T cell killing function
The in vitro killing activity of two EHCAR-EK-28TIZ T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells obtained in example 3 was detected by selecting MHC class I-typed matched effector cells and target cells using a real-time label-free cell function analyzer (RTCA) from the Eisen company, and the specific procedures were as follows:
(1) Zeroing: adding 50 μl of DMEM or 1640 culture solution into each well, placing into an instrument, selecting step 1, and zeroing;
(2) Target cell plating: human hepatoma cell HCCLM3, human hepatoma cell Hep3B and human non-small cell lung carcinoma H23 (purchased from American type culture Collection ATCC) at 10 per well 4 The individual cells/50. Mu.l were spread in a plate containing detection electrodes and left for several minutes until the cells were stableThen, putting the cells into an instrument, and starting the step 2 to culture the cells;
(3) Adding effector cells: after the target cells are cultured for 24 hours, the step 2 is paused, effector cells are added, 50 mu l of effector cells are added in each hole, the effective target ratio is set to be 4:1, the Mock T cells transferred into pNB328 empty vectors are used as a control, the step 3 is started, and after the co-culture is continued for 24 hours, the cell proliferation curve is observed;
the results are shown in FIG. 6. The killing effect of EHCAR-EK-28 TIZ-anti-ipd 1T cells from PD1 antibody-expressing EHCAR-EK-28TIZ T cells was significantly stronger on various tumor cells than EHCAR-EK-28TIZ T cells as well as control T cells.
Example 7: comparison of cytokine release by EHCAR-EK-28TIZ T cells and EHCAR-EK-28TIZ-antiPD1T cells under specific stimulation of EGFR antigen
Coating 96-well plate with EGFR antigen 5ug/ml, coating overnight at 4deg.C, washing 3 times with PBS, adding 1×10 5 (100 ul volumes) EHCAR-EK-28TIZ T cells and EHCAR-EK-28TIZ-antiPD1T cells prepared in example 3 and control Mock T cells (transferred into pNB328 empty vector) were cultured for 24h and cell supernatants were collected. The secretion of cytokines by these three T cells after stimulation with EGFR antigen was examined.
The results are shown in FIG. 7, where the IL-2 and IFN-gamma secretion amounts of EHCAR-EK-28 TIZ-anti-iPD 1T cells were significantly higher than those of EHCAR-EK-28TIZ T cells and Mock-T, indicating that self-expression of PD1 antibodies can promote cytokine secretion by CAR-T cells.
Example 8: in vivo anti-tumor effects of EHCAR-EK-28TIZ T cells, EHCAR-EK-28 TIZ-anti-iPD 1-wt T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells.
20 NSG mice with the age of 4-6 weeks are purchased and divided into 5 groups, 4 mice in each group are inoculated with hepatoma cell strain HCCLM3-LUC, and each group is 1 multiplied by 10 7 10 days after tumor formation, PBS (100 ul), mock-T, EHCAR-EK-28TIZ T cells, EHCAR-EK-28 TIZ-anti-iPD 1-wt T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells (1X 10) 7 Individual cells/mouse), the change in tumor fluorescence in mice was recorded by observation.
The results show that PBS, mock-T, EHCAR-EK-28 TIZ-anti-iPD 1-wt T cells have no therapeutic effect on tumor models, and EHCAR-EK-28TIZ T cells and EHCAR-EK-28 TIZ-anti-iPD 1T cells have good anti-tumor effects, but the EHCAR-EK-28 TIZ-anti-iPD 1T cells have obviously better effects. As shown in particular in fig. 8.
Although specific embodiments of the invention have been described in detail. Those skilled in the art will understand. Numerous modifications and substitutions of details are possible in light of all the teachings disclosed, and such modifications are contemplated as falling within the scope of the present invention. The full scope of the invention is given by the appended claims and any equivalents thereof.
Sequence listing
<110> Shanghai cell therapy institute
SHANGHAI ENGINEERING RESEARCH CENTER FOR CELL THERAPY GROUP Co.,Ltd.
<120> CAR-T cells targeting ErbB receptor family and self-expressing PD-1 antibodies and uses thereof
<130> 17A010
<160> 6
<170> SIPOSequenceListing 1.0
<210> 1
<211> 495
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 1
Met Glu Ala Pro Ala Gln Leu Leu Phe Leu Leu Leu Leu Trp Leu Pro
1 5 10 15
Asp Thr Thr Gly Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser
20 25 30
Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Lys Gly
35 40 45
Val Ser Thr Ser Gly Tyr Ser Tyr Leu His Trp Tyr Gln Gln Lys Pro
50 55 60
Gly Gln Ala Pro Arg Leu Leu Ile Tyr Leu Ala Ser Tyr Leu Glu Ser
65 70 75 80
Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr
85 90 95
Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr Cys
100 105 110
Gln His Ser Arg Asp Leu Pro Leu Thr Phe Gly Gly Gly Thr Lys Val
115 120 125
Glu Ile Lys Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly
130 135 140
Gly Ser Gln Val Gln Leu Val Gln Ser Gly Val Glu Val Lys Lys Pro
145 150 155 160
Gly Ala Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr
165 170 175
Asn Tyr Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu
180 185 190
Trp Met Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu
195 200 205
Lys Phe Lys Asn Arg Val Thr Leu Thr Thr Asp Ser Ser Thr Thr Thr
210 215 220
Ala Tyr Met Glu Leu Lys Ser Leu Gln Phe Asp Asp Thr Ala Val Tyr
225 230 235 240
Tyr Cys Ala Arg Arg Asp Tyr Arg Phe Asp Met Gly Phe Asp Tyr Trp
245 250 255
Gly Gln Gly Thr Thr Val Thr Val Ser Ser Glu Ser Lys Tyr Gly Pro
260 265 270
Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Glu Gly Gly Pro Ser Val
275 280 285
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
290 295 300
Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu
305 310 315 320
Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
325 330 335
Thr Lys Pro Arg Glu Glu Gln Phe Gln Ser Thr Tyr Arg Val Val Ser
340 345 350
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
355 360 365
Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile
370 375 380
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
385 390 395 400
Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
405 410 415
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
420 425 430
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
435 440 445
Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg
450 455 460
Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
465 470 475 480
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
485 490 495
<210> 2
<211> 1488
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 2
atggaagccc cagctcagct tctcttcctc ctgctactct ggctcccaga taccaccgga 60
gaaattgtgt tgacacagtc tccagccacc ctgtctttgt ctccagggga aagagccacc 120
ctctcctgca gggccagcaa aggtgtcagt acatctggct atagttattt gcactggtat 180
caacagaaac ctggccaggc tcccaggctc ctcatctatc ttgcatccta cctagaatct 240
ggcgtcccag ccaggttcag tggtagtggg tctgggacag acttcactct caccatcagc 300
agcctagagc ctgaagattt tgcagtttat tactgtcagc acagcaggga ccttccgctc 360
acgttcggcg gagggaccaa agtggagatc aaaggtggag gcggttcagg cggaggtggc 420
agcggcggtg gcgggtcgca ggtgcagctg gtgcagtccg gcgtggaggt gaagaagcct 480
ggcgcctccg tcaaggtgtc ctgtaaggcc tccggctaca ccttcaccaa ctactacatg 540
tactgggtgc ggcaggcccc aggccaggga ctggagtgga tgggcggcat caacccttcc 600
aacggcggca ccaacttcaa cgagaagttc aagaaccggg tgaccctgac caccgactcc 660
tccaccacaa ccgcctacat ggaactgaag tccctgcagt tcgacgacac cgccgtgtac 720
tactgcgcca ggcgggacta ccggttcgac atgggcttcg actactgggg ccagggcacc 780
accgtgaccg tgtcctccga gtccaaatat ggtcccccat gcccaccatg cccagcacct 840
gagttcgagg ggggaccatc agtcttcctg ttccccccaa aacccaagga cactctcatg 900
atctcccgga cccctgaggt cacgtgcgtg gtggtggacg tgagccagga agaccccgag 960
gtccagttca actggtacgt ggatggcgtg gaggtgcata atgccaagac aaagccgcgg 1020
gaggagcagt tccagagcac gtaccgtgtg gtcagcgtcc tcaccgtcct gcaccaggac 1080
tggctgaacg gcaaggagta caagtgcaag gtctccaaca aaggcctccc gtcctccatc 1140
gagaaaacca tctccaaagc caaagggcag ccccgagagc cacaggtgta caccctgccc 1200
ccatcccagg aggagatgac caagaaccag gtcagcctga cctgcctggt caaaggcttc 1260
taccccagcg acatcgccgt ggagtgggag agcaatgggc agccggagaa caactacaag 1320
accacgcctc ccgtgctgga ctccgacggc tccttcttcc tctacagcag gctaaccgtg 1380
gacaagagca ggtggcagga ggggaatgtc ttctcatgct ccgtgatgca tgaggctctg 1440
cacaaccact acacacagaa gagcctctcc ctgtctctgg gtaaatga 1488
<210> 3
<211> 495
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 3
Met Glu Ala Pro Ala Gln Leu Leu Phe Leu Leu Leu Leu Trp Leu Pro
1 5 10 15
Asp Thr Thr Gly Glu Ile Val Leu Thr Gln Ser Pro Ala Thr Leu Ser
20 25 30
Leu Ser Pro Gly Glu Arg Ala Thr Leu Ser Cys Arg Ala Ser Lys Gly
35 40 45
Val Ser Thr Ser Gly Tyr Ser Tyr Leu His Trp Tyr Gln Gln Lys Pro
50 55 60
Gly Gln Ala Pro Arg Leu Leu Ile Tyr Leu Ala Ser Tyr Leu Glu Ser
65 70 75 80
Gly Val Pro Ala Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr
85 90 95
Leu Thr Ile Ser Ser Leu Glu Pro Glu Asp Phe Ala Val Tyr Tyr Cys
100 105 110
Gln His Ser Arg Asp Leu Pro Leu Thr Phe Gly Gly Gly Thr Lys Val
115 120 125
Glu Ile Lys Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly
130 135 140
Gly Ser Gln Val Gln Leu Val Gln Ser Gly Val Glu Val Lys Lys Pro
145 150 155 160
Gly Ala Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr
165 170 175
Asn Tyr Tyr Met Tyr Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu
180 185 190
Trp Met Gly Gly Ile Asn Pro Ser Asn Gly Gly Thr Asn Phe Asn Glu
195 200 205
Lys Phe Lys Asn Arg Val Thr Leu Thr Thr Asp Ser Ser Thr Thr Thr
210 215 220
Ala Tyr Met Glu Leu Lys Ser Leu Gln Phe Asp Asp Thr Ala Val Tyr
225 230 235 240
Tyr Cys Ala Arg Arg Asp Tyr Arg Phe Asp Met Gly Phe Asp Tyr Trp
245 250 255
Gly Gln Gly Thr Thr Val Thr Val Ser Ser Glu Ser Lys Tyr Gly Pro
260 265 270
Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser Val
275 280 285
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
290 295 300
Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu
305 310 315 320
Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
325 330 335
Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val Ser
340 345 350
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
355 360 365
Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile
370 375 380
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
385 390 395 400
Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
405 410 415
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
420 425 430
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
435 440 445
Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg
450 455 460
Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
465 470 475 480
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
485 490 495
<210> 4
<211> 1428
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 4
gaaattgtgt tgacacagtc tccagccacc ctgtctttgt ctccagggga aagagccacc 60
ctctcctgca gggccagcaa aggtgtcagt acatctggct atagttattt gcactggtat 120
caacagaaac ctggccaggc tcccaggctc ctcatctatc ttgcatccta cctagaatct 180
ggcgtcccag ccaggttcag tggtagtggg tctgggacag acttcactct caccatcagc 240
agcctagagc ctgaagattt tgcagtttat tactgtcagc acagcaggga ccttccgctc 300
acgttcggcg gagggaccaa agtggagatc aaaggtggag gcggttcagg cggaggtggc 360
agcggcggtg gcgggtcgca ggtgcagctg gtgcagtccg gcgtggaggt gaagaagcct 420
ggcgcctccg tcaaggtgtc ctgtaaggcc tccggctaca ccttcaccaa ctactacatg 480
tactgggtgc ggcaggcccc aggccaggga ctggagtgga tgggcggcat caacccttcc 540
aacggcggca ccaacttcaa cgagaagttc aagaaccggg tgaccctgac caccgactcc 600
tccaccacaa ccgcctacat ggaactgaag tccctgcagt tcgacgacac cgccgtgtac 660
tactgcgcca ggcgggacta ccggttcgac atgggcttcg actactgggg ccagggcacc 720
accgtgaccg tgtcctccga gtccaaatat ggtcccccat gcccaccatg cccagcacct 780
gagttcctgg ggggaccatc agtcttcctg ttccccccaa aacccaagga cactctcatg 840
atctcccgga cccctgaggt cacgtgcgtg gtggtggacg tgagccagga agaccccgag 900
gtccagttca actggtacgt ggatggcgtg gaggtgcata atgccaagac aaagccgcgg 960
gaggagcagt tcaacagcac gtaccgtgtg gtcagcgtcc tcaccgtcct gcaccaggac 1020
tggctgaacg gcaaggagta caagtgcaag gtctccaaca aaggcctccc gtcctccatc 1080
gagaaaacca tctccaaagc caaagggcag ccccgagagc cacaggtgta caccctgccc 1140
ccatcccagg aggagatgac caagaaccag gtcagcctga cctgcctggt caaaggcttc 1200
taccccagcg acatcgccgt ggagtgggag agcaatgggc agccggagaa caactacaag 1260
accacgcctc ccgtgctgga ctccgacggc tccttcttcc tctacagcag gctaaccgtg 1320
gacaagagca ggtggcagga ggggaatgtc ttctcatgct ccgtgatgca tgaggctctg 1380
cacaaccact acacacagaa gagcctctcc ctgtctctgg gtaaatga 1428
<210> 5
<211> 580
<212> PRT
<213> Artificial sequence (Artificial Sequence)
<400> 5
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala Ala Arg Pro Ser Val Val Ser His Phe Asn Asp Cys Pro Leu
20 25 30
Ser His Asp Gly Tyr Cys Leu His Asp Gly Val Cys Met Tyr Ile Glu
35 40 45
Ala Leu Asp Lys Tyr Ala Cys Asn Cys Val Val Gly Tyr Ile Gly Glu
50 55 60
Arg Cys Gln Tyr Arg Asp Leu Lys Trp Trp Glu Leu Arg Glu Ala Ala
65 70 75 80
Ala Lys Glu Ala Ala Ala Lys Glu Ala Ala Ala Lys Gly Thr His Ser
85 90 95
Leu Pro Pro Arg Pro Ala Ala Val Pro Val Pro Leu Arg Met Gln Pro
100 105 110
Gly Pro Ala His Pro Val Leu Ser Phe Leu Arg Pro Ser Trp Asp Leu
115 120 125
Val Ser Ala Phe Tyr Ser Leu Pro Leu Ala Pro Leu Ser Pro Thr Ser
130 135 140
Val Pro Ile Ser Pro Val Ser Val Gly Arg Gly Pro Asp Pro Asp Ala
145 150 155 160
His Val Ala Val Asp Leu Ser Arg Tyr Glu Gly Glu Ser Lys Tyr Gly
165 170 175
Pro Pro Cys Pro Pro Cys Pro Ala Pro Pro Val Ala Gly Pro Ser Val
180 185 190
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
195 200 205
Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro Glu
210 215 220
Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
225 230 235 240
Thr Lys Pro Arg Glu Glu Gln Phe Gln Ser Thr Tyr Arg Val Val Ser
245 250 255
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
260 265 270
Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr Ile
275 280 285
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
290 295 300
Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
305 310 315 320
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
325 330 335
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
340 345 350
Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser Arg
355 360 365
Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
370 375 380
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys Pro
385 390 395 400
Phe Trp Val Leu Val Val Val Gly Gly Val Leu Ala Cys Tyr Ser Leu
405 410 415
Leu Val Thr Val Ala Phe Ile Ile Phe Trp Val Arg Ser Lys Arg Ser
420 425 430
Arg Leu Leu His Ser Asp Tyr Met Asn Met Thr Pro Arg Arg Pro Gly
435 440 445
Pro Thr Arg Lys His Tyr Gln Pro Tyr Ala Pro Pro Arg Asp Phe Ala
450 455 460
Ala Tyr Arg Ser Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala
465 470 475 480
Tyr Gln Gln Gly Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg
485 490 495
Arg Glu Glu Tyr Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu
500 505 510
Met Gly Gly Lys Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn
515 520 525
Glu Leu Gln Lys Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met
530 535 540
Lys Gly Glu Arg Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly
545 550 555 560
Leu Ser Thr Ala Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala
565 570 575
Leu Pro Pro Arg
580
<210> 6
<211> 1740
<212> DNA
<213> Artificial sequence (Artificial Sequence)
<400> 6
atggccttac cagtgaccgc cttgctcctg ccgctggcct tgctgctcca cgccgccagg 60
ccgagcgtgg tgtcccattt taatgactgt cccctgtccc acgatgggta ctgcctccat 120
gatggtgtgt gcatgtatat tgaagcattg gacaagtatg catgcaactg tgttgttggc 180
tacatcgggg agcgatgtca gtaccgagac ctgaagtggt gggaactgcg cgaagctgcc 240
gctaaggagg ccgcagccaa agaggccgct gcaaagggca cccacagcct gcccccccgc 300
cccgccgccg tgcccgtgcc cctgcgcatg cagcccggcc ccgcccaccc cgtgctgagc 360
ttcctgcgcc ccagctggga cctggtgagc gccttctaca gcctgcccct ggcccccctg 420
agccccacca gcgtgcccat cagccccgtg agcgtgggcc gcggccccga ccccgacgcc 480
cacgtggccg tggacctgag ccgctacgag ggcgagtcca aatatggtcc cccatgccca 540
ccatgcccag cacctcccgt ggccggacca tcagtcttcc tgttcccccc aaaacccaag 600
gacactctca tgatctcccg gacccctgag gtcacgtgcg tggtggtgga cgtgagccag 660
gaagaccccg aggtccagtt caactggtac gtggatggcg tggaggtgca taatgccaag 720
acaaagccgc gggaggagca gttccagagc acgtaccgtg tggtcagcgt cctcaccgtc 780
ctgcaccagg actggctgaa cggcaaggag tacaagtgca aggtctccaa caaaggcctc 840
ccgtcctcca tcgagaaaac catctccaaa gccaaagggc agccccgaga gccacaggtg 900
tacaccctgc ccccatccca ggaggagatg accaagaacc aggtcagcct gacctgcctg 960
gtcaaaggct tctaccccag cgacatcgcc gtggagtggg agagcaatgg gcagccggag 1020
aacaactaca agaccacgcc tcccgtgctg gactccgacg gctccttctt cctctacagc 1080
aggctaaccg tggacaagag caggtggcag gaggggaatg tcttctcatg ctccgtgatg 1140
catgaggctc tgcacaacca ctacacacag aagagcctct ccctgtctct gggtaaaccc 1200
ttttgggtgc tggtggtggt tggtggagtc ctggcttgct atagcttgct agtaacagtg 1260
gcctttatta ttttctgggt gaggagtaag aggagcaggc tcctgcacag tgactacatg 1320
aacatgactc cccgccgccc cgggcccacc cgcaagcatt accagcccta tgccccacca 1380
cgcgacttcg cagcctatcg ctccagagtg aagttcagca ggagcgcaga cgcccccgcg 1440
taccagcagg gccagaacca gctctataac gagctcaatc taggacgaag agaggagtac 1500
gatgttttgg acaagagacg tggccgggac cctgagatgg ggggaaagcc gagaaggaag 1560
aaccctcagg aaggcctgta caatgaactg cagaaagata agatggcgga ggcctacagt 1620
gagattggga tgaaaggcga gcgccggagg ggcaaggggc acgatggcct ttaccagggt 1680
ctcagtacag ccaccaagga cacctacgac gcccttcaca tgcaggccct gccccctcgc 1740

Claims (22)

1. A T cell, wherein the T cell:
(1) Comprising a coding sequence for expressing a chimeric antigen receptor that targets the ErbB receptor family and a coding sequence for a secreted PD1 antibody; and/or
(2) Expression of chimeric antigen receptor and secreted PD1 antibodies targeting the ErbB receptor family;
the secretory PD1 antibody comprises an anti-PD 1 single-chain antibody and an IgG4Fc; wherein the amino acid sequence of the IgG4Fc is shown as amino acid residues 267-495 of SEQ ID NO. 1, the amino acid sequence of the light chain variable region of the anti-PD 1 single-chain antibody is shown as amino acid residues 21-131 of SEQ ID NO. 1, the amino acid sequence of the heavy chain variable region of the anti-PD 1 single-chain antibody is shown as amino acid sequences 147-266 of SEQ ID NO. 1,
the chimeric antigen receptor comprises a T1E, EAAAK joint, a Herin, an IgG4 CH2CH3 hinge region, a CD28 transmembrane region, a CD28 intracellular domain and a CD3 zeta intracellular signaling domain from the N end to the C end in sequence,
the amino acid sequence of the T1E is shown as the 23 rd to 77 th amino acid residues of SEQ ID NO. 5;
the amino acid sequence of the Herin is shown as amino acid residues 93-171 of SEQ ID NO. 5;
the amino acid sequence of the EAAAK linker is shown as the 78 th-92 th amino acid residue of SEQ ID NO. 5.
2. The T cell of claim 1, wherein the chimeric antigen receptor further comprises an N-terminal signal peptide, wherein the signal peptide is a CD8 signal peptide.
3. The T cell of claim 2, wherein the amino acid sequence of the CD8 signal peptide is shown as amino acid residues 1-22 of SEQ ID No. 5.
4. The T cell of claim 1, wherein the amino acid sequence of the hinge region of IgG4 CH2CH3 is shown as amino acid residues 172-399 of SEQ ID No. 5.
5. The T cell of claim 1, wherein the amino acid sequence of the CD28 transmembrane region is shown as amino acid residues 400-427 of SEQ ID No. 5.
6. The T cell of claim 1, wherein the amino acid sequence of the CD28 intracellular domain is shown as amino acid residues 428-468 of SEQ ID No. 5.
7. The T cell of claim 1, wherein the amino acid sequence of the cd3ζ intracellular signaling domain is depicted as amino acid residues 469-580 of SEQ ID No. 5.
8. The T cell of any one of claims 1-7, wherein the chimeric antigen receptor has one or more of the following characteristics:
the coding sequence of the signal peptide is shown as the 1 st to 66 th base sequence of SEQ ID NO. 6;
the coding sequence of the T1E is shown as the 67 th to 231 th base sequence of SEQ ID NO. 6;
the coding sequence of the Herin is shown in the 277 th to 513 th base sequence of SEQ ID NO. 6;
The coding sequence of the EAAAK linker is shown as 232 th-276 th base sequence of SEQ ID NO. 6;
the coding sequence of the hinge region is shown as 514 th to 1197 th base sequence of SEQ ID NO. 6;
the coding sequence of the transmembrane region is shown as the base sequence of 1198-1281 of SEQ ID NO. 6;
the coding sequence of the intracellular co-stimulatory signal domain is shown as the 1282 th to 1404 th base sequence of SEQ ID NO. 6; and
the coding sequence of the intracellular signal domain is shown as the 1405 th to 1740 th base sequence of SEQ ID NO. 6.
9. The T cell of claim 1, wherein the chimeric antigen receptor has an amino acid sequence as set forth in amino acid residues 23-580 of SEQ ID No. 5, or as set forth in SEQ ID No. 5.
10. The T cell of claim 9, wherein the chimeric antigen receptor has a nucleotide sequence as set forth in SEQ ID No. 6 at nucleotide sequences 67-1740 or as set forth in SEQ ID No. 6.
11. The T cell of claim 1, wherein the secreted PD1 antibody further comprises a light chain signal peptide.
12. The T cell of claim 11, wherein the amino acid sequence of the light chain signal peptide is set forth in base sequences 1-20 of SEQ ID No. 1.
13. The T cell of claim 1, wherein the anti-PD 1 single chain antibody has an amino acid sequence as set forth in amino acid residues 21-266 of SEQ ID No. 1.
14. The T cell of claim 13, wherein the anti-PD 1 single chain antibody has a coding sequence as set forth in base sequence nos. 61-798 of SEQ ID No. 2.
15. The T cell of claim 13, wherein the secreted PD1 antibody has an amino acid sequence as set forth in SEQ ID No. 1 at amino acid positions 21-495, or as set forth in SEQ ID No. 1.
16. The T cell of claim 15, wherein the secretory PD1 antibody has a coding sequence shown as amino acid residues 61-1485 of SEQ ID No. 2 or as SEQ ID No. 2.
17. A composition or kit, the composition comprising:
(1) A vector comprising an expression cassette for a chimeric antigen receptor as defined in any one of claims 1 to 10 for integration of said expression cassette into the genome of a host cell; and
(2) A vector comprising an expression cassette for a secreted PD1 antibody as defined in any one of claims 1, 11-16 for integration of said expression cassette into the genome of a host cell.
18. A pharmaceutical composition comprising the T cell of any one of claims 1-16 or the T cell and its expressed secreted PD1 antibody.
19. Use of the T cell of any one of claims 1-16 or the T cell and its expressed secreted PD1 antibody or a pharmaceutical composition thereof in the manufacture of a medicament for the treatment or prevention of cancer.
20. The use of claim 19, wherein the cancer is a cancer in which the surface of the cancer cell abnormally expresses at least one EGFR family member protein.
21. The use of claim 19, wherein the cancer is selected from the group consisting of: liver cancer, lung cancer, colon cancer, rectal cancer, colorectal cancer, breast cancer, ovarian cancer, cervical cancer, gastric cancer, head and neck cancer, bile duct cancer, gall bladder cancer, melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck squamous cell carcinoma, hodgkin's lymphoma, esophageal cancer, pancreatic cancer or prostate cancer.
22. The use of claim 19, wherein the cancer is an adenocarcinoma.
CN201711461266.6A 2017-12-28 2017-12-28 CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof Active CN109971724B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201711461266.6A CN109971724B (en) 2017-12-28 2017-12-28 CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof
PCT/CN2018/124316 WO2019129138A1 (en) 2017-12-28 2018-12-27 Car-t cells targeting erbb receptor family and self-expressing pd-1 antibody and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201711461266.6A CN109971724B (en) 2017-12-28 2017-12-28 CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof

Publications (2)

Publication Number Publication Date
CN109971724A CN109971724A (en) 2019-07-05
CN109971724B true CN109971724B (en) 2023-10-31

Family

ID=67066624

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201711461266.6A Active CN109971724B (en) 2017-12-28 2017-12-28 CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof

Country Status (2)

Country Link
CN (1) CN109971724B (en)
WO (1) WO2019129138A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230054413A1 (en) * 2019-12-13 2023-02-23 Samsung Bioepis Co., Ltd. Stable anti-pd1 antibody pharmaceutical formulations
CN111235113A (en) * 2020-01-21 2020-06-05 南京北恒生物科技有限公司 Immune cells comprising chimeric antigen receptors and uses thereof
CN112142854B (en) * 2020-09-18 2021-06-15 南京凯地生物科技有限公司 Immune regulation specific chimeric antigen receptor cell and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105153315A (en) * 2015-10-09 2015-12-16 重庆倍思益生物科技有限公司 Chimeric receptor combining immunosuppression receptor and tumor antigen receptor and application of chimeric receptor
WO2017181119A2 (en) * 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK3221355T3 (en) * 2014-11-20 2020-12-07 Hoffmann La Roche Combination therapy with T cell activating bispecific antigen binding molecules CD3 and folate receptor 1 (FolR1) as well as PD-1 axis binding antagonists
GB201513540D0 (en) * 2015-07-31 2015-09-16 King S College London Therapeutic agents

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105153315A (en) * 2015-10-09 2015-12-16 重庆倍思益生物科技有限公司 Chimeric receptor combining immunosuppression receptor and tumor antigen receptor and application of chimeric receptor
WO2017181119A2 (en) * 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Antibody-modified T cells:CARs take the seat for hematologic malignancies;Maus MV等;《Blood》;20141231;第123卷;第2625-2635页 *
嵌合抗原受体T细胞肿瘤免疫治疗的风险与对策;张苗苗等;《药学学报》;20160712(第07期);第1032-1038页 *
靶向EGFR家族成员分泌PD-1抗体的CAR-T细胞治疗肺癌的实验研究;胥阶英;中国优秀硕士学位论文全文数据库 医药卫生科技辑(第第7期期);E072-106 *

Also Published As

Publication number Publication date
CN109971724A (en) 2019-07-05
WO2019129138A1 (en) 2019-07-04

Similar Documents

Publication Publication Date Title
AU2017305366B2 (en) Anti-PD-L1 nanobody and use thereof
CN108341872B (en) Antibodies targeting BCMA and uses thereof
US11292841B2 (en) Anti-PD-1 nano-antibody and application thereof
CN109971712B (en) CAR-T cell specifically targeting CD19 antigen and stably expressing PD-1 antibody at high level and application thereof
CN113402609B (en) Canine antibodies with modified CH2-CH3 sequences
CN109721657B (en) Fusion protein for blocking PD-1/PD-L1 signal transduction pathway and activating T cells and application thereof
CN106715470B (en) Covalently bound diabodies immunoreactive with PD-1 and LAG-3 and methods of use thereof
CN112851816B (en) PD-L1 antibodies that bind canine PD-L1
WO2018233574A1 (en) Anti-pd-l1 humanized nanobody and use thereof
CN109971725B (en) Antibody-modified chimeric antigen receptor-modified T cells and uses thereof
WO2019020088A1 (en) Chimeric antigen receptor-modified t cell targeting mesothelin and use thereof
WO2019129146A1 (en) Egfr-specific car-t cell capable of autocrine secretion of cd47 antibody, and application of car-t cell
WO2021031716A1 (en) Anti-pd-l1 single domain antibodies
WO2019128994A1 (en) Muc1-specific car-t cell stably expressing pd-1 antibody and use thereof
CN108727504A (en) The fusion protein and its application of a kind of IFN and anti-PD-L1 antibody
CN109971724B (en) CAR-T cells targeting the ErbB receptor family and self-expressing PD-1 antibodies and uses thereof
WO2019128998A1 (en) Self-expressing pd-1 antibody, chimeric antigen receptor-modified t cells targeting mesothelin, and use thereof
WO2019129124A1 (en) T cell containing cd40 antibody and muc1 specific chimeric antigen receptor gene and use thereof
WO2021218874A1 (en) Bispecific antibody targeting human claudin and human pdl1 proteins, and application thereof
WO2019129174A1 (en) Car-t cell targeting cd19 and expressing cd40 antibody at high level of stability, and use thereof
CN111423512A (en) Multi-targeting fusion protein blocking vascular endothelial cell growth and activating T cells and pharmaceutical composition comprising the same
CN109971719B (en) Car-T cells that autocrine CD40 antibodies and target ErbB receptor family and uses thereof
WO2023241522A1 (en) T cell receptor targeting kras g12v mutant polypeptide, and use thereof
CN110177568A (en) Combination treatment for treating cancer
WO2019128996A1 (en) Mesothelin-specific car-t cell expressing cd47 antibody, and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP03 Change of name, title or address

Address after: No. 1585 Yuanguo Road, Jiading District, Shanghai, 201805

Patentee after: SHANGHAI CELL THERAPY Research Institute

Country or region after: China

Patentee after: Shanghai Cell Therapy Group Co.,Ltd.

Address before: No. 1585 Yuanguo Road, Jiading District, Shanghai, 201805

Patentee before: SHANGHAI CELL THERAPY Research Institute

Country or region before: China

Patentee before: SHANGHAI CELL THERAPY GROUP Co.,Ltd.