WO2021109977A1 - 一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用 - Google Patents

一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用 Download PDF

Info

Publication number
WO2021109977A1
WO2021109977A1 PCT/CN2020/133024 CN2020133024W WO2021109977A1 WO 2021109977 A1 WO2021109977 A1 WO 2021109977A1 CN 2020133024 W CN2020133024 W CN 2020133024W WO 2021109977 A1 WO2021109977 A1 WO 2021109977A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
car
sequence
cell
lentiviral
Prior art date
Application number
PCT/CN2020/133024
Other languages
English (en)
French (fr)
Inventor
李建强
何晋元
王琳
牛星
巴敏
王庆龙
Original Assignee
河北森朗生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 河北森朗生物科技有限公司 filed Critical 河北森朗生物科技有限公司
Priority to AU2020397271A priority Critical patent/AU2020397271B2/en
Publication of WO2021109977A1 publication Critical patent/WO2021109977A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the invention relates to the field of biotechnology, in particular to a chimeric antigen receptor T cell that can be efficiently prepared and applied safely, and a preparation method and application thereof.
  • CAR-T therapy is chimeric antigen receptor T-cell immunotherapy, the full English name is Chimeric Antigen Receptor T-Cell Immunotherapy. This is a new type of precision targeted therapy for tumor treatment. In recent years, it has achieved good results in clinical tumor treatment through optimization and improvement. It is a very promising, accurate, fast, efficient, and possible cure for cancer. New tumor immunotherapy methods.
  • the current conventional solution is to adopt suitable transfection reagents, maintain the best cell condition or choose the best transfection method; but these changes are still low in transfection efficiency.
  • the preparation cost is still high.
  • An object of the present invention is to provide a lentiviral recombinant vector.
  • the lentiviral recombinant vector provided by the present invention contains a CAR structure coding sequence and a promoter MND that drives the expression of the CAR structure coding sequence;
  • the nucleotide sequence of the promoter MND is 2838-3236 of sequence 1;
  • the CAR structure in turn includes an extracellular binding domain, one or more hinge domains or spacer domains, a transmembrane domain, one or more intracellular costimulatory signal transduction domains, and a primary signal transduction domain.
  • the extracellular domain is selected from at least one of the following target antigen antibodies or antigen-binding fragments, binding ligands or co-receptor extracellular domains: ⁇ folate receptor, 5T4, ⁇ v ⁇ 6 Integrin, BCMA, B7-H3, B7-H6, CAIX, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a, CD79b, CD123, CD138, CD171, CEA, CSPG4, EGFR, EGFR family including ErbB2 (HER2), EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, fetal AchR, FR ⁇ , GD2, GD3, Glypican-3 (GPC3), HLA-A1 +MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ES
  • the hinge domain or spacer domain is selected from at least one of the transmembrane domains of CD8 ⁇ , CD4, CD45, PD1 and CD152; but it is not limited thereto;
  • the transmembrane domain is selected from at least one of the transmembrane domains of CD8, CD4, CD45, PD1, and CD152; but not limited thereto;
  • the intracellular costimulatory signal transduction domain is selected from CD28, CD54 (ICAM), CD134 (OX40), CD137 (41BB), CD152 (CTLA4), CD273 (PD-L2), CD274 (PD-L1) and CD278 At least one of (ICOS); but not limited to this;
  • the primary signal transduction domain is selected from at least one of the primary signal transduction domain of CD3 ⁇ and the primary signal transduction domain of FcR ⁇ ; but it is not limited thereto.
  • the extracellular binding domain (also known as the antigen-specific binding domain) includes the light chain variable region of the CD19 antibody and the heavy chain variable region of the CD19 antibody;
  • the hinge domain is a CD8 hinge domain
  • the transmembrane domain is a CD8 transmembrane domain
  • the intracellular costimulatory signal transduction domain is 4-1BB;
  • the primary signal transduction domain is CD3 ⁇ .
  • amino acid sequence of the light chain variable region of the CD19 antibody is 23-129 of sequence 3 in the sequence listing;
  • amino acid sequence of the heavy chain variable region of the CD19 antibody is the 148-267th position in the sequence 3 in the sequence table;
  • the amino acid sequence of the CD8 hinge domain is positions 268-307 of sequence 3 in the sequence listing;
  • the amino acid sequence of the CD8 transmembrane domain is positions 308-335 of sequence 3 in the sequence table;
  • amino acid sequence of the intracellular costimulatory signal transduction domain 4-1BB is the 336-377th position in the sequence 3 in the sequence table;
  • amino acid sequence of the primary signal transduction domain CD3 ⁇ is positions 378-489 of sequence 3 in the sequence listing.
  • the nucleotide sequence of the CAR structure coding sequence is 3251-4717 of SEQ ID NO: 1.
  • the lentiviral recombination vector is the homologous recombination of DNA molecules at positions 2838-5863 of sequence 1 into a lentiviral expression vector to obtain a vector expressing the CAR structure; in the implementation of the present invention
  • the specific lentiviral expression vector used is the pDBR vector.
  • the lentiviral recombinant vector in the embodiment of the present invention is the lentiviral recombinant plasmid pLV-M19BBz (M19BBz), and its nucleotide sequence is sequence 1.
  • the recombinant virus packaged by the above-mentioned lentiviral recombinant vector is also within the protection scope of the present invention.
  • the above-mentioned recombinant virus is a virus capable of expressing the CAR structure shown at positions 3251-4717 of sequence 1, and capable of infecting immune effector cells.
  • the virus is lentivirus, herpes virus, macrophage virus, Epstein-Barr virus, hepatitis B virus, hepatitis C virus or HIV.
  • Recombinant cells containing the aforementioned lentiviral recombination vector are also within the protection scope of the present invention.
  • the aforementioned recombinant cell is an immune effector cell expressing the aforementioned lentiviral recombinant vector
  • the immune effector cells are cytotoxic T lymphocytes, NKT cells, NK cells, helper T cells or gamma delta T cells.
  • promoter MND to drive the expression of CAR structure in CAR-T cells is also within the scope of this protection.
  • the nucleotide sequence of the promoter MND is 2838-3236 of SEQ ID NO: 1.
  • Another object of the present invention is to provide a method for preparing CAR-T cells.
  • the method provided by the present invention includes the following steps: transfecting the recombinant virus obtained by packaging the above-mentioned lentiviral recombinant vector into a recombinant cell obtained by transfecting a T cell.
  • the present invention also provides the following product for treating tumors or having the function of treating tumors and having a low cytokine storm, which includes the above-mentioned lentiviral recombinant vector or the above-mentioned recombinant cell.
  • the invention also provides the following applications:
  • the present invention provides the application of the above-mentioned lentiviral recombinant vector, the above-mentioned recombinant virus or the above-mentioned recombinant cell in the preparation of products for treating tumors;
  • the present invention provides the application of the above-mentioned lentiviral recombinant vector, the above-mentioned recombinant virus or the above-mentioned recombinant cell in the preparation of products with tumor treatment and low cytokine storm functions.
  • the present invention also provides a method for reducing the production of cytokine storm in CAR-T cell therapy, which is to use the promoter MND to drive the expression of CAR structure in T cells;
  • the nucleotide sequence of the promoter MND is 2838-3236 of SEQ ID NO: 1.
  • the above includes the following steps: 1) preparing the lentiviral recombinant vector in the first object;
  • the present invention also provides a method for treating tumors, which includes the following steps:
  • the present invention provides genetically engineered immune effector cells comprising a vector designed to express a chimeric antigen receptor that directs cytotoxicity to tumor cells.
  • CARs are molecules that combine an antibody based on the specificity of a target antigen (such as a tumor antigen) with a T cell receptor activating intracellular domain to produce A chimeric protein with specific anti-tumor cell immune activity.
  • chimeric as used herein describes being composed of parts of different proteins or composed of DNA from different sources.
  • the vectors considered herein include promoters such as MND, EF1 ⁇ , CMV, PGK, etc. and polynucleotides encoding CAR (including colony stimulating factor signal peptide, FMC63 single-chain antibody sequence, CD8 hinge region and transmembrane region sequence, 4- 1BB costimulatory signal domain and CD3 ⁇ signal domain as well as T2A and tEGFR).
  • the CAR considered herein includes an extracellular domain (also called a binding domain or an antigen-specific binding domain) that binds to a specific target antigen, a transmembrane domain, and an intracellular signal transduction domain.
  • the binding of the antigen-binding domain of the CAR to its target antigen on the surface of the target cell results in the aggregation of the CAR and delivers an activation stimulus to the CAR-containing cells.
  • the main characteristic of CAR is its ability to redirect immune effector cells specifically, thereby triggering proliferation, cytokine production, phagocytosis, or the ability to mediate the expression of targets in a major histocompatibility complex (MHC)-independent manner
  • MHC major histocompatibility complex
  • the production of molecules for cell death of antigens uses the cell-specific targeting ability of monoclonal antibodies, soluble ligands or cell-specific co-receptors.
  • Figure 1 shows the CAR vector construction of different promoters EF1 ⁇ and MND, (a) CAR vector map using EF1 ⁇ promoter; (b) CAR vector map using MND promoter, (c) CAR vector map of different promoters EF1 ⁇ and MND Schematic diagram of the structure of the CAR carrier.
  • FIG. 1 shows the measurement of transfection efficiency.
  • Figure 3 is a flow chart of the determination of transfection efficiency.
  • Figure 4 shows the difference in the expression of CAR molecules in T cells after transduction of E19BBz and M19BBz.
  • Figure 5 shows the antigen-specific tumor clearance by T cells expressing CAR with different promoters and the amount of cytokine produced by CAR-T cells in the corresponding experiments.
  • Figure 6 shows the regression effect of CAR-modified T cells under different promoters on tumor cells in tumor model mice after adoptive transfer.
  • Figure 7 shows the transfection efficiency of the two CAR-T cells in clinical trials, Relative MFI, the expansion rate of T cell culture in vitro and the expansion rate of CAR-T in vivo.
  • Figure 8 is a summary of the patient's response after treatment in the clinical trial, (a) the patient's body temperature change within 14 days after reinfusion; (b) the highest fever temperature of the patient after treatment; (c) the CRS series statistics of the patient after treatment .
  • Figure 9 shows the long-term clinical results of CAR-T cells produced by two promoters after treatment.
  • the lentiviral recombinant plasmid pLV-M19BBz (M19BBz) containing the MND promoter (as shown in Figure 1b and 1c below) has a nucleotide sequence of sequence 1 (full gene synthesis).
  • This recombinant plasmid can also be used for sequence 1 to 2838- Homologous recombination of 5863 DNA molecules into the pDBR vector (Calderón-Gómez, E., et al., Reprogrammed quiescent B cells provide an effective cellular therapy against encephalomyelitis.European encephalomyelitis.European of Immunology, 2011.41(6) 1696-1708.).
  • positions 2838-3236 of sequence 1 are MND promoters
  • positions 3251-3316 are the nucleic acid encoding colony stimulating factor signal peptide (amino acid sequence is positions 1-22 of sequence 3)
  • positions 3317-3637 are extra-membrane antigen binding Region CD19 antibody light chain variable region VL19 encoding nucleic acid (amino acid sequence is sequence 3 23-129)
  • 3638-3691 is Linker
  • 3692-4051 is extra-membrane antigen binding region CD19 antibody heavy chain can be Variable region VH19 encoding nucleic acid (VH19 amino acid sequence is sequence 3, positions 148-267; Sequence 1, positions 3317-4051 are FMC63 single-chain antibody coding nucleic acid), and positions 4052-4171 are CD8 hinge region coding nucleic acid (amino acid sequence 3 268-307), 4172-4255 are the CD8 transmembrane region coding nucleic acid (amino acid sequence 3, 308-335), and 4256-
  • the lentiviral recombinant plasmid pLV-E19BBz (E19BBz) containing the EF1 ⁇ promoter (as shown in Figure 1a and 1c above) has the nucleotide sequence of sequence 2.
  • the recombinant plasmid is the MND promoter from the 2838-3236 of sequence 1 Replace with the promoter EF1 ⁇ (sequence 2 at positions 2838-4131 are the promoter EF1 ⁇ ), and the remaining nucleotide sequence remains unchanged, to obtain a fragment.
  • the above-mentioned recombinant vector is prepared according to the following method: separately clone the synthetic promoter sequence and the polynucleotide sequence of CAR, design primers to connect the two parts by overlapping PCR, and after the above sequence is successfully spliced, cloned into the pDBR vector (GenBank: FR822201.1) to construct pLV-E19BBz.
  • pDBR vector GenBank: FR822201.1
  • the promoter EF1 ⁇ of pLV-E19BBz (E19BBz) was replaced with MND and named pLV-M19BBz (M19BBz).
  • M19BBz pLV-M19BBz
  • the lentiviral recombinant plasmids pLV-E19BBz (E19BBz) and pLV-M19BBz (M19BBz) were packaged respectively to obtain the corresponding lentiviral particles. Specific steps are as follows:
  • each petri dish 500 ⁇ l buffer (purchased from Polyplus Transfection, product catalog number B161116), 6 ⁇ g of the lentiviral recombinant plasmid prepared in Example 1 pLV-E19BBz (E19BBz) or pLV-M19BBz (M19BBz) , 3 ⁇ g psPAX2 (purchased from Wuhan Miaoling Biotechnology Co., Ltd., product catalog number P026) and 1.5 ⁇ g pMD2.G (purchased from Guangzhou Jisai Biotechnology Co., Ltd., product catalog number 161220L08), mix well, and then add to the system Add packaging reagents (purchased from Polyplus Transfection, product catalog number 114-15), 25 ⁇ l/10 cm petri dish, mix again, and let stand at room temperature for 10 minutes to obtain a mixed solution.
  • 500 ⁇ l buffer purchased from Polyplus Transfection, product catalog number B161116)
  • the culture supernatant is collected as the virus stock solution, and the collected virus stock solution is filtered into a 50 ml centrifuge tube with a 0.45 ⁇ m filter, and centrifuged at a high speed of 18500 g at 4° C. for 2 hours. The supernatant is discarded, and DMEM complete medium (the volume ratio of the added medium to the virus stock solution is 1:500) is added to the pellet to resuspend the virus particles, which is the virus concentrate.
  • the virus titer was detected.
  • the titers of E19BBz virus and M19BBz virus were 2.6 ⁇ 10 8 /ml and 4.3 ⁇ 10 8 /ml, respectively.
  • the E19BBz virus stock solution and the M19BBz virus stock solution obtained by quantification (4) using the p24 ELISA kit (TAKARA, 632200) were detected.
  • the result is shown in Figure 2b. It can be seen that the amount of p24 in the M19BBz virus stock solution is greater than the E19BBz virus stock solution.
  • lymphocyte separation solution (Dongfang Huahui; 25710) into a new centrifuge tube, and then slowly add 2 times the volume (ie 30ml) of the blood sample diluted in step (1) to the upper layer of lymphocyte separation solution.
  • the centrifugation parameters are 2000rpm, 20min, rise 7 fall 4, 25°C;
  • the monocyte layer is transferred to a new centrifuge tube and washed with saline;
  • step (3) Purify CD3+ T cells with the kit for monocytes obtained in step (3) (Miltenyi Biotec: 130-050-101) to obtain CD3+ T cells.
  • E19BBz virus or M19BBz virus (MOI of 0.25, 0.5, 1, 2) prepared in step 1 (5) above to the culture flask with CD3+ T cells at room temperature. , 4, 8), then put the culture flask in a centrifuge, and centrifuge at 2000rpm, 2h, up 4 down 4, 35°C centrifugation parameters to complete the lentiviral transduction, and get CAR-T of M19BBz with different MOI Cells and CAR-T cells of E19BBz with different MOIs;
  • step (8) of the above one were tested by flow cytometry, and the transduction rate was tested by CD4-FITC (Biolegend; 357405) and 7AAD (Biolegend; 420403).
  • FIG. 3 is a representative graph with MOI value of 0.25. It can be seen that the T cell transduction rate of M19BBz is higher than that of E19BBz.
  • the cells on the fifth day (MOI value of 0.5) cultured in step (8) of the above one were tested by flow cytometry, and the biotinylated Erbitux (anti-EGFR monoclonal antibody, Merck: Erbitux) and SA-PE (Biolegend: 405204) or SA-APC (Biolegend: 405207) detects the expression of tEGFR, as well as the detection of specific antibodies with antigen CD19Fc-FITC (ACRO: P15391-1) and FMC63scFv (Bioswan: 019-01-647M) CAR molecule expression. Untransduced CD3+ T cells were used as controls.
  • MFI Fluorescence Intensity
  • tEGFR Detect the expression of tEGFR (erb).
  • M19BBz positive cell MFI/negative cell MFI
  • E19BBz E19BBz
  • FMC63scFv antibody scFv
  • scFv FMC63scFv antibody
  • the target cells were first stained with 1mg/ml calcein (Calcein-AM, Invitrogen, C3099), and then in 200 ⁇ l cell culture medium (RPMI 1640, Gibco, 22400-089+10% FBS ExCell Bio, FND500) target cells were added to 2 ⁇ 10 4, each CAR-T cells were added to each 2 ⁇ 10 5, after 37 °C for four hours, the supernatant was detected calcein released (detected using a microplate reader Multiskan Sky, excitation detection condition The wavelength is 490nm and the emission wavelength is 515nm.).
  • Spontaneous release group the target cells directly detect the supernatant after the same time, the value of this group is the spontaneous release of fluorescence;
  • Specific killing efficiency (%) (fluorescence value of experimental group-fluorescence value of spontaneous release group)/(fluorescence value of complete release group-fluorescence value of spontaneous release group) ⁇ 100%
  • NOD-SCID mice were used to establish a Raji-luciferase tumor model.
  • This model was constructed by infusion of Raji human B-cell lymphoma cells containing luc (luciferase) labeled Raji-luciferase (by Yikang (Beijing) Pharmaceutical Provided by Science and Technology Co., Ltd.), the tumor cells in the logarithmic growth phase were injected into the blood vessels of the mouse through the tail vein of the mouse, and the details are as follows:
  • the Raji-luciferase tumor model cells were prepared into a cell suspension with a concentration of 1 ⁇ 10 7 /ml with PBS, and inoculated into mice through the tail vein, 100 ⁇ l/mouse; each mouse was inoculated with 1 ⁇ 10 6 cells.
  • the mice will be injected with luciferin for three days. After anesthesia, the fluorescence intensity will be detected on the equipment (IVIS Lumina, Series III, PE). After the detection, mice with the same fluorescence intensity will be screened and then randomly divided and administered (six mice in each group).
  • Control group Inject 200 ⁇ l of physiological saline containing 2% (mass volume percentage content, unit is g/ml) human albumin (Hebei Daan Pharmaceutical Co., Ltd., S200443042);
  • M19BBz group (CAR-T M19BBz): Inject 200 ⁇ l of a solution containing 5 ⁇ 10 6 M19BBz CAR-T cells (MOI value is 0.25) (convert the total cell number according to the transfection rate, and resuspend the cells in 2% Human albumin in normal saline);
  • E19BBz group (CAR-T E19BBz): Inject 200 ⁇ l of a solution containing 5 ⁇ 10 6 E19BBz CAR-T cells (MOI value is 0.25) (convert the total number of cells according to the transfection rate, and resuspend the cells in a 2% Human albumin in normal saline);
  • the leukocytes were prepared according to the method of 1 and 3 in Example 1, and prepared multiple batches of M19BBz CAR-T cells (a certain batch of MOI value is 1) and E19BBz CAR-T cells (a certain batch of MOI value 1) (each is a different batch of cells), CAR-T cells were subjected to flow cytometry to detect the transduction rate (by biotinylated Erbitux (anti-EGFR monoclonal antibody, Merck: Love) before reinfusion on the fourteenth day). Biduo) and SA-PE (Biolegend: 405204) or SA-APC (Biolegend: 405207) to detect the expression of tEGFR), and the MFI value. Patients undergo reinfusion and stay in the hospital for observation until they are discharged.
  • the number of chimeric antigen receptors on the surface of CAR-T cells can be changed, the safety of the product can be further adjusted, cytokine storms can be reduced, and safer products can be provided.
  • the experiment of the present invention proves that, in order to overcome the shortcomings of low transfection efficiency and large amount of cytokine produced in the current CAR-T treatment process, the present invention can significantly increase the transfection rate, and the process development of clinical-grade cell production is easier to carry out.
  • Reduce production costs reduce the density of CAR molecules on the cell surface and reduce the intensity of the reaction, thereby reducing the occurrence of CRS and making CAR-T treatment safer.
  • the specifics are as follows: by improving the initiation of the CAR structure by the promoter, that is, using the MND promoter to promote the CAR structure, a new chimeric antigen receptor is prepared, and T cells expressing the chimeric antigen receptor are prepared.
  • the T cells have an impact
  • the CAR molecule has the characteristic of less expression on the cell surface.
  • it has the advantage of lessening the cell killing effect. This can greatly reduce the occurrence of cytokine storms and effectively improve the safety.
  • the preparation method obtains Chimeric antigen receptor T cells also have high transduction efficiency, and can be stably and persistently expressed on the surface of T cells, so that when they bind to the antigen on the surface of the target cell, they can play a killing effect on the target cell, and in addition, the expression can be sustained. It can also make its killing effect lasting; make it safer clinically for the treatment of cancer.

Landscapes

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

Abstract

提供了能高效制备且应用安全的嵌合抗原受体T细胞及其制备方法与应用,还涉及一种慢病毒重组载体,其含有CAR结构编码序列和驱动所述CAR结构编码序列表达的启动子MND;所述启动子MND的核苷酸序列为序列1第2838-3236位。为了克服当前CAR-T治疗过程中的转染效率低和产生细胞因子量大的缺点,通过改善启动子对CAR结构的启动,即用MND启动子启动CAR结构,制备出新的嵌合抗原受体,并制备表达该嵌合抗原受体的T细胞,该T细胞具有影响CAR分子在细胞表面表达较少的特性,与CD19阳性细胞结合时,有着使细胞杀伤作用较缓和的优点,如此可大大降低细胞因子风暴的发生,有效提高了安全性。

Description

一种能高效制备且应用安全的嵌合抗原受体T细胞及其制备方法与应用 技术领域
本发明涉及生物技术领域,具体涉及一种能高效制备且应用安全的嵌合抗原受体T细胞及其制备方法与应用。
背景技术
CAR-T疗法就是嵌合抗原受体T细胞免疫疗法,英文全称Chimeric Antigen Receptor T-Cell Immunotherapy。这是一种治疗肿瘤的新型精准靶向疗法,近几年通过优化改良在临床肿瘤治疗上取得很好的效果,是一种非常有前景的,能够精准、快速、高效,且有可能治愈癌症的新型肿瘤免疫治疗方法。
尽管CAR-T在临床效果上取得了巨大的成功,其商业化推广上仍有不小的问题:其一,价格昂贵,诺华的KymriahTM上市定价高达47.5万美元,支付方式可以创新,生产制备成本却因无法规模化而居高不下。影响生产成本的因素有很多,其中转染率低是很重要的一个原因,并且在通用性CAR-T中依然会面临转染效率低的问题,提高转染效率可以有效促进临床级别细胞生产的工艺开发,显著降低制备成本;其二,一些施用CAR-T细胞疗法的患者会发生危险且有时常常是危及生命的副作用,该副作用称为细胞因子释放综合征(CRS)或细胞因子风暴,其中输注的活化T细胞产生的细胞因子快速且大量释放到血流中的全身炎症反应,从而导致危险的低血压,高烧和寒战。在严重的CRS情况下,患者会发生生命危险。
针对CAR-T转染效率低制备成本高问题,目前常规解决办法是为采用合适的转染试剂、保持最佳的细胞状态或选择最佳的转染方法;但是这些改变转染效率还是低,制备成本还是高。
关于施用CAR-T细胞疗法的患者的细胞因子风暴副作用产生,目前研究发现,CAR-T细胞上CAR分子的数量会严重影响到对靶细胞的结合效率与活化效率;如果CAR分子表达效率低,可能导致CAR-T细胞不能有效活化,从而影响治疗效果;但是细胞表面的CAR分子密度过高,在治疗过程中反应强度激烈,会导致大量的细胞因子产生,提高细胞因子风暴(CRS)发生的风险。
因此,寻求一种可以高效低成本制备且在治疗同时可以降低细胞因子风暴的抗原受体T细胞是目前急需要解决的难题。
发明公开
本发明的一个目的是提供一种慢病毒重组载体。
本发明提供的慢病毒重组载体,其含有CAR结构编码序列和驱动所述CAR结构编码序列表达的启动子MND;
所述启动子MND的核苷酸序列为序列1第2838-3236位;
所述CAR结构依次包括胞外结合结构域、一个或多个铰链结构域或间隔结构域、跨膜结构域、一个或多个胞内共刺激信号转导结构域以及初级信号转导结构域。
上述慢病毒重组载体中,所述胞外结构域选自如下靶抗原的抗体或其抗原结合片段、束缚配体或者共受体的胞外结构域中至少一种:α叶酸受体、5T4、αvβ6整合素、BCMA、B7-H3、B7-H6、CAIX、CD19、CD20、CD22、CD30、CD33、CD44、CD44v6、CD44v7/8、CD70、CD79a、CD79b、CD123、CD138、CD171、CEA、CSPG4、EGFR、包括ErbB2(HER2)的EGFR家族、EGFRvIII、EGP2、EGP40、EPCAM、EphA2、EpCAM、FAP、胎儿型AchR、FRα、GD2、GD3、磷脂酰肌醇聚糖-3(GPC3)、HLA-A1+MAGE1、HLA-A2+MAGE1、HLA-A3+MAGE1、HLA-A1+NY-ESO-1、HLA-A2+NY-ESO-1、HLA-A3+NY-ESO-1、IL-11Rα、IL-13Rα2、λ、Lewis-Y、κ、间皮素、Muc1、Muc16、NCAM、NKG2D配体、NY-ESO-1、PRAME、PSCA、PSMA、ROR1、SSX、生存素、TAG72、TEM和VEGFR2;但不限于此;
所述铰链结构域或间隔结构域选自CD8α、CD4、CD45、PD1和CD152的跨膜结构域中的至少一种;但不限于此;
所述跨膜结构域选自CD8、CD4、CD45、PD1和CD152的跨膜结构域中的至少一种;但不限于此;
所述胞内共刺激信号转导结构域选自CD28、CD54(ICAM)、CD134(OX40)、CD137(41BB)、CD152(CTLA4)、CD273(PD-L2)、CD274(PD-L1)和CD278(ICOS)中至少一种;但不限于此;
所述初级信号转导结构域选自CD3ζ的初级信号转导结构域和FcRγ 的初级信号转导结构域中至少一种;但不限于此。
上述慢病毒重组载体中,所述胞外结合结构域(又名抗原特异性结合结构域)包括CD19抗体的轻链可变区和CD19抗体的重链可变区;
所述铰链结构域为CD8铰链结构域;
所述跨膜结构域为CD8跨膜结构域;
所述胞内共刺激信号转导结构域为4-1BB;
所述初级信号转导结构域为CD3ζ。
上述慢病毒重组载体中,
所述CD19抗体的轻链可变区的氨基酸序列为序列表中序列3第23-129位;
所述CD19抗体的重链可变区的氨基酸序列为序列表中序列3第148-267位;
所述CD8铰链结构域的氨基酸序列为序列表中序列3第268-307位;
所述CD8跨膜结构域的氨基酸序列为序列表中序列3第308-335位;
所述胞内共刺激信号转导结构域4-1BB的氨基酸序列为序列表中序列3第336-377位;
所述初级信号转导结构域CD3ζ的氨基酸序列为序列表中序列3第378-489位。
上述慢病毒重组载体中,所述CAR结构编码序列的核苷酸序列为序列1第3251-4717位。
在本发明的实施例中,所述慢病毒重组载体为将序列1第2838-5863位的DNA分子同源重组到慢病毒表达载体中,得到表达所述CAR结构的载体;在本发明的实施例中,具体采用的慢病毒表达载体为pDBR载体。本发明实施例中的慢病毒重组载体为慢病毒重组质粒pLV-M19BBz(M19BBz),其核苷酸序列为序列1。
由上述慢病毒重组载体包装得到的重组病毒也是本发明保护的范围。
上述重组病毒为能够表达序列1第3251-4717位所示的CAR结构,且能够侵染免疫效应细胞的病毒。所述病毒为慢病毒、疱疹病毒、巨噬细胞病毒、EB病毒、乙型肝炎病毒、丙型肝炎病毒或艾滋病毒。
含有上述的慢病毒重组载体的重组细胞也是本发明保护的范围。
上述重组细胞为表达上述的慢病毒重组载体的免疫效应细胞;
所述免疫效应细胞为细胞毒性T淋巴细胞、NKT细胞、NK细胞、辅助性T细胞或gamma delta T细胞。
启动子MND在CAR-T细胞中用作驱动CAR结构表达也是本保护的范围;
所述启动子MND的核苷酸序列为序列1第2838-3236位。
本发明另一个目的是提供一种制备CAR-T细胞的方法。
本发明提供的方法,包括如下步骤:将由上述慢病毒重组载体包装得到的重组病毒转染T细胞得到的重组细胞。
本发明还提供了如下一种治疗肿瘤或具有治疗肿瘤且低细胞因子风暴功能的产品,其包括上述慢病毒重组载体或上述重组细胞。
本发明还提供了如下应用:
本发明提供了上述的慢病毒重组载体、上述重组病毒或上述重组细胞在制备治疗肿瘤的产品中的应用;
或,本发明提供了上述的慢病毒重组载体、上述重组病毒或上述重组细胞在制备具有治疗肿瘤且低细胞因子风暴功能的产品中的应用。
本发明还提供了一种降低CAR-T细胞治疗中细胞因子风暴产生量的方法,为用启动子MND驱动CAR结构在T细胞中表达;
所述启动子MND的核苷酸序列为序列1第2838-3236位。
上述包括如下步骤:1)制备第一个目的中的慢病毒重组载体;
2)将所述慢病毒重组载体导入自体T细胞中,得到CAR-T细胞;
3)将所述CAR-T细胞回输体内,实现降低CAR-T细胞治疗中细胞因子风暴产生量。
本发明还提供了一种治疗肿瘤的方法,包括如下步骤:
1)制备第一个目的中的慢病毒重组载体;
2)将所述慢病毒重组载体导入自体T细胞中,得到CAR-T细胞;
3)将所述CAR-T细胞回输体内,实现治疗肿瘤。
在不同的实施方案中,本发明提供基因工程化的免疫效应细胞,其包含设计以表达使细胞毒性定向至肿瘤细胞的嵌合抗原受体的载体。这些基因工程化的受体在本文中称作CAR,CAR是这样的分子:将基于对靶抗 原(例如肿瘤抗原)特异性的抗体与T细胞受体活化胞内结构域组合,以产生显示出特异性的抗肿瘤细胞免疫活性的嵌合蛋白。
本文使用的术语“嵌合的”描述了由不同蛋白的部分组成或者由来自不同来源的DNA组成。
本文考虑的载体包含如MND、EF1α、CMV、PGK等启动子和编码CAR的多核苷酸(包括集落刺激因子信号肽、FMC63单链抗体序列、CD8的绞链区与跨膜区序列、4-1BB共刺激信号域以及CD3ζ信号域以及T2A和tEGFR)。本文考虑的CAR包含与特异性的靶抗原结合的胞外结构域(也被称为结合结构域或者抗原特异性结合结构域)、跨膜结构域和胞内信号转导结构域。CAR的抗原结合结构域与其在靶细胞表面上的靶抗原的结合导致CAR的聚集,并向包含CAR的细胞递送活化刺激。CAR的主要特性是其使免疫效应细胞的特异性重定向的能力,从而引发增殖、细胞因子的产生、吞噬作用或者能够以不依赖主要组织相容性复合物(MHC)的方式介导表达靶抗原的细胞死亡的分子的产生,利用单克隆抗体、可溶性配体或细胞特异性共受体的细胞特异性靶向能力。
附图说明
图1为不同启动子EF1α和MND的CAR载体构建,(a)使用EF1α启动子的CAR载体图谱;(b)使用MND启动子的CAR载体谱图,(c)为不同启动子EF1α和MND的CAR载体的结构示意图。
图2为转染效率的测定。
图3为转染效率的测定流式图。
图4为转导E19BBz、M19BBz后的T细胞中CAR分子的表达差异。
图5为利用不同启动子表达CAR的T细胞进行的抗原特异性肿瘤清除以及相对应的实验中CAR-T细胞产生细胞因子的量。
图6为不同启动子下CAR修饰的T细胞在过继性转移之后,在肿瘤模型小鼠中对肿瘤细胞的消退作用。
图7为临床试验中两种CAR-T细胞的转染效率,Relative MFI,T细胞体外培养扩增倍率以及CAR-T体内扩增倍率。
图8为临床试验中患者治疗后出现反应的总结,(a)患者回输后14天内的体温变化;(b)患者治疗后发烧温度的最高值;(c)患者治疗后 CRS的级数统计。
图9为采用两种启动子生产的CAR-T细胞,患者治疗后的长期临床结果。
实施发明的最佳方式
下述实施例中所使用的实验方法如无特殊说明,均为常规方法。
下述实施例中所用的材料、试剂等,如无特殊说明,均可从商业途径得到。
下述实施例中的定量试验,均设置三次重复实验,结果取平均值。
实施例1、慢病毒重组质粒的构建
1、含有MND启动子的慢病毒重组质粒pLV-M19BBz(M19BBz)
含有MND启动子的慢病毒重组质粒pLV-M19BBz(M19BBz)(如图1b和1c下图),其核苷酸序列为序列1(全基因合成),该重组质粒也可以将序列1第2838-5863位的DNA分子同源重组到pDBR载体(Calderón-Gómez,E.,et al.,Reprogrammed quiescent B cells provide an effective cellular therapy against chronic experimental autoimmune encephalomyelitis.European Journal of Immunology,2011.41(6):p.1696-1708.)中得到。
其中,序列1第2838-3236位为MND启动子,第3251-3316位为集落刺激因子信号肽编码核酸(氨基酸序列为序列3第1-22位)、第3317-3637位为膜外抗原结合区CD19抗体的轻链可变区VL19编码核酸(氨基酸序列为序列3第23-129位)、第3638-3691位为Linker、第3692-4051位为膜外抗原结合区CD19抗体的重链可变区VH19编码核酸(VH19氨基酸序列为序列3第148-267位;序列1第3317-4051位为FMC63单链抗体编码核酸),第4052-4171位为CD8铰链区编码核酸(氨基酸序列序列3第268-307位)、第4172-4255位为CD8跨膜区编码核酸(氨基酸序列序列3第308-335位),第4256-4381位为胞内共刺激信号转导结构域4-1BB编码核酸(氨基酸序列序列3第336-377位),第4382-4717位为初级信号转导结构域CD3ζ编码核酸(氨基酸序列序列3第378-489位);第4718-4789位为自切割序列T2A编码核酸(编码氨基酸序列3第490-513位),第4790-5863位为tEGFR(编码核酸氨基酸序列3第514-870),慢病毒重组质粒pLV-M19BBz(M19BBz)表达CAR结构(序列3第1-489 位),所述CAR结构的核苷酸序列为序列1第3251-4717位。
2、含有EF1α启动子的慢病毒重组质粒pLV-E19BBz(E19BBz)
含有EF1α启动子的慢病毒重组质粒pLV-E19BBz(E19BBz)(如图1a和1c上图),其核苷酸序列为序列2,该重组质粒为将序列1第2838-3236位为MND启动子替换为启动子EF1α(序列2第2838-4131位为启动子EF1α),且其余核苷酸序列不变,得到的片段。
上述重组载体按照如下方法制备:分别克隆合成启动子序列和CAR的多核苷酸序列,设计引物通过重叠PCR将其两部分进行连接,将上述序列拼接成功后,克隆到pDBR的载体上(GenBank:FR822201.1),构建出pLV-E19BBz。通过序列合成与克隆的方式,将pLV-E19BBz(E19BBz)的启动子EF1α更换为MND,命名为pLV-M19BBz(M19BBz)。载体各部分元件如图1所示。
实施例2、CAR-T细胞的制备
一、CAR-T细胞的制备
1、慢病毒重组质粒的包装
分别将慢病毒重组质粒pLV-E19BBz(E19BBz)和pLV-M19BBz(M19BBz)进行包装,分别得到对应的慢病毒颗粒。具体步骤如下:
(1)将已长到80%-90%的293FT细胞(赛业(广州)HEKFT-30001)培养瓶(T175)从37℃、5%CO 2的细胞培养箱中取出,消化后收集洗涤细胞,每10cm细胞培养皿中加入4.5×10 6个细胞和9ml DMEM完全培养基(购于Gibco公司,产品目录号11965-084),轻轻摇匀,放入37℃、5%CO 2培养箱中培养。第2天,每个培养皿加入如下试剂:500μl buffer(购于Polyplus Transfection公司,产品目录号B161116)、6μg实施例1制备的慢病毒重组质粒pLV-E19BBz(E19BBz)或pLV-M19BBz(M19BBz)、3μg psPAX2(购于武汉淼灵生物科技有限公司,产品目录号P026)和1.5μg pMD2.G(购于广州吉赛生物科技股份有限公司,产品目录号161220L08),混合均匀,然后再向体系中加入包装试剂(购于Polyplus Transfection公司,产品目录号114-15),25μl/10cm培养皿,再次混合均匀,室温静置10min,得到混合液。
(3)将用于包装病毒的293FT细胞从37℃、5%CO 2的细胞培养箱中取 出,将混合液平均加到每个培养皿中,轻轻摇匀,放入37℃、5%CO 2培养箱中继续培养。培养4h后,弃旧培养基,加入5ml已预热的PBS清洗细胞,再加入9ml新鲜的已预热的含10%(体积分数)FBS的DMEM完全培养基,放入37℃、5%CO 2培养箱中培养。
(4)继续培养48-72h后收取培养上清作为病毒原液,并将收集的病毒原液用0.45μm过滤器过滤到50ml离心管中,4℃、18500g高速离心2h。弃上清液,向沉淀中加入DMEM完全培养基(加入的培养基与病毒原液的体积比为1:500)重悬病毒颗粒,此即为病毒浓缩液。
(5)将病毒浓缩液按200μl/管分装,另外留取10μl进行病毒滴度测定。将分装好的浓缩液置于-80℃冰箱保存,得到E19BBz病毒和M19BBz病毒。
检测病毒滴度,E19BBz病毒和M19BBz病毒的滴度分别为2.6×10 8/ml与4.3×10 8/ml。
检测利用p24的ELISA试剂盒(TAKARA,632200)定量(4)得到的E19BBz病毒原液和M19BBz病毒原液,结果如图2b所示,可以看出,M19BBz病毒原液的p24量大于E19BBz病毒原液。
2、T细胞的分选
(1)将健康供者新鲜外周血(供体者知情)样本转移至离心管中,用等体积的0.9%生理盐水稀释并混合均匀;
(2)将15ml淋巴细胞分离液(东方华辉;25710)置入新的离心管中,随后将2倍体积(即30ml)步骤(1)中稀释的血样缓慢加到淋巴细胞分离液上层,进行离心,离心参数为2000rpm,20min,升7降4,25℃;
(3)离心结束后,会分为四层:稀释的血浆层、单核细胞层、淋巴细胞分离液层以及红细胞层,将单核细胞层转移至新的离心管中,并用生理盐水洗涤;
(4)将步骤(3)中得到的单核细胞用试剂盒进行CD3+T细胞的纯化(美天旎:130-050-101),得到CD3+T细胞。
3、慢病毒颗粒转染靶细胞制备嵌合抗原受体T细胞
(5)取纯化后1×10 7的CD3+T细胞铺培养瓶,同时加入25μl的CTS CD3/CD28Dynabeads(Gibco:40203D)刺激活化培养;
(6)培养的第二天,在室温下向有CD3+T细胞的培养瓶中加入一定 量的上述1步骤(5)制备的E19BBz病毒或M19BBz病毒(MOI分别为0.25、0.5、1、2、4、8),随后将培养瓶放至在离心机中,在2000rpm,2h,升4降4,35℃离心参数下进行离心以完成慢病毒转导,得到不同MOI的M19BBz的CAR-T细胞和不同MOI的E19BBz的CAR-T细胞;
(7)培养的第三天对培养瓶中的细胞进行半量补液(加入含200IU IL-2的TexMACS(美天旎:130-097-748与170-076-309));
(8)培养的第五天,采用流式细胞仪检测嵌合抗原受体在T细胞中的表达情况;
(9)继续扩大培养,在培养的第十二天时,采用流式细胞术进行转导率检测;
(10)继续培养至14天,收集细胞,完成嵌合抗原受体T细胞制备。
二、CAR-T细胞的鉴定检测
1、转导率检测
将上述一中的(8)步骤培养的第五天的细胞进行流式细胞术检测,其中,通过CD4-FITC(Biolegend;357405)、7AAD(Biolegend;420403)检测转导率。
不同MOI值的转导率结果如图2a所示,可以看出,M19BBz的T细胞转导率较E19BBz高。
不同MOI值的阳性CAR+的细胞百分比(代表转导率)结果如图2c所示,可以看出,等量的T细胞分别转导不同MOI的E19BBz病毒与M19BBz病毒,检测转导率后发现E19BBz转导率无法像M19BBz一样随着MOI的提高,而有相应的转导率增加。表明,M19BBz的T细胞转导率较E19BBz高。
图3为MOI值为0.25的代表图,可以看出M19BBz的T细胞转导率较E19BBz高。
2、检测CAR分子表达
将上述一中的(8)步骤培养的第五天的细胞(MOI值为0.5)进行流式细胞术检测,通过生物素化的Erbitux(anti-EGFR单抗,默克:爱必妥)与SA-PE(Biolegend:405204)或SA-APC(Biolegend:405207)检测tEGFR表达,以及用抗原CD19Fc-FITC(ACRO:P15391-1)和FMC63scFv(Bioswan: 019-01-647M)的特异性抗体检测CAR分子表达。以未转导的CD3+T细胞作为对照。
结果如图4a所示,可以看出,使用不同的启动子均可以使CAR正常表达;tEGFR分子的确与CAR分子共表达,且表达效率成正相关。
进一步分析流式结果的平均萤光强度(Mean Fluorescence Intensity,MFI)(流式分析软件可以直接产生MFI数据)。
结果如图4b所示:
检测tEGFR的表达(erb),表达M19BBz的CAR-T细胞表面上tEGFR的相对MFI值(阳性细胞MFI/阴性细胞MFI)(12.19±1.30)较表达E19BBz的CAR-T细胞(10.47±1.16)略高,但两者的差异约为16%。通过检测tEGFR,显示二者之间的表达无明显差异。
在检测CAR分子的表达上,用CD19Fc检测时,表达E19BBz的CAR-T细胞与表达M19BBz的CAR-T细胞的相对MFI(4.37±0.59vs 1.13±0.08),两者的差异是-74%。
用FMC63scFv抗体(scFv)检测时,E19BBz与M19BBz的相对MFI(2.26±0.37vs 1.24±0.07),两者的差异是-46%;两种抗体检测都显示了,CAR分子在E19BBz表面表达量较高。
实施例3、嵌合抗原受体T细胞的体外功能性实验测定
1、杀伤和细胞因子的检测
A.特异性杀伤效率检测
将实施例2的一的3制备的M19BBz的CAR-T细胞(MOI值为0.25)和E19BBz的CAR-T细胞(MOI值为0.25)分别各自与三种不同的肿瘤靶细胞(NALM-6、Raji、697:依序为广州赛库生物:B系急性淋巴细胞白血病CC1928;ATCC:Burkitt’s淋巴瘤细胞CCL86;上海宾穗:人前B细胞白血病细胞系BSC-5209479641-01)共培养,具体如下:
实验组:靶细胞先用1mg/ml的钙黄绿素(Calcein-AM,Invitrogen,C3099)染色后,在200μl的细胞培养基(RPMI 1640,Gibco,22400-089+10%FBS ExCell Bio,FND500)中,靶细胞加入2×10 4,每种CAR-T细胞各加入2×10 5,37℃培养四小时后,检测上清液中释放的钙黄绿素(用酶标仪Multiskan Sky检测,检测条件激发波长490nm,发射波长515nm。)。
自发释放组:靶细胞在相同时间后直接检测上清,该组数值为自发释放萤光;
完全释放组:靶细胞加入2%Triton X-100每孔100μl,在相同时间后直接检测上清,该数值为完全释放组。
特异性杀伤效率(%)=(实验组荧光值-自发释放组荧光值)/(完全释放组荧光值-自发释放组荧光值)×100%
结果如图5a所示,可以看出在NAML-6与697两种细胞系中,E19BBz的CAR-T细胞与M19BBz的CAR-T细胞的杀伤能力没有显著差异,而在Raji细胞系上,M19BBz的CAR-T细胞较E19BBz的CAR-T细胞为高。结合实施例2的结果,显示M19BBz的CAR-T细胞表面的CAR分子数虽然减少了,但是对于肿瘤细胞的杀伤能力,并未随之降低。
B.细胞因子风暴检测
在200μl的细胞培养基(RPMI 1640,Gibco,22400-089+10%FBS ExCell Bio,FND500)中,靶细胞加入2×10 4,CAR-T细胞加入6×10 4,37℃培养20小时后,取上清液用多因子检测试剂盒(BioLegend LEGENDplex#740118)进行细胞因子检测。
结果如图5b和5c所示,发现M19BBz的CAR-T细胞中的细胞因子TNF-α和IFN-γ的表达量比E19BBz的CAR-T细胞显著降低,结合图5a的结果,说明M19BBz的CAR-T细胞能在不降低肿瘤杀伤能力的情况下,相比E19BBz的CAR-T细胞更加的安全,有效降低细胞因子风暴。
2、动物实验检测体内杀瘤效果
采用NOD-SCID小鼠来建立Raji-luciferase的肿瘤模型,此模型的构建是通过输注含有luc(荧光素酶)标记的Raji人B细胞淋巴瘤细胞Raji-luciferase(由亦康(北京)医药科技有限公司提供),将处于对数生长期的肿瘤细胞通过小鼠尾静脉注射进到小鼠血管内构建,具体如下:
用PBS将Raji-luciferase的肿瘤模型细胞制备成浓度为1×10 7/ml的细胞悬液,通过尾静脉接种至小鼠体内,100μl/鼠;每只小鼠接种1×10 6细胞,第三天老鼠会注射luciferin,麻醉后在设备(IVIS Lumina,Series III,PE)上进行萤光强度检测,检测后筛选荧光强度一致的小鼠再进行随机分组并给药(每组六只)。
对照组(no treatment):注射200μl含2%(质量体积百分含量,单位为g/ml)人类白蛋白(河北大安制药有限公司,国药准字S200443042)的生理盐水;
M19BBz组(CAR-T M19BBz):注射200μl含有5×10 6个M19BBz的CAR-T细胞(MOI值为0.25)的溶液(根据转染率换算总细胞数,并将细胞重悬于含2%人类白蛋白的生理盐水中);
E19BBz组(CAR-T E19BBz):注射200μl含有5×10 6个E19BBz的CAR-T细胞(MOI值为0.25)的溶液(根据转染率换算总细胞数,并将细胞重悬于含2%人类白蛋白的生理盐水中);
每隔七天量测一次发光信号,并观察小鼠存活情况。
测定分析的结果如图6所示,(a):使用体内成像系统,检测对比未治疗组、E19BBz和M19BBz治疗之后肿瘤负荷;(b)(纵坐标是平均光强度代表肿瘤负荷量):对a图进行量化处理,并进行统计学分析;M19BBz的CAR-T细胞的小鼠体内杀瘤效果明显优于E19BBz的CAR-T细胞,并且具有统计学上的差异。
3、临床水平上的实验测定
通过临床实验结果,来统计产品质量与安全性。难治复发的急性B细胞白血病患者,经过医师筛选后由患者代表签署知情同意,入组进行临床试验,在医院内采集白细胞后进行细胞生产。
表1入组详情
Figure PCTCN2020133024-appb-000001
Figure PCTCN2020133024-appb-000002
白细胞按照实施例1的一的3的方法,分别制备了多批次的M19BBz的CAR-T细胞(某一批次MOI值为1)和E19BBz的CAR-T细胞(某一批次MOI值为1)(各为不同批次细胞),在第十四天回输前CAR-T细胞进行流式细胞术检测转导率(通过生物素化的Erbitux(anti-EGFR单抗,默克:爱必妥)与SA-PE(Biolegend:405204)或SA-APC(Biolegend:405207)检测tEGFR表达),以及MFI数值。患者在医院进行回输后留院观察直到出院,住院时每日监测体温变化并观察反应,并根据对应状况进行对症治疗,在固定时间检测外周血,计算CAR-T细胞扩增情况。治疗后14-28日进行治疗效果的早期评估。
结果如图7a和7b所示,可以看出,E19BBz的转导率平均较M19BBz低。而tEGFR相对萤光强度,在这两种结构中都没有显著差异。说明在进行临床细胞制备时,两种病毒的转导率与tEGFR的表达情况和先前体外功能结果相符。
体外培养时计算收获细胞数量后,统计培养扩增倍数(图7c),以及细胞在体内扩增后最大数值与起始回输剂量的倍数(图7d),都显示E19BBz和M19BBz没有显著统计差异。
但是在观察两组受试者每日的体温变化情况,M19BBz的平均体温上升较缓(图8a),发烧温度的最高数值也较低(图8b),发生2级以上CRS的比例也较低(图8c)。对患者进行长期随访后,M19BBz的总体生存期与E19BBz并没有显著差异(图9),代表副反应较和缓的M19BBzCAR-T细胞在治疗效果上与E19BBz相似。
总结而言,通过更换启动子,可以改变CAR-T细胞表面的嵌合抗原受体数量,进一步调整产品的安全性,降低细胞因子风暴,提供更安全的产品。
工业应用
本发明的实验证明,本发明为了克服当前CAR-T治疗过程中的转染效率低和产生细胞因子量大的缺点,实现显著提升转染率,临床级别细胞生产的工艺开发较容易进行,能够降低制备成本;通过降低细胞表面的CAR分子密度,降低反应的强度,从而降低CRS的发生,使CAR-T治疗更加安全。具体如下:通过改善启动子对CAR结构的启动,即用MND启动子启动CAR结构,制备出新的嵌合抗原受体,并制备表达该嵌合抗原受体的T细胞,该T细胞具有影响CAR分子在细胞表面表达较少的特性,与CD19阳性细胞结合时,有着使细胞杀伤作用较缓和的优点,如此可大大降低细胞因子风暴的发生,有效提高了安全性,同时该制备方法得到的嵌合抗原受体T细胞还具有较高的转导效率,并且能在T细胞表面稳定、持久的表达,如此在与靶细胞表面的抗原结合时,可发挥杀伤靶细胞效应,另外持久的表达还能使其杀伤效果持久;使其对用于治疗癌症的临床上更为安全。

Claims (18)

  1. 慢病毒重组载体,其含有CAR结构和驱动所述CAR结构表达的启动子MND;
    所述启动子MND的核苷酸序列为序列1第2838-3236位;
    所述CAR结构依次包括胞外结合结构域、一个或多个铰链结构域或间隔结构域、跨膜结构域、一个或多个胞内共刺激信号转导结构域以及初级信号转导结构域。
  2. 根据权利要求1所述的慢病毒重组载体,其特征在于:
    所述胞外结合结构域选自如下靶抗原的抗体或其抗原结合片段、束缚配体或者共受体的胞外结构域中至少一种:α叶酸受体、5T4、αvβ6整合素、BCMA、B7-H3、B7-H6、CAIX、CD19、CD20、CD22、CD30、CD33、CD44、CD44v6、CD44v7/8、CD70、CD79a、CD79b、CD123、CD138、CD171、CEA、CSPG4、EGFR、包括ErbB2的EGFR家族、EGFRvIII、EGP2、EGP40、EPCAM、EphA2、EpCAM、FAP、胎儿型AchR、FRα、GD2、GD3、磷脂酰肌醇聚糖-3、HLA-A1+MAGE1、HLA-A2+MAGE1、HLA-A3+MAGE1、HLA-A1+NY-ESO-1、HLA-A2+NY-ESO-1、HLA-A3+NY-ESO-1、IL-11Rα、IL-13Rα2、λ、Lewis-Y、κ、间皮素、Muc1、Muc16、NCAM、NKG2D配体、NY-ESO-1、PRAME、PSCA、PSMA、ROR1、SSX、生存素、TAG72、TEM和VEGFR2;
    所述铰链结构域或间隔结构域选自CD8α、CD4、CD45、PD1和CD152的跨膜结构域中的至少一种;
    所述跨膜结构域选自CD8、CD4、CD45、PD1和CD152的跨膜结构域中的至少一种;
    所述胞内共刺激信号转导结构域选自CD28、CD54(ICAM)、CD134(OX40)、CD137(41BB)、CD152(CTLA4)、CD273(PD-L2)、CD274(PD-L1)和CD278(ICOS)中至少一种;
    所述初级信号转导结构域选自CD3ζ的初级信号转导结构域和FcRγ的初级信号转导结构域中至少一种。
  3. 根据权利要求1或2所述的慢病毒重组载体,其特征在于:
    所述胞外结合结构域包括CD19抗体的轻链可变区和CD19抗体的重 链可变区;
    所述铰链结构域为CD8铰链结构域;
    所述跨膜结构域为CD8跨膜结构域;
    所述胞内共刺激信号转导结构域为4-1BB;
    所述初级信号转导结构域为CD3ζ。
  4. 根据权利要求3所述的慢病毒重组载体,其特征在于:
    所述CD19抗体的轻链可变区的氨基酸序列为序列表中序列3第23-129位;
    所述CD19抗体的重链可变区的氨基酸序列为序列表中序列3第148-267位;
    所述CD8铰链结构域的氨基酸序列为序列表中序列3第268-307位;
    所述CD8跨膜结构域的氨基酸序列为序列表中序列3第308-335位;
    所述胞内共刺激信号转导结构域4-1BB的氨基酸序列为序列表中序列3第336-377位;
    所述初级信号转导结构域CD3ζ的氨基酸序列为序列表中序列3第378-489位。
  5. 根据权利要求1-4中任一所述的慢病毒重组载体,其特征在于:
    所述CAR结构的核苷酸序列为序列1第3251-4717位。
  6. 根据权利要求5所述的慢病毒重组载体,其特征在于:
    所述慢病毒表达载体为pDBR载体;
    所述慢病毒重组载体的核苷酸序列为序列1。
  7. 由权利要求1-6中任一所述的慢病毒重组载体包装得到的重组病毒。
  8. 含有权利要求1-6中任一所述的慢病毒重组载体的重组细胞。
  9. 根据权利要求8所述的重组细胞,其特征在于:所述重组细胞为表达权利要求1-6中任一所述的慢病毒重组载体的免疫效应细胞。
  10. 根据权利要求9所述的重组细胞,其特征在于:所述免疫效应细胞为细胞毒性T淋巴细胞、NKT细胞、NK细胞、辅助性T细胞或gamma delta T细胞。
  11. 启动子MND在CAR-T细胞中用作驱动CAR结构表达的应用;
    所述启动子MND的核苷酸序列为序列1第2838-3236位。
  12. 一种制备CAR-T细胞的方法,包括如下步骤:将由权利要求1-6中任一所述的慢病毒重组载体包装得到的重组病毒转染T细胞得到的重组细胞。
  13. 一种治疗肿瘤或具有治疗肿瘤且低细胞因子风暴功能的产品,其包括权利要求1-6中任一所述的慢病毒重组载体或权利要求8-10中任一所述重组细胞。
  14. 权利要求1-6中任一所述的慢病毒重组载体、权利要求7所述重组病毒或权利要求8-10中任一所述重组细胞在制备治疗肿瘤的产品中的应用;
  15. 权利要求1-6中任一所述的慢病毒重组载体、权利要求7所述重组病毒或权利要求8-10中任一所述重组细胞在制备具有治疗肿瘤且低细胞因子风暴功能的产品中的应用。
  16. 一种降低CAR-T细胞治疗中细胞因子风暴产生量的方法,为用启动子MND驱动CAR结构在T细胞中表达;
    所述启动子MND的核苷酸序列为序列1第2838-3236位。
  17. 根据权利要求16所述的方法,其特征在于:所述方法包括如下步骤:1)制备权利要求1-6中任一所述的慢病毒重组载体;
    2)将所述慢病毒重组载体导入自体T细胞中,得到CAR-T细胞;
    3)将所述CAR-T细胞回输体内,实现降低CAR-T细胞治疗中细胞因子风暴产生量。
  18. 一种治疗肿瘤的方法,包括如下步骤:
    1)制备权利要求1-6中任一所述的慢病毒重组载体;
    2)将所述慢病毒重组载体导入自体T细胞中,得到CAR-T细胞;
    3)将所述CAR-T细胞回输体内,实现治疗肿瘤。
PCT/CN2020/133024 2019-12-02 2020-12-01 一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用 WO2021109977A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2020397271A AU2020397271B2 (en) 2019-12-02 2020-12-01 Chimeric antigen receptor T cell which can be efficiently prepared and safely used, preparation method therefor and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911212444.0 2019-12-02
CN201911212444.0A CN112980886B (zh) 2019-12-02 2019-12-02 一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用

Publications (1)

Publication Number Publication Date
WO2021109977A1 true WO2021109977A1 (zh) 2021-06-10

Family

ID=76221462

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/133024 WO2021109977A1 (zh) 2019-12-02 2020-12-01 一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用

Country Status (3)

Country Link
CN (1) CN112980886B (zh)
AU (1) AU2020397271B2 (zh)
WO (1) WO2021109977A1 (zh)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111315402A (zh) * 2017-08-15 2020-06-19 艾达普特免疫有限公司 T细胞修饰
US20210032658A1 (en) * 2014-04-25 2021-02-04 Bluebird Bio, Inc. Mnd promoter chimeric antigen receptors
US12006369B2 (en) 2014-07-24 2024-06-11 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US12029784B2 (en) 2014-12-12 2024-07-09 2Seventy Bio, Inc. BCMA chimeric antigen receptors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106536549A (zh) * 2014-04-25 2017-03-22 蓝鸟生物公司 Mnd启动子嵌合抗原受体
CN107207598A (zh) * 2014-12-12 2017-09-26 蓝鸟生物公司 Bcma嵌合抗原受体
CN109311984A (zh) * 2016-03-11 2019-02-05 蓝鸟生物公司 基因组编辑的免疫效应细胞
WO2019104245A1 (en) * 2017-11-22 2019-05-31 La Jolla Institute For Allergy And Immunology Use and production of engineered immune cells

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109415687A (zh) * 2016-04-07 2019-03-01 蓝鸟生物公司 嵌合抗原受体t细胞组合物
EP3595684A1 (en) * 2017-03-17 2020-01-22 Senti Biosciences, Inc. Immunomodulating cell circuits
SG10202111394XA (en) * 2017-04-13 2021-12-30 Senti Biosciences Inc Combinatorial cancer immunotherapy
JP2021506260A (ja) * 2017-12-15 2021-02-22 ジュノー セラピューティクス インコーポレイテッド 抗cct5結合分子およびその使用方法

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106536549A (zh) * 2014-04-25 2017-03-22 蓝鸟生物公司 Mnd启动子嵌合抗原受体
CN107207598A (zh) * 2014-12-12 2017-09-26 蓝鸟生物公司 Bcma嵌合抗原受体
CN109311984A (zh) * 2016-03-11 2019-02-05 蓝鸟生物公司 基因组编辑的免疫效应细胞
WO2019104245A1 (en) * 2017-11-22 2019-05-31 La Jolla Institute For Allergy And Immunology Use and production of engineered immune cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LIN LAN, XU ZHILIANG: "Comparision of the Efficiency of Different Promoters Driving the Expression of CD19-specifc Chimeric Antigen Receptor(CAR)-redirected T Lymphocytes Mediated by Lentiviral Vector", JOURNAL OF MEDICAL RESEARCH, vol. 46, no. 6, 1 June 2017 (2017-06-01), pages 92 - 98, XP055818806, DOI: 10.11969/j.issn.1673-548X.2017.06.024 *
YING ZHITAO; HUANG XUE F.; XIANG XIAOYU; LIU YANLING; KANG XI; SONG YUQIN; GUO XIAOKAI; LIU HANZHI; DING NING; ZHANG TINGTING; DUA: "A safe and potent anti-CD19 CAR T cell therapy", NATURE MEDICINE, NATURE PUB. CO, NEW YORK, vol. 25, no. 6, 22 April 2019 (2019-04-22), New York, pages 947 - 953, XP036901065, ISSN: 1078-8956, DOI: 10.1038/s41591-019-0421-7 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210032658A1 (en) * 2014-04-25 2021-02-04 Bluebird Bio, Inc. Mnd promoter chimeric antigen receptors
US12006369B2 (en) 2014-07-24 2024-06-11 2Seventy Bio, Inc. BCMA chimeric antigen receptors
US12029784B2 (en) 2014-12-12 2024-07-09 2Seventy Bio, Inc. BCMA chimeric antigen receptors
CN111315402A (zh) * 2017-08-15 2020-06-19 艾达普特免疫有限公司 T细胞修饰
CN111315402B (zh) * 2017-08-15 2024-05-07 艾达普特免疫有限公司 T细胞修饰

Also Published As

Publication number Publication date
CN112980886B (zh) 2022-02-22
AU2020397271A1 (en) 2022-07-21
AU2020397271B2 (en) 2023-08-03
CN112980886A (zh) 2021-06-18

Similar Documents

Publication Publication Date Title
WO2021109977A1 (zh) 一种能高效制备且应用安全的嵌合抗原受体t细胞及其制备方法与应用
US20210317183A1 (en) Enhanced antigen presenting ability of rna car t cells by co-introduction of costimulatory molecules
EP3490585B1 (en) Immunomodulatory polypeptides and related compositions and methods
Dargel et al. T cells engineered to express a T-cell receptor specific for glypican-3 to recognize and kill hepatoma cells in vitro and in mice
CA2864688C (en) Use of icos-based cars to enhance antitumor activity and car persistence
JP7399157B2 (ja) 改良された治療用t細胞
Perro et al. Generation of multi-functional antigen-specific human T-cells by lentiviral TCR gene transfer
WO2015179801A1 (en) Car based immunotherapy
JP2022507830A (ja) 治療薬を発現する改変細胞とその使用
AU2016341527A1 (en) Methods, kits, agents and apparatuses for transduction
WO2013044225A1 (en) A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens
CN109266667B (zh) 靶向cd5的嵌合抗原受体及其应用
JP2018510652A (ja) Her2/erbb2キメラ抗原受容体
CN113677353A (zh) 修饰细胞的扩增及其用途
EP4151660A1 (en) Fusion protein for reversing tumor microenvironment and use thereof
JP2017522859A (ja) グリピカン3に特異的なt細胞受容体、及び肝細胞癌の免疫療法のためのその使用
JP2021525101A (ja) 改変された抗cd19 car−t細胞
CN110257338B (zh) 嵌合细胞因子受体
TW201930342A (zh) 包含nkg2d結構域的多特異性嵌合受體及其使用方法
CN112423793A (zh) 肌肉特异性激酶嵌合自身抗体受体细胞的组合物和方法
WO2020019983A1 (zh) 一种用于治疗肿瘤的基因工程细胞
CN115820697B (zh) 一种免疫细胞及其制备方法和应用
WO2024011335A1 (zh) 经修饰的免疫细胞
CN118047853A (zh) 识别肿瘤相关抗原的tcr分子及其用途
CN118725080A (zh) 识别肿瘤相关抗原的tcr分子及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20895274

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020397271

Country of ref document: AU

Date of ref document: 20201201

Kind code of ref document: A

122 Ep: pct application non-entry in european phase

Ref document number: 20895274

Country of ref document: EP

Kind code of ref document: A1