WO2022007665A1 - Non-viral method for preparing stable nk cells with high chimeric receptor expression - Google Patents

Non-viral method for preparing stable nk cells with high chimeric receptor expression Download PDF

Info

Publication number
WO2022007665A1
WO2022007665A1 PCT/CN2021/103235 CN2021103235W WO2022007665A1 WO 2022007665 A1 WO2022007665 A1 WO 2022007665A1 CN 2021103235 W CN2021103235 W CN 2021103235W WO 2022007665 A1 WO2022007665 A1 WO 2022007665A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
natural killer
car
chimeric receptor
Prior art date
Application number
PCT/CN2021/103235
Other languages
French (fr)
Chinese (zh)
Inventor
肖�琳
裘新红
彭群武
张亮
朱苏闽
徐芳芳
Original Assignee
杭州优凯瑞医药科技有限公司
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 杭州优凯瑞医药科技有限公司 filed Critical 杭州优凯瑞医药科技有限公司
Publication of WO2022007665A1 publication Critical patent/WO2022007665A1/en

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464417Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • 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
    • A61K39/464429Molecules with a "CD" designation not provided for elsewhere
    • 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
    • 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
    • C07K14/70521CD28, CD152
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian 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
    • 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
    • C12N5/0646Natural killers cells [NK], NKT 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
    • 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
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/50Colon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/90Vectors containing a transposable element

Definitions

  • the present application belongs to the technical field of medical bioengineering, and relates to a method for preparing NK cells modified by stable and high expression of chimeric receptors by using a non-viral method and its application.
  • NK cells Natural killer cells are an important part of the innate immune system. The proportion of NK cells in peripheral blood lymphocytes is about 10%, and they are the third largest lymphocyte population after B cells and T cells. NK cells have multiple biological functions and are the body's first line of defense against infections and tumors. NK cell function is regulated by a sophisticated and complex network of activating and inhibitory receptors. Most circulating NK cells are in a quiescent phase and can be activated by cytokines and infiltrate pathogen-infected or malignant cells-containing tissues. When the receptors of NK cells are combined with the corresponding ligands, NK cells can also secrete some cytokines, such as interferon (IFN)- ⁇ , to play an immunoregulatory role.
  • IFN interferon
  • NK cells are involved in controlling the occurrence and development of various diseases.
  • Several in vitro studies on mammalian cells, including human cells, and in vivo studies in mice and rats have shown that NK cells can recognize tumor cells as targets and control tumor cell growth, metastasis, and spread in vivo. Decreased peripheral blood NK cell activity increases the risk of cancer in adults, an 11-year follow-up epidemiological survey showed.
  • NK cells As host immunity has also been studied in various cases of flavivirus infection such as Japanese encephalitis virus, yellow fever virus, dengue virus, tick-borne encephalitis virus and West Nile virus (WNV) ; their role in viral hepatitis, influenza virus and HIV-1 infection has also been well documented in several studies; likewise, their role in preventing respiratory tract infections by viruses such as bacteria, respiratory syncytial virus (RSV) and influenza
  • RSV respiratory syncytial virus
  • the role of Aspects has also been described in detail in mouse studies. (For details, please refer to: Mandal A, Viswanathan C. et al., Natural killer cells: In health and diseas[J]. Hematology/oncology & Stem Cell Therapy, 2015, 8(2): 47-55.).
  • NK cells do not require HLA matching, so they can be used as heterologous off-the-shelf cell drugs for immunotherapy of patients, which has important clinical application prospects.
  • Adoptive therapy based on autologous or allogeneic NK cells has been clinically applied to anti-tumor and anti-viral infection, especially allogeneic NK cells have shown good therapeutic effects in the treatment of hematological cancers (Reference: Lupo K B, Matosevic S. et al., Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy [J]. Cancers, 2019, 11(6).
  • NK cells have powerful immune effector functions
  • many cancer cells and viruses suppress the function of NK cells through mechanisms such as antigen escape and immunosuppression.
  • immunosuppression such as by expressing endogenous cytokines to enhance NK after adoptive reinfusion Cell proliferation ability, inhibiting immunosuppressive signals or enhancing the killing function of NK cells, etc.
  • CARs chimeric antigen receptors
  • a chimeric antigen receptor is a fusion of an antibody's scFv with an intracellular signal transduction structure, and the scFv is used to mediate the recognition and binding of tumor cell surface antigens.
  • primary NK cells are more resistant to gene transfection, which results in a low rate of uptake by NK cells to various vector systems, low transgene expression in NK cells, and only viral vectors achieve high levels in primary NK cells gene transfer efficiency.
  • the modification of chimeric receptors in primary NK cells is generally achieved by electrotransformation of mRNA encoding genes of chimeric receptors or by lentivirus/retrovirus infection. mRNA electrotransformation can only make the gene encoding the chimeric receptor transiently expressed, and the cost of synthesizing mRNA in vitro is relatively high, requiring multiple injections in clinical practice, and the treatment cost is high.
  • Lentiviral/retroviral transfection can produce NK cells with stable expression of chimeric receptor genes.
  • primary NK cells are extremely difficult to infect. So far, only a few scientific research teams have successfully applied NK cells.
  • Virus/retrovirus prepares chimeric antigen receptor-modified primary NK cells. After repeated infection, the expression ratio of its transgene in the final product is between 20% and 80%. Among them, retrovirus transfection The efficiency is higher than that of lentivirus (see literature: Sandro et al., Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies [J]. Journal of Immunology Research, 2018.).
  • retroviruses and/or lentiviruses are themselves pathogenic and have the potential for insertional mutagenesis, they pose a significant regulatory hurdle to the implementation of human clinical trials.
  • the large-scale production of viral vectors in cell lines has the problems of high cost and low efficiency, which constitutes an obstacle to the clinical transformation of chimeric receptor-modified NK cells and significantly increases the production cost.
  • the non-viral vector system is an artificial synthesis system, which does not depend on any viral components or mammalian cells for production, and is low in cost, and at the same time, it is beneficial to avoid the immune problems caused by the use of viral vectors.
  • DNA transposon is a typical non-viral vector system with the advantages of sustainable gene expression, low immunogenicity, low cost and high efficiency.
  • DNA transposons are discrete DNA segments containing transposase genes flanked by inverted terminal repeats (ITRs) containing transposase binding sites.
  • ITRs inverted terminal repeats
  • the process of transposition is that the transposase binds to the ITRs, "cuts” the transposition from one location, and "pastes” it to another new location.
  • Transposon-based vector systems such as Sleeping Beauty (SB) system, PiggyBac (PB) system, and Tol transposon system, use the method of transposition to introduce transgenes into the host genome.
  • the present application provides a non-viral preparation of chimeric receptors
  • the receptor-modified NK cell method uses a transposon system to introduce stable and highly expressed chimeric receptors into NK cells to achieve efficient genetic modification of NK cells.
  • the present invention provides a method for preparing natural killer cells modified by chimeric receptors by utilizing a non-viral transposon system and artificial antigen-presenting cells, comprising:
  • the NK cells are primary human NK cells, which can be derived from human peripheral blood, umbilical cord blood, placental tissue or induced pluripotent stem cells (iPSCs).
  • iPSCs induced pluripotent stem cells
  • the chimeric receptors include chimeric antigen receptors, chimeric switching receptors, or other chimeric receptors synthesized by recombinant DNA technology.
  • the transposon system of step (1) includes the PiggyBac transposon system, the Sleeping Beauty transposon system, the Tol2 transposon system or other transposon systems using transposase.
  • the transposon system in step (1) includes a transposon element and a transposase element; the transposon element includes a transposon 5' inverted terminal repeat (5' ITR) and a 3' inverse terminal repeat.
  • the transposon can Is a plasmid or a linearized nucleic acid fragment; the transposase element can be encoded by a plasmid or a linearized nucleic acid fragment, or can be encoded by mRNA, or can be a protein.
  • the nucleic acid sequence of the transposase and the nucleic acid sequence of the transposon may be on one vector or on two different vectors.
  • genes encoding cytokines that promote the survival of NK cells can be added to the transposon element.
  • the cytokine can be secreted, membrane-bound, or a combination of cytokine-linked receptors; the cytokine encoding gene can be in the same transposon expression vector with the chimeric receptor encoding gene , also in different transposon expression vectors.
  • the IL-15 encoding gene that promotes the survival of NK cells is added to the transposon element, and is in the same transposon expression vector as the encoding gene of the chimeric receptor.
  • the method for using the transposon system to transfer the encoding gene of the chimeric receptor into NK cells includes electroporation, chemical transfection, and other non-viral transfection methods.
  • the relevant vectors of the transposon system are transferred into primary NK cells by electroporation.
  • the NK cells may or may not be activated in advance.
  • the NK cells are activated in advance.
  • the NK cells are activated in advance, it can be 0 to 14 days after the activation of the NK cells, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14.
  • the gene encoding the chimeric receptor was transferred into NK cells using the transposon system.
  • the gene encoding the chimeric receptor is transferred into the NK cells using a transposon system.
  • the gene encoding the chimeric receptor is transferred into the NK cells using a transposon system.
  • NK cells are activated in advance, artificial antigen-presenting cells, feeder cells, cytokines, antibodies or compounds can be used for activation.
  • the artificial antigen-presenting cells include cell-based, artificially synthesized or exosome-based artificial antigen-presenting cells.
  • the cell-based artificial antigen presenting cells are selected from human myeloid leukemia K562 cells, human Burkitt lymphoma Daudi cells, EBV transformed B lymphoblastoid cells (EBV-LCL) cells or mouse embryonic fibroblasts Cell line NIH/3T3 cells.
  • the cell-based artificial antigen presenting cells are selected from K562 cells.
  • the cell-based artificial antigen presenting cell is an engineered cell that expresses an antigen recognized by a chimeric receptor.
  • the engineered cells express ligand molecules, cytokines, etc. required for NK cell activation, wherein the cytokines may be secreted or membrane-bound.
  • the ligand molecules required for NK cell activation include CD137L and/or OX40L
  • the cytokines required for NK activation include any one of human IL-12, human IL-15, human IL-18 or human IL-21 or a combination of at least two.
  • the cell-based artificial antigen presenting cells and feeder cells are treated with gamma radiation (100 Gy) or mitomycin C (20 ⁇ g/mL).
  • both NK cell activation and CAR-NK cell expansion can be performed using artificial antigen presenting cells.
  • step (1) 0 to 14 days after the gene encoding the chimeric receptor is transferred into NK cells using the transposon system, for example, 1, 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11, 12, 13, or 14 days, using artificial antigen-presenting cells to stimulate the proliferation of chimeric receptor-modified NK cells.
  • the transposon system for example, 1, 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11, 12, 13, or 14 days, using artificial antigen-presenting cells to stimulate the proliferation of chimeric receptor-modified NK cells.
  • the chimeric receptor-modified NK cells in step (2) are expanded multiple times, and each expansion cycle is 3 to 14 days, such as 3, 4, 5, 6, 7, 8, 9, 10 , 11, 12, 13 or 14 days.
  • magnetic sorting can be used to enrich NK cells or CD3+ cells can be removed before step (1); or 0 to 14 days after step (1), magnetic sorting can be used to enrich NK cells cells or remove CD3+ cells; magnetic sorting can also be used to enrich NK cells or remove CD3+ cells after step (2).
  • the method further comprises the step of sorting CAR+ cells.
  • the method further includes the step of sorting CAR+ cells.
  • step (2) that is, after one round of co-culture with artificial antigen-presenting cells, sorting of CAR+ cells is performed.
  • a cytokine composition such as a combination of hrIL-12 and hrIL-18 or a combination of hrIL-12, hrIL-15 and hrIL-18, can be added during the preparation of chimeric receptor-modified NK cells using the transposon system The combination.
  • the cytokine composition can be added on any day during the preparation of the chimeric receptor-modified NK cells.
  • the cytokine composition can be removed by centrifugation one day after being added during the production of chimeric receptor-modified NK cells.
  • the final concentration of hrIL-12 is 1-100 ng/mL
  • the final concentration of hrIL-15 is 1-100 ng/mL
  • the final concentration of hrIL-18 is 1-100 ng/mL
  • the final concentration of hrIL-12 is 1-100 ng/mL.
  • the final concentration of hrIL-15 or hrIL-18 is the same or different, for example, it can be 1ng/mL, 10ng/mL, 20ng/mL, 30ng/mL, 40ng/mL, 50ng/mL, 60ng/mL, 70ng/mL, 80ng/mL, 90ng/mL or 100ng/mL.
  • the chimeric receptor-modified NK cell culture is carried out using a liquid medium
  • the liquid medium can be a cell culture medium commonly used in the art, such as AIM V TM (Gibco), X-VIVO TM 15 ( Lonza), SCGM TM (CellGenix), RPMI, NK MACS TM (Miltenyi), OpTmizer TM (Gibco), ImmunoCult TM -XF T Cell Expansion Medium (STEMCELL) or StemSpan TM (STEMCELL).
  • the present application provides a method for preparing chimeric receptor-modified NK cells using the PiggyBac transposon system, the method comprising:
  • PBMCs peripheral blood mononuclear cells
  • the helper plasmid encoding the PiggyBac (PB) transposon system and the donor plasmid encoding the chimeric acceptor were electroporated into NK cells;
  • chimeric receptor-modified NK cells were co-cultured with ⁇ -ray-treated engineered K562 cells, and added at the beginning of the culture.
  • the cell culture medium is AIMV medium supplemented with 1-5% human AB serum/human autologous plasma, and hrIL-2 is present during the culture.
  • the method for preparing chimeric receptor-modified NK cells using the PiggyBac transposon system according to the present invention is:
  • the helper plasmid encoding the PiggyBac (PB) transposon system and the donor plasmid encoding the chimeric acceptor were electroporated into NK cells;
  • the cell culture medium is AIMV medium supplemented with 1-5% human AB serum/human autologous plasma, and hrIL-2 is present during the culture.
  • lentiviral vectors or retroviral vectors also brings certain safety hazards to operators. Not only that, when using lentiviral vectors or retroviral vectors to prepare chimeric receptor-modified NK cells, There is also a potential risk of contamination of NK cell products by replicating retrovirus (RCR) or replicating lentivirus (RCL). Although the viral vector design and production system are constantly improving, this risk has been greatly reduced, but it cannot be completely ruled out. , the transposon system used in this application is a non-viral method, which is very safe and will not have these hidden dangers;
  • the integration efficiency of the gene encoding the chimeric receptor in NK cells is between 40% and 80%, which has reached the same level as that of using retrovirus to modify primary NK cells in the prior art.
  • FIG. 1A is the map of plasmid 2
  • FIG. 1B is the map of plasmid 4;
  • Figure 2A is the expression of membrane-bound human interleukin 15 on the surface of K562-NK1 cells
  • Figure 2B is the expression of membrane-bound human interleukin 21 on the surface of K562-NK1 cells
  • the abscissa is the membrane-bound human interleukin IL-15 or IL-21
  • the expression intensity of , the ordinate is the cell count;
  • Fig. 3 is the flow chart that utilizes PiggyBac transposon system to prepare CAR-NK;
  • Figure 4A shows the proportion of NK cells in the cell population on the 10th, 17th, and 24th days of culture for different CAR-NK preparation methods
  • Figure 4B shows the proportion of CAR+ cells in the cell population for different CAR-NK preparation methods on the 10th, 17th, and 24th days of culture. the proportion of NK cells;
  • Figure 5 shows the cumulative expansion fold of the total cell number on the 10th, 17th, and 24th days of culture for different CAR-NK preparation methods
  • Figure 6A shows the proportion of NK cells in the cell population of mononuclear cells (MNCs) from different sources at the end of the 1st, 2nd, 3rd, and 4th rounds of co-culture on day 0, and Figure 6B shows the ratios of MNCs from different sources in the first At the end of 2, 3, and 4 rounds of co-culture, the proportion of CAR+ cells in NK cells;
  • MNCs mononuclear cells
  • Figure 7 is the cumulative expansion fold of the total cell number of MNCs from different sources at the end of the first, second, third, and fourth rounds of co-culture;
  • Figure 8A shows the real-time killing map of NK and NKG2D CAR-NK cells on the human colorectal cancer cell line HCT116 on the 21st day after the third round of co-culture
  • Figure 8B shows the NK and NKG2D CAR on the 21st day after the third round of co-culture -
  • Figure 10A shows the expression of GFP in NK cells 1 day after the pmax-GFP plasmid was electroporated.
  • the NK cells used were NK cells on the 17th day of expansion.
  • the abscissa is the expression intensity of GFP
  • the ordinate is the cell count
  • Figure 10B is pmax -Number of NK cells before and after 1 day of electroporation of GFP plasmid
  • the NK cells used are NK cells on the 17th day of expansion
  • the abscissa is before and 1 day after electroporation, respectively
  • the ordinate is the number of cells;
  • Figure 11A shows the proportion of NK cells in the cell population on the 10th, 17th, 24th and 31st days of culture by different CAR-NK preparation methods
  • Figure 11B shows the ratio of NK cells in the cell population by different CAR-NK preparation methods on the 10th, 17th, 24th and 31st days of culture , the proportion of CAR+ cells in NK cells;
  • Figure 12 shows the cumulative expansion fold of the total cell number on the 10th, 17th, 24th, and 31st days of culture for different CAR-NK preparation methods
  • Figure 13A is the real-time killing map of human ovarian cancer cell line SKOV3 by NK, CAR-NK and eCAR-NK cells on the 17th day.
  • the coordinate is the cell index
  • Figure 13B shows the data analysis after adding effector cells for 24 hours to calculate the tumor growth inhibition rate, the abscissa is the different experimental groups, and the ordinate is the tumor growth inhibition rate;
  • Figure 14A is the detection of the survival of NK, NKG2D CAR-NK and NKG2D eCAR-15NK on the 22nd day of culture in the presence or absence of IL-2 for one week in vitro;
  • Figure 14B is the detection of the NK on the 21st day of culture , NKG2D eCAR-15NK and NKG2D eCAR-15 NK(sorted) were cultured in vitro for one week in the presence or absence of IL-2;
  • Figure 14C is the detection of NK, NKG2D eCAR-NK ( sorted) and NKG2D eCAR-mbIL15 NK(sorted) in vitro cultured without IL-2 for one week; ) Survival of one week in vitro culture with or without IL-2; among them, "-IL2" in the abscissa is the culture group without IL-2, and "+IL2" is the culture group with IL-2 added , the ordinate is the percentage change relative to the number of cells before one week
  • Figure 15A shows the killing results of NK, NKG2D CAR-mbIL15 NK, and NKG2D eCAR-mbIL15 NK (sorted) on the 24th day after the third round of co-culture on human acute myeloid leukemia cell line KG1, the abscissa is the effect-target ratio (E : T), the killing percentage on the ordinate;
  • Figure 15B is the killing result of NK and NKG2D eCAR-mbIL15 NK (sorted) on the human colorectal cancer cell line HCT116 on the 24th day after the third round of co-culture, the abscissa is the tumor cell plating
  • the time after (that is, after the start of the experiment), in hours, the ordinate is the cell index;
  • Figure 16 is the expression of BCMA on the surface of K562-NK2 cells
  • Figure 17A shows the proportion of NK cells in the cell population of peripheral blood mononuclear cells (PBMC) derived from a healthy donor at the end of the 1st, 2nd and 3rd rounds of co-culture on day 0;
  • Figure 17B shows a healthy donor The proportion of CAR+ cells in NK cells at the end of the 1st, 2nd and 3rd rounds of co-culture of the derived PBMC;
  • PBMC peripheral blood mononuclear cells
  • Figure 18 is the expansion fold of the total cell number in each round of PBMC derived from a healthy donor at the end of the 1st, 2nd, and 3rd rounds of co-culture;
  • Figure 19 shows the killing results of human multiple myeloma cell line U266 by NK and BCMA eCAR-NK on the 36th day at the end of the fifth round of co-culture, wherein the abscissa is the effect-target ratio (E:T), and the ordinate is the killing percentage.
  • PBMC peripheral blood of solid tumor patients or healthy donors
  • MNC derived from umbilical cord blood was purchased from Maishun Biotechnology Co., Ltd.
  • human chronic myeloid leukemia cell line wild-type K562 cells were purchased from ATCC
  • human multiple myeloma cell line U266 was purchased from Wuhan Proceeds Life Technology Co., Ltd.
  • the other reagents or consumables not mentioned were all from conventional reagent manufacturers in the field or prepared by conventional means in the field;
  • the medium of human ovarian cancer cell line SKOV3 and human colorectal cancer cell line HCT116 is McCoy 5A (Gibco) + 10% FBS (Gibco)
  • the medium of human gastric cancer cell lines MGC803 and U266 is RPMI (Gibco) + 10% FBS (Gibco)
  • the medium of K562 and human AML cell line KG1 is IMDM (Gibco) + 10% FBS (Gibco);
  • the K562-NK1 cells used in the examples are ⁇ -ray-treated (100 Gy) genetically engineered K562 encoding membrane-bound human IL-15 (mbIL15), membrane-bound human IL-21 (mbIL21), eGFP and Puromycin cell. Since K562 cells naturally express NKG2D ligands, the antigens recognized by NKG2D CAR are not additionally loaded into K562-NK1 cells here.
  • Membrane-bound human IL-15 is composed of the GM-CSF Ra signal peptide (amino acids 1-22 of UniprotKB P15509), human IL-15 (amino acids 30-162 of UniprotKB P40933) and the hinge transmembrane region of human CD8 (UniprotKB P01732 Amino acids 128-213) are fused together; membrane-bound human IL-21 is composed of GM-CSF R ⁇ signal peptide (amino acids 1-22 of UniprotKB P15509), human IL-21 (amino acids 25-162 of UniprotKB Q9HBE4) ), the hinge constant region of human IGHG4 (the 99th to 327th amino acids of UniprotKB P01861), the transmembrane region of human CD4 (the 397th to 418th amino acids of UniprotKB P01730) are fused together, and the amino acid sequence of eGFP is as shown in SEQ ID No:3 Shown, the amino acid sequence of Puro
  • the K562-NK2 cells used in the examples were genetically engineered to encode membrane-bound human IL-15 (mbIL15), membrane-bound human IL-21 (mbIL21), truncated BCMA (tBCMA), eGFP and Puromycin K562 cells.
  • mbIL15 membrane-bound human IL-15
  • mbIL21 membrane-bound human IL-21
  • tBCMA truncated BCMA
  • eGFP Puromycin K562 cells.
  • the sequence of tBCMA is the amino acid sequence of positions 1-77 of UniProt KB-Q02223.
  • Human recombinant IL-2 (hrIL2) was purchased from Beijing Shuanglu Company, and human recombinant IL-12 (hrIL12), human recombinant IL-15 (hrIL15), and human recombinant IL-18 (hrIL18) were purchased from Nearshore Protein Company;
  • NK cell culture medium AIM medium (Gibco company) + 5% human AB serum (Gemini company);
  • P3 Primary Cell 4D-Nucleofector X Kit was purchased from Lonza Company;
  • PE-conjugated anti-human CD3 antibody, APC-conjugated anti-human CD56 antibody were purchased from BD
  • FITC-conjugated anti-Strep-tag II antibody was purchased from GenScript
  • Biotin-conjugated anti-human IL-15 antibody and APC Conjugated Streptavidin was purchased from Biolegend Company
  • Biotin-conjugated anti-human IL-21 antibody was purchased from eBioscience Company
  • anti-Biotin microbeads and Streptavidin micobeads were purchased from Miltenyi Company
  • Flow cytometer was purchased from BD Company, model C6 Sampler; real-time killing detector was purchased from ACEA Bio Company, model xCELLigence RTCA DP; human IFN ⁇ ELISA detection kit was purchased from Biolegend Company; Calcein-AM was purchased from Yisheng Biotechnology; pmax-GFP was purchased from Lonza Company;
  • the amino acid sequence of PiggyBac transposase is from GenBank: AAA87375.2; the NKG2D CAR is: NKG2D-CD28-4-1BB-CD3 ⁇ , the antigen-binding domain is from amino acids 83-216 of NKG2D (UniProtKB-P26718), and the hinge region is from Amino acids 99-110 of IgG4 (UniProtKB-P01861), transmembrane region from amino acids 153-179 of CD28 (UniProtKB-P10747), intracellular domain from 209-255 of 4-1BB (UniProtKB-Q07011) BCMA CAR is: anti-BCMA ScFv-CD28-4-1BB-DAP12 ⁇ , the antigen binding domain is from the scFv, hinge of Anti-BCMA antibody C11D5.3 The region is derived from amino acids 99-110 of IGHG4 (UniProtKB-P01861), the transmembrane region is derived from amino acids 153
  • DNA fragment 1 CMV promoter nucleotide sequence, puromycin coding nucleotide sequence, T2A, EGFP coding nucleotide sequence were synthesized by conventional biotechnology service companies in the field;
  • DNA fragment 2 CMV promoter nucleotide sequence, mbIL-15 coding nucleotide sequence, P2A coding nucleotide sequence, mbIL-21 coding nucleotide sequence are synthesized by conventional biotechnology service companies in the field;
  • DNA Fragment 1 and DNA Fragment 2 were cloned into pFastBac1 plasmid (purchased from Thermofisher) and designated as plasmid 1 .
  • the 5' inverted terminal repeat of the PiggyBac transposon (SEQ ID NO: 1), the chicken ⁇ -globin chromatin insulator cHS4 (GenBank: AY040835.1), the EF1 ⁇ promoter, the EcoRI and SalI restriction site sequences , SV40 polyA sequence, reverse complementary cHS4 sequence and 3' reverse terminal repeat sequence (SEQ ID NO: 2) were fused, and synthesized by an outsourced service company, cloned into pmaxCloning vector (purchased from Lonza company) through BsaI, named For pZTS4, the expression of the fusion gene is controlled by the EF1 ⁇ promoter;
  • SEQ ID NO: 1 5'-ccctagaaagataatcatattgtgacgtacgttaagataatcatgtgtaaaattgacgcatg-3';
  • SEQ ID NO: 2 5'-catgcgtcaattttacgcagactatctttctaggg-3';
  • the extracellular antigen-binding domain (ED) of NKG2D was fused with the IGHG4 hinge region, CD28 transmembrane region, 4-1BB and CD3 ⁇ signaling domain to generate the second-generation NKG2D CAR vector; CAR expression was detected by cytometry, and 3 repeats of Strept-tag II (ST2) were added to the sequence of the CAR; the CAR fragment was synthesized by a conventional biotechnology service company in the field, and the 5' and 3' ends of the sequence included restriction Sexual restriction sites EcoRI and SalI; after the synthesized DNA fragment was digested by EcoRI and SalI, it was cloned into pFastBac1 plasmid (purchased from Thermofisher) and named as plasmid 3.
  • pFastBac1 plasmid purchased from Thermofisher
  • the CAR sequence in plasmid 2 was digested with EcoRI and SalI and cloned into pZTS4, named plasmid 4. As shown in Figure 1B, the expression of the CAR gene is controlled by the EF1 ⁇ promoter. control.
  • the sequence of IRES and human IL-15 (UniprotKB: P40933) was fused, and the 5' and 3' ends of the sequence included the restriction site SalI .
  • the synthesized DNA fragment was digested with SalI and cloned into plasmid 4, which was named plasmid 5.
  • the NKG2D CAR transposon donor vector containing the membrane-bound human IL-15 (mbIL-15) gene the IRES and the sequence of membrane-bound human IL-15 were fused, including restriction at the 5' and 3' ends of the sequence The cleavage site SalI.
  • the synthesized DNA fragment was digested with SalI and cloned into plasmid 4, which was named plasmid 6.
  • the anti-BCMA scFv (C11D5.3) was fused with the IGHG44 hinge region, CD28 transmembrane region, 4-1BB and CD3 ⁇ signaling domains to generate a second-generation anti-BCMA CAR vector; in order to To facilitate the detection of CAR expression by flow cytometry, 3 repeats of Strept-tag II (ST2) are added to the CAR sequence; the CAR fragment is synthesized by an outsourced service company, and the 5' and 3' ends of the sequence include restriction Sex restriction sites EcoRI and SalI; the synthesized DNA fragment was digested by EcoRI and SalI, and then cloned into pFastBac1 plasmid (purchased from Thermofisher), named plasmid 7.
  • ST2 Strept-tag II
  • the CAR sequence in plasmid 7 was digested with EcoRI and SalI and cloned into pZTS4, which was named plasmid 8.
  • the expression of the CAR gene was controlled by the EF1 ⁇ promoter.
  • plasmid 9 the DNA sequence of tBCMA was synthesized by an outsourcing company, and cloned into pZTS4 by EcoRI and SalI, named plasmid 9.
  • the wild-type K562 cells were resuspended in 10 mL Opti-MEM, centrifuged at 200 ⁇ g for 5 min, the cell pellet was resuspended in 100 ⁇ L P3 buffer, 10 ⁇ g plasmid 1 was added, and after mixing, it was transferred to the Lonza electric shock cup;
  • puromycin screening was performed at a concentration of 200 ⁇ g/mL for 1 month, and the medium was changed every 2 days;
  • K562-NK1 cells expressed high levels of human IL-15 and human-IL21 on the surface, with positive rates of 100.0% and 78.9%, respectively.
  • PBMCs and 5 ⁇ 10 6 K562-NK1 were resuspended in 10 mL of NK cell culture medium, seeded into T25 cell culture flasks, and hrIL-2 was added to make the final concentration is 100IU/mL;
  • the suspended cells were taken out and counted, and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet in 100 ⁇ L P3 buffer, and added 5 ⁇ g plasmid 2 and 10 ⁇ g Plasmid 4, mix well and transfer to the electroporation Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module), and perform electroporation.
  • NK cell suspension After the electroporation, slowly transfer the NK cell suspension in one electroporation cup Transfer to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing a final concentration of 100IU/mL hrIL-2, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
  • This example compares the ratio of NK cells in CAR-NK, the ratio of CAR expression in NK cells, and the total cells obtained by electroporation before NK cell activation or on the 2nd or 4th day after NK cell activation. the amplification fold.
  • Example 3 Electrically At day 2 activated NK cells transfected is the procedure of Example 3 is referred to as Day2 EP; method of electroporation prior to activation of NK cells is different from the embodiment 3 in that the 5 ⁇ 10 6 PBMC were as described in embodiment The method of Example 3 was electroporated. Immediately after electroporation, it was co-cultured with 5 ⁇ 10 6 K562-NK1 (100Gy ⁇ -ray irradiation), which was recorded as Day0 EP; electroporation was performed on the 4th day after the activation of NK cells, which was different from that in Example 3.
  • 5 ⁇ 10 6 K562-NK1 100Gy ⁇ -ray irradiation
  • the above three methods were used to prepare CAR-NK cells from a normal donor PBMC, and the obtained NK cell purity (CD3-CD56+) was compared.
  • the results are shown in Figure 4A.
  • the purity of NK cells was 68%, 77%, and 85% on the 10th, 17th, and 24th days of culture, respectively, while the purity of NK cells in the CAR-NK cells prepared by electroporation before NK cell activation was on the 10th, 17th day of culture. were 11% and 13%, respectively, and the purity of NK cells in CAR-NK cells prepared by electroporation 4 days after NK cell activation were 46%, 81%, and 88% on culture days 10, 17, and 24, respectively. It shows that the purity of NK cells is higher in the CAR-NK cells prepared by electroporation after NK cell activation than that prepared by electroporation before NK activation.
  • the expression ratio of CAR in the obtained CAR-NK cells was compared, and the results were shown in Figure 4B. were 14%, 30%, and 45%, while the proportion of CAR+ NK cells in CAR-NK cells prepared by electroporation before NK cell activation was 14% and 27% on the 10th and 17th days of culture, respectively.
  • the proportion of CAR+ NK cells in the CAR-NK cells prepared by electroporation on day 4 was 2%, 15%, and 19% on the 10th, 17th, and 24th days of culture, respectively. It shows that the earlier electroporation is performed, the higher the proportion of CAR+ NK cells.
  • This example also compares the amplification multiples of the total number of cells obtained.
  • the results are shown in Figure 5.
  • the amplification multiples of the total number of cells obtained by the method of Example 3 for 24 days is 3022 times; After a total of 17 days of expansion, the total number of cells was expanded by 133 times; 4 days after the activation of NK cells, electroporated for 24 days, the total number of cells was expanded by 736 times. This indicated that the cells obtained by electroporation on the second day after the activation of NK cells had the highest expansion fold.
  • Example 3 Based on the comparison of these three parameters, the method of Example 3, that is, electroporation was performed 2 days after the activation of NK cells, the prepared CAR-NK cells, the purity of NK cells, the proportion of CAR+ NK cells, and the amplification fold of the total number of cells. Relatively best.
  • Example 5 Mononuclear cells from multiple sources can use the PiggyBac transposon system to prepare NKG2D CAR-NK
  • NKG2D CAR-NK 7 mononuclear cells (MNCs) from different sources were used to prepare NKG2D CAR-NK according to the method described in Example 3, wherein HD001, HD002, HD003, HD004, HD005, and HD006 were extracted from peripheral blood of healthy donors.
  • PBMC, CB001 was derived from MNC extracted from umbilical cord blood.
  • Example 6 Direct electroporation of NK cells, transgene expression efficiency and NK cell survival rate
  • the green fluorescent protein GFP gene was used as the reporter gene, and the pmax-GFP plasmid was directly used to electroporate the NK cells amplified for 17 days.
  • the NK cell expansion procedure is as follows: On day 0, 2 ⁇ 10 6 PBMCs and 2 ⁇ 10 6 K562-NK1 (100Gy ⁇ -ray irradiated) were resuspended in 10 mL of NK cell culture medium, and seeded in T75 cell culture flasks (vertical).
  • hrIL-2 was added to make the final concentration 100IU/mL; from the 2nd day to the 10th day, according to the cell growth, an appropriate amount of fresh NK cell culture medium containing hrIL-2 was added; on the 10th day, all cells were collected Counting, taking 2 ⁇ 10 6 cells for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody); 2 ⁇ 10 6 cells and 2 ⁇ 10 6 K562-NK1 (100Gy ⁇ -ray irradiation) ) was resuspended in 10 mL of medium, and hrIL-2 was added to make its final concentration 100 IU/mL; from the 10th day to the 17th day, according to the cell growth, an appropriate amount of fresh NK cell culture medium containing hrIL-2 was added; the 17th day On the next day, all cells were collected and counted, and 2 ⁇ 10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD
  • the NK cells on the 17th day of expansion were taken, and the electroporation method in Example 3 was used.
  • the number of electroporated NK cells was 2 ⁇ 10 6 , and the plasmid was replaced by the pmax-GFP plasmid.
  • cell counting and flow cytometry were performed to detect the proportion of GFP in NK cells.
  • Figure 10A and Figure 10B on the second day of electroporation, although the expression ratio of GFP was 17.99%, the survival rate of NK cells was only 3.2%.
  • PBMCs and 5 ⁇ 10 6 K562-NK1 were resuspended in 10 mL of NK cell culture medium, seeded into T25 cell culture flasks, and hrIL-2 was added to make the final concentration is 100IU/mL;
  • the suspended cells were taken out for counting, and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet in 100 ⁇ L P3 buffer, and added 5 ⁇ g plasmid 2 and 10 ⁇ g Plasmid 4, mix well and transfer to the Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module) and perform electroporation.
  • NK cell culture medium containing hrIL-2 After the electroporation, slowly transfer the NK cell suspension in one electroporation cup to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 100IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
  • Example 8 Effects of different cytokine compositions on the expansion fold and cell phenotype of CAR-NK cells
  • This example compares the effect of adding cytokine compositions (IL-12, IL-15, IL-18) on the growth of CAR-NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation. Impact.
  • cytokine compositions IL-12, IL-15, and IL-18 were added (Example 7).
  • NK cell purity (CD3-CD56+) was compared.
  • the purity of NK cells in NK cells was 79%, 81%, 90%, and 93% on the 10th, 17th, 24th, and 31st days of culture, respectively, while the purity of NK cells in the eCAR-NK cells prepared in Example 4 was on the 1st day of culture. At 10, 17, 24, and 31 days, they were 79%, 87%, 93%, and 93%, respectively.
  • cytokines IL-12, IL-15 and IL-18 can slightly increase the proportion of NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation.
  • the ratio of NK cells of CAR+ in the obtained cells was compared.
  • the results are shown in Figure 11B.
  • the ratio of NK cells of CAR+ in the CAR-NK cells prepared in Example 3 was on the 10th, 17th, 24th, and 31st days of culture, respectively. were 34%, 47%, 49%, and 74%, while the proportion of CAR+ NK cells in the eCAR-NK cells prepared in Example 7 was 34%, 57%, and 58% on the 10th, 17th, 24th, and 31st days of culture, respectively. , 85%.
  • the total cell expansion folds obtained in the preparation process of CAR-NK and eCAR-NK cells are compared.
  • the results are shown in Figure 12.
  • the total expansion fold of the CAR-NK cells prepared in Example 3 is in 0-10 days, 10-17 days, 17-24 days and 24-31 days were 27, 21, 9, and 8 times respectively; while the eCAR-NK cells prepared in Example 7 had a total expansion fold at the 0- 10 days, 10-17 days, 17-24 days and 24-31 days were 27, 27, 9, and 7 times, respectively.
  • Example 7 Comprehensively comparing the data of the above three aspects, the method described in Example 7, that is, adding the cytokine compositions IL-12, IL-15, IL- 18. Contribute to the expansion of CAR+ NK cells.
  • Example 9 The effect of adding cytokine composition on the 10th day of CAR-NK preparation on the killing of human ovarian cancer cell line SKOV3 in vitro (RTCA)
  • This example compares the effect of adding cytokine compositions (IL-12, IL-15, IL-18) on the in vitro effects of CAR-NK on the 10th day of NKG2D CAR-NK preparation, when it was co-cultured with K562-NK1 for the second time. impact on lethality.
  • cytokine compositions IL-12, IL-15, IL-18
  • SKOV3 cells were plated in a 16-well electrode plate (ACEA Bio Company), 5000 cells/well; about 24 hours later, the NKG2D CAR-NK cells, NKG2D eCAR-NK cells and NKG2D eCAR-NK cells were cultured on the 17th day.
  • NK cells effector cells
  • SKOV3:NK 1:1” in the figure or CAR-NK cells (shown as “SKOV3:CAR-NK 1:1” in the figure)
  • the growth of tumor cells can be significantly inhibited, in which the killing of CAR-NK cells
  • the tumor effect was significantly better than that of NK cells, which indicated the effect of CAR in NKG2D CAR-NK cells; while adding eCAR-NK cells (shown as "SKOV3:eCAR-NK 1:1” in the figure) or CAR-NK cells (Fig. shown as "SKOV3:CAR-NK 1:2"), the tumor cells were almost completely killed.
  • the specific steps include: on the 0th day, 2 ⁇ 10 7 PBMC cells were taken, and CD3+ cells were removed with CD3 magnetic beads (Miltenyi) according to the manufacturer’s recommended procedure; 5 ⁇ 10 6 sorted PBMCs and 5 ⁇ 10 6 cells were removed K562-NK1 (100Gy ⁇ -ray irradiation) was resuspended in 10mL of NK cell culture medium, inoculated into a T25 cell culture flask, and hrIL-2 was added to make the final concentration 50IU/mL;
  • the suspended cells were taken out for counting, and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet in 100 ⁇ L P3 buffer, and added 5 ⁇ g plasmid 2 and 10 ⁇ g Plasmid 4, mix well and transfer to the Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module) and perform electroporation.
  • NK cell culture medium containing hrIL-2 After the electroporation, slowly transfer the NK cell suspension in one electroporation cup to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 50IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
  • CAR-NK cells were resuspended in 80 ⁇ L of DPBS+1% AB serum buffer, added with 20 ⁇ L of anti-Strep tag II antibody, mixed and incubated at 2-8°C for 20 min; after incubation, 5 mL of buffer was added and centrifuged at 300g 10min, discard the supernatant and resuspend in 80 ⁇ L DPBS+1% AB serum buffer, add 20 ⁇ L anti-Biotin magnetic beads, mix well, incubate at 2-8°C for 15min; add 5mL buffer after incubation, centrifuge at 300g for 10min , discard the supernatant and resuspend in 500 ⁇ L of buffer, and then follow the steps recommended by the Mil
  • Example 11 CAR+ cell sorting can significantly increase the proportion of CAR+ NK cells
  • the NKG2D CAR-NK cells on the 10th day after the first round of co-culture were taken and divided into three groups: in the first group, 2 ⁇ 10 6 cells were co-cultured with 2 ⁇ 10 6 K562-NK1 (100Gy ⁇ -ray irradiation) cells1 Week, named as the unsorted group; in the second group, 1 ⁇ 10 7 cells were sorted according to the method described in Example 10.
  • NK cells:artificial antigen-presenting cells 1:2 and K562 -NK1 (100Gy ⁇ -ray irradiation) was co-cultured for 1 week, named Biotin Anti-Strep tag II Ab+anti-Biotin microbeads group; 1 ⁇ 10 7 cells were taken from the third group and resuspended in 90 ⁇ L DPBS+1% AB serum Add 10 ⁇ L of Streptavidin microbeads to the buffer, mix well and incubate at 2-8 °C for 20 min; after incubation, add 5 mL of buffer, centrifuge at 300 ⁇ g for 10 min, discard the supernatant and resuspend in 500 ⁇ L of buffer, and then proceed with magnetic beads The labeled positive cells were sorted, and the remaining cells were taken after sorting and co-cultured with K562-NK1 (100Gy ⁇ -ray irradiation) for 1 week according to NK cells:art
  • the sorting yield of Biotin Anti-Strep tag II Ab+anti-Biotin microbeads is higher, and the proportion of CAR+ cells in NK cells after one round of expansion can be increased to more than 90%.
  • Example 12 Adding secreted or membrane-bound human IL-15 to the CAR carrier can promote the survival of CAR-NK cells in vitro
  • NK NKG2D CAR-NK
  • eCAR-IL15 NKG2D eCAR-IL15 NK
  • NKG2D CAR-NK refers to Example 3; the preparation of NKG2D eCAR-IL15 NK refers to Example 7, and plasmid 4 is replaced with plasmid 5. Since the CAR vector contains IL-15, the cytokine composition on the 17th day IL-15 was removed. Take 2 ⁇ 10 6 cells in each group on the 22nd day after the third round of co-culture, and count them after 1 week of in vitro culture with and without addition of hrIL-2 (50 IU/mL), and calculate the percentage change of cells ( 100% ⁇ the number of cells after 1 week of culture/2 ⁇ 10 6 ).
  • NK, NKG2D eCAR-IL15 NK and NKG2D eCAR-IL15 NK were prepared from umbilical cord blood, respectively.
  • NKG2D eCAR-IL15 NK refer to Example 7, and replace plasmid 4 with plasmid 5. Since the CAR vector contains IL-15, IL-15 was removed from the cytokine composition on day 17; NKG2D eCAR-IL15 NK (sorted) Preparation Refer to Example 10, replace plasmid 4 with plasmid 5, since IL-15 is contained in the CAR vector, IL-15 is removed from the cytokine composition on day 17.
  • the percentage of cell change in NK, NKG2D eCAR-NK(sorted), NKG2D eCAR-mbIL15 NK(sorted) were 6%, 13%, and 140%, respectively, and the membrane Binding of IL-15 (mbIL-15) can also promote the survival of CAR-NK in vitro.
  • NK, NKG2D CAR-mbIL15 NK and NKG2D eCAR-mbIL15 NK were prepared from PBMCs, respectively.
  • the preparation of NKG2D CAR-mbIL15 NK refers to Example 3, and plasmid 4 is replaced with plasmid 6. Since the CAR vector contains mbIL-15, IL-15 is removed from the cytokine composition on day 17; NKG2D eCAR-mbIL15 NK (sorted ) Preparation of ) Refer to Example 10, replace plasmid 4 with plasmid 6, since mbIL-15 is contained in the CAR vector, IL-15 is removed from the cytokine composition on day 17.
  • NK, NKG2D eCAR-mIL15 NK and NKG2D eCAR-mbIL15 NK were prepared respectively with PBMC as the source, wherein the preparation of NKG2D eCAR-mbIL15 NK refers to Example 7, and plasmid 4 was replaced with plasmid 6, because CAR The vector contains mbIL-15, and IL-15 is removed from the cytokine composition on day 17; the preparation of NKG2D eCAR-mbIL15 NK (sorted) refers to Example 10, and plasmid 4 is replaced with plasmid 6, because the CAR vector contains mbIL -15, Depletion of IL-15 in the cytokine composition on day 17.
  • the cells on the 16th day after the second round of co-culture were taken for in vitro killing test.
  • the human AML cell line KG1 cells were taken and resuspended in DPBS at 1 ⁇ 10 6 cells/ml, and Calcein-AM (final concentration 0.2 ⁇ M) was added for staining at 37° C. for 15 min. After staining, FBS was added in an equal volume to stop the staining, and washed three times with DPBS.
  • the stained KG1 was resuspended in the medium, the cell density was adjusted to 2 ⁇ 10 5 cells/ml, and KG1 was plated in 100 ⁇ L per well in a U-shaped 96-well plate.
  • the tumor growth inhibition rate was calculated according to the following formula:
  • the K562-NK1 cells were resuspended in 10 mL Opti-MEM, centrifuged at 300 ⁇ g for 10 min, the cell pellet was resuspended in 100 ⁇ L P3 buffer, 5 ⁇ g plasmid 2 and 10 ⁇ g plasmid 9 were added, and the cells were transferred to the Lonza electric shock cup; The cups were placed in a Lonza 4D-NucleofectorTM X Unit (in a single shock cup module) for electroporation.
  • K562 cell suspension in an electroporation cup was slowly transferred to a well of a 6-well plate, and K562 medium (IMDM+10% FBS) was pre-added to the well; on the 7th day after electroporation, the cells were used
  • the sorting instrument BD Fusion sorts single cells into 96-well plates, and analyzes the amplified single cell clones by flow cytometry to detect the expression of BCMA.
  • the detection antibody is APC-conjugated anti-human BMCA.
  • Antibody, the screened single cell clone was named K562-NK2.
  • K562-NK2 cells expressed high levels of BCMA on the surface, with a positive rate of 99.4%.
  • the suspended cells were taken out for counting, and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 ⁇ g for 10 min; resuspended the cell pellet in 100 ⁇ L P3 buffer, and added 5 ⁇ g plasmid 2 and 10 ⁇ g Plasmid 8, after mixing, transfer to the electroporation Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module), and perform electroporation.
  • NK cell suspension After the electroporation, slowly transfer the NK cell suspension in one electroporation cup Transfer to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 50IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
  • Figure 18 shows the expansion fold of the total number of cells in each round at the end of the first, second, and third rounds of co-culture during the preparation of BCMA eCAR-NK from a healthy donor-derived PBMC, and the expansion of the first round of co-culture.
  • the multiplication factor was 13 times
  • the second round of co-culture was 60 times
  • the third round of co-culture was 46 times.
  • U266 cells were taken and resuspended in DPBS at 1 ⁇ 10 6 cells/mL, and Calcein-AM (final concentration 0.2 ⁇ M) was added for staining at 37° C. for 15 min. After staining, FBS was added in an equal volume to stop the staining, and washed three times with DPBS. The stained U266 cells were resuspended in the medium, the cell density was adjusted to 2 ⁇ 10 5 cells/mL, and 100 ⁇ L per well of U266 was plated in a U-shaped 96-well plate.
  • the present application adopts a non-viral method and uses a transposon system to introduce a stable and highly expressed transgene into NK cells to achieve efficient genetic modification of NK cells.
  • the production cycle is short and the production cost is low.
  • the prepared transgenic modification NK cells have high purity and good safety, and have significant killing and/or inhibitory effects on tumor cells.
  • the present application illustrates the detailed method of the present application through the above-mentioned embodiments, but the present application is not limited to the above-mentioned detailed method, which does not mean that the present application must rely on the above-mentioned detailed method for implementation.
  • Those skilled in the art should understand that any improvement to the application, the equivalent replacement of each raw material of the product of the application, the addition of auxiliary components, the selection of specific methods, etc., all fall within the scope of protection and disclosure of the application.

Abstract

Provided are a method for preparing stable natural killer (NK) cells with high chimeric receptor expression by means of a non-viral method and the use thereof, specifically comprising: (1) transferring the coding gene of the chimeric receptor into a natural killer cell using a transposon system to obtain an initial chimeric receptor-modified natural killer cell; and (2) amplifying the initial chimeric receptor-modified natural killer cell using an artificial antigen-presenting cell. The chimeric receptor-modified NK cell prepared by means of the method has a relatively strong tumor killing ability, and shows a relatively good anti-tumor effect in vitro.

Description

非病毒方法制备稳定高表达嵌合受体的NK细胞Preparation of NK cells with stable and high expression of chimeric receptors by non-viral method 技术领域technical field
本申请属于医学生物工程技术领域,涉及应用非病毒方法制备稳定高表达嵌合受体修饰的NK细胞的方法及其应用。The present application belongs to the technical field of medical bioengineering, and relates to a method for preparing NK cells modified by stable and high expression of chimeric receptors by using a non-viral method and its application.
背景技术Background technique
自然杀伤(NK)细胞是先天性免疫系统的重要组成部分,在外周血淋巴细胞中NK细胞的比例约10%,是继B细胞和T细胞之后的第三大淋巴细胞群体。NK细胞具有多种生物学功能,是人体对抗感染和肿瘤的第一道防线。NK细胞的功能通过精密且复杂的激活型和抑制型受体网络进行调节。大部分循环的NK细胞处于静息期,可以被细胞因子活化并浸润到病原体感染或含有恶性细胞的组织中。当NK细胞的受体与相应的配体结合后,NK细胞还可以分泌一些细胞因子,例如干扰素(IFN)-γ,发挥免疫调节作用。NK细胞的主要功能之一是对机体进行免疫监督,研究者已发现NK细胞参与控制多种疾病的发生发展。对哺乳动物细胞(包括人体细胞)的一些体外研究以及对小鼠和大鼠的体内研究表明,NK细胞可以将肿瘤细胞识别为靶标,并在体内控制肿瘤细胞的生长、转移和扩散。一项为期11年的随访流行病学调查显示,外周血NK细胞活性降低会增加成人患癌症的风险。不仅如此,NK细胞作为宿主免疫的作用也在各种黄病毒感染病例中进行了研究,如日本脑炎病毒、黄热病病毒、登革热病毒、蜱传脑炎病毒和西尼罗病毒(WNV);它们在病毒性肝炎、流感病毒和HIV-1感染中的作用也在一些研究中得到了很好的证明;同样,它们在防止细菌、呼吸道合胞病毒(RSV)和流感等病毒的呼吸道感染方面的作用在小鼠研究中也有详细描述。(具体可参考文献:Mandal A,Viswanathan C.等,Natural killer cells:In health and diseas[J].Hematology/oncology&Stem Cell Therapy,2015,8(2):47-55.)。Natural killer (NK) cells are an important part of the innate immune system. The proportion of NK cells in peripheral blood lymphocytes is about 10%, and they are the third largest lymphocyte population after B cells and T cells. NK cells have multiple biological functions and are the body's first line of defense against infections and tumors. NK cell function is regulated by a sophisticated and complex network of activating and inhibitory receptors. Most circulating NK cells are in a quiescent phase and can be activated by cytokines and infiltrate pathogen-infected or malignant cells-containing tissues. When the receptors of NK cells are combined with the corresponding ligands, NK cells can also secrete some cytokines, such as interferon (IFN)-γ, to play an immunoregulatory role. One of the main functions of NK cells is to supervise the immune system of the body. Researchers have found that NK cells are involved in controlling the occurrence and development of various diseases. Several in vitro studies on mammalian cells, including human cells, and in vivo studies in mice and rats have shown that NK cells can recognize tumor cells as targets and control tumor cell growth, metastasis, and spread in vivo. Decreased peripheral blood NK cell activity increases the risk of cancer in adults, an 11-year follow-up epidemiological survey showed. Not only that, the role of NK cells as host immunity has also been studied in various cases of flavivirus infection such as Japanese encephalitis virus, yellow fever virus, dengue virus, tick-borne encephalitis virus and West Nile virus (WNV) ; their role in viral hepatitis, influenza virus and HIV-1 infection has also been well documented in several studies; likewise, their role in preventing respiratory tract infections by viruses such as bacteria, respiratory syncytial virus (RSV) and influenza The role of Aspects has also been described in detail in mouse studies. (For details, please refer to: Mandal A, Viswanathan C. et al., Natural killer cells: In health and diseas[J]. Hematology/oncology & Stem Cell Therapy, 2015, 8(2): 47-55.).
与MHC限制的T细胞不同,NK细胞不需要进行HLA配型,因此可以作为异源性的现货细胞药品对病人进行免疫治疗,在临床上具有重要的应用前景。基于自体或同种异体NK细胞的过继性疗法在临床上已经应用于抗肿瘤、抗病毒感染,特别是异体NK细胞对于治疗血液系统癌症显示出良好的治疗效果(参考文献:Lupo K B,Matosevic S.等,Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy[J].Cancers,2019,11(6).)。Unlike MHC-restricted T cells, NK cells do not require HLA matching, so they can be used as heterologous off-the-shelf cell drugs for immunotherapy of patients, which has important clinical application prospects. Adoptive therapy based on autologous or allogeneic NK cells has been clinically applied to anti-tumor and anti-viral infection, especially allogeneic NK cells have shown good therapeutic effects in the treatment of hematological cancers (Reference: Lupo K B, Matosevic S. et al., Natural Killer Cells as Allogeneic Effectors in Adoptive Cancer Immunotherapy [J]. Cancers, 2019, 11(6).
尽管NK细胞具有强大的免疫效应功能,但是许多癌细胞和病毒会通过抗原逃逸和免疫抑制等机制来抑制NK细胞的功能。由于这些原因,在过去的十年时间中,科学家们一直在探索通过基因工程的方法来增强NK细胞的功能活性并避免免疫抑制,例如通过表达内源性细胞因子来增强过继回输后的NK细胞的增殖能力,抑制免疫抑制信号或增强NK细胞的杀伤功能等。后一种方法目前主要通过修饰嵌合抗原受体(CARs)使NK细胞重新定向于肿瘤细胞。嵌合抗原受体是将抗体的scFv与胞内信号转导结构融合在一起,scFv用于介导识别和结合肿瘤细胞表面抗原。Although NK cells have powerful immune effector functions, many cancer cells and viruses suppress the function of NK cells through mechanisms such as antigen escape and immunosuppression. For these reasons, over the past decade, scientists have been exploring genetic engineering approaches to enhance the functional activity of NK cells and avoid immunosuppression, such as by expressing endogenous cytokines to enhance NK after adoptive reinfusion Cell proliferation ability, inhibiting immunosuppressive signals or enhancing the killing function of NK cells, etc. The latter approach currently redirects NK cells to tumor cells mainly by modifying chimeric antigen receptors (CARs). A chimeric antigen receptor is a fusion of an antibody's scFv with an intracellular signal transduction structure, and the scFv is used to mediate the recognition and binding of tumor cell surface antigens.
然而,原代NK细胞对于基因转染的抵抗力较强,这导致NK细胞对多种载体系统的摄取率低,NK细胞的转基因表达量低,只有病毒载体在原代NK细胞中实现了较高的基因转移效率。目前,对原代NK细胞进行嵌合受体的修饰一般通过电转嵌合受体编码基因的mRNA或者采用慢病毒/逆转录病毒感染的方式来实现。mRNA电转只能使嵌合受体编码基因瞬时表达,且体外合成mRNA的成本较高,临床上需要多次注射,治疗费用较高。慢病毒/逆转录病毒转染则可以制备嵌合受体基因稳定表达的NK细胞,然而与NK细胞株如NK92比较,原代NK细胞感染难度极大,迄今为止,只有少数科研团队成功应用慢病毒/逆转录病毒制备嵌合抗原受体修饰的原代NK细胞,经多次重复感染后,其转基因在终产品中的表达比例在20%~80%之间,其中逆转录病毒的转染效率高于慢病毒(参见文献:Sandro等,Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies[J].Journal of Immunology Research,2018.)。然而,由于逆转录病毒和/或慢病毒本身是致病原,具有插入突变的潜力,对人类临床试验的实施构成了重大的监管障碍。不仅如此,在细胞系中大规模制造病毒载体存在成本高、效率低的问题,对嵌合受体修饰的NK细胞的临床转化构成了障碍,显著提高了生产成本。However, primary NK cells are more resistant to gene transfection, which results in a low rate of uptake by NK cells to various vector systems, low transgene expression in NK cells, and only viral vectors achieve high levels in primary NK cells gene transfer efficiency. At present, the modification of chimeric receptors in primary NK cells is generally achieved by electrotransformation of mRNA encoding genes of chimeric receptors or by lentivirus/retrovirus infection. mRNA electrotransformation can only make the gene encoding the chimeric receptor transiently expressed, and the cost of synthesizing mRNA in vitro is relatively high, requiring multiple injections in clinical practice, and the treatment cost is high. Lentiviral/retroviral transfection can produce NK cells with stable expression of chimeric receptor genes. However, compared with NK cell lines such as NK92, primary NK cells are extremely difficult to infect. So far, only a few scientific research teams have successfully applied NK cells. Virus/retrovirus prepares chimeric antigen receptor-modified primary NK cells. After repeated infection, the expression ratio of its transgene in the final product is between 20% and 80%. Among them, retrovirus transfection The efficiency is higher than that of lentivirus (see literature: Sandro et al., Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies [J]. Journal of Immunology Research, 2018.). However, because retroviruses and/or lentiviruses are themselves pathogenic and have the potential for insertional mutagenesis, they pose a significant regulatory hurdle to the implementation of human clinical trials. Not only that, the large-scale production of viral vectors in cell lines has the problems of high cost and low efficiency, which constitutes an obstacle to the clinical transformation of chimeric receptor-modified NK cells and significantly increases the production cost.
为了克服病毒载体系统的上述局限性,研究者进一步探索了非病毒载体的可行性。通常,非病毒载体系统为人工合成系统,不依赖于任何病毒成分或哺乳动物细胞进行生产制造,成本低,同时有利于避免使用病毒载体带来的免疫问题。DNA转座子是一种典型的非病毒载体系统,具有基因可持续性表达、免疫原性低、成本低、效率高等优点。To overcome the above limitations of viral vector systems, the researchers further explored the feasibility of non-viral vectors. Generally, the non-viral vector system is an artificial synthesis system, which does not depend on any viral components or mammalian cells for production, and is low in cost, and at the same time, it is beneficial to avoid the immune problems caused by the use of viral vectors. DNA transposon is a typical non-viral vector system with the advantages of sustainable gene expression, low immunogenicity, low cost and high efficiency.
在自然界中,DNA转座子是包含转座酶基因的离散DNA片段,其两侧是包含转座酶结合位点的反向末端重复序列(ITRs)。转座的过程为转座酶结合到ITRs上,从一个位置“剪切”转座,并“粘贴”到另一个新的位置。基于转座子的载体系统,如睡美人(SB)系统、PiggyBac(PB)系统、Tol转座子系统,是利用转座的方法,将转基因引入到宿主基因组。与逆转录病毒载体类似,稳定的基因组整合到宿主中可以实现长期高效的转基因表达(参见文献:Munoz-Lopez M,Garcia-Perez J L.,等DNA Transposons:Nature and Applications in Genomics[J].Current Genomics,2010.)。SB和PB转座子都已成功用于修饰T细胞,使其表达CD19特异性CAR(参见文献:Harjeet S等,Manufacture of Clinical-Grade CD19-Specific T Cells Stably Expressing Chimeric Antigen Receptor Using Sleeping Beauty System and Artificial Antigen Presenting Cells[J].Plos One,2013,8(5):e64138.;和David C.等,PiggyBac-Engineered T Cells Expressing CD19-Specific CARs that Lack IgG1 Fc Spacers Have Potent Activity against B-ALL Xenografts[J],Molecular Therapy Volume 26,Issue 8,1 August 2018,Pages 1883-1895)。然而目前尚无采用转座子系统制备转基因修饰的原代NK细胞的报道。In nature, DNA transposons are discrete DNA segments containing transposase genes flanked by inverted terminal repeats (ITRs) containing transposase binding sites. The process of transposition is that the transposase binds to the ITRs, "cuts" the transposition from one location, and "pastes" it to another new location. Transposon-based vector systems, such as Sleeping Beauty (SB) system, PiggyBac (PB) system, and Tol transposon system, use the method of transposition to introduce transgenes into the host genome. Similar to retroviral vectors, stable genome integration into the host can achieve long-term and efficient transgene expression (see references: Munoz-Lopez M, Garcia-Perez J L., et al. DNA Transposons: Nature and Applications in Genomics [J]. Current Genomics, 2010.). Both SB and PB transposons have been successfully used to modify T cells to express CD19-specific CARs (see: Harjeet S et al., Manufacture of Clinical-Grade CD19-Specific T Cells Stably Expressing Chimeric Antigen Receptor Using Sleeping Beauty System and Artificial Antigen Presenting Cells[J].Plos One,2013,8(5):e64138.; and David C. et al., PiggyBac-Engineered T Cells Expressing CD19-Specific CARs that Lack IgG1 Fc Spacers Have Potent Activity again B-ALL Xenografts [J], Molecular Therapy Volume 26, Issue 8, 1 August 2018, Pages 1883-1895). However, there is no report on the use of transposon system to prepare transgenic primary NK cells.
发明内容SUMMARY OF THE INVENTION
为了克服目前的技术手段对原代NK细胞进行嵌合受体修饰效率低的问题,以及使用逆转录病毒造成 的监管障碍、生产成本高的问题,本申请提供了一种非病毒的制备嵌合受体修饰的NK细胞方法,应用转座子系统在NK细胞中引入稳定高表达的嵌合受体,实现对NK细胞的高效遗传修饰。In order to overcome the problems of low efficiency of chimeric receptor modification of primary NK cells by current technical means, as well as the problems of regulatory barriers and high production costs caused by the use of retroviruses, the present application provides a non-viral preparation of chimeric receptors The receptor-modified NK cell method uses a transposon system to introduce stable and highly expressed chimeric receptors into NK cells to achieve efficient genetic modification of NK cells.
为达此目的,本发明采用以下技术方案:For this purpose, the present invention adopts the following technical solutions:
本发明提供一种利用非病毒转座子系统和人工抗原呈递细胞制备嵌合受体修饰的自然杀伤细胞的方法,包括:The present invention provides a method for preparing natural killer cells modified by chimeric receptors by utilizing a non-viral transposon system and artificial antigen-presenting cells, comprising:
(1)应用转座子系统将嵌合受体的编码基因转入NK细胞;(1) Using the transposon system to transfer the gene encoding the chimeric receptor into NK cells;
(2)利用人工抗原呈递细胞扩增嵌合受体修饰的NK细胞。(2) Using artificial antigen-presenting cells to expand chimeric receptor-modified NK cells.
其中,所述NK细胞是人原代NK细胞,可以来自人的外周血、脐带血、胎盘组织或诱导多能干细胞(iPSC)等。Wherein, the NK cells are primary human NK cells, which can be derived from human peripheral blood, umbilical cord blood, placental tissue or induced pluripotent stem cells (iPSCs).
所述嵌合受体包括嵌合抗原受体、嵌合转换受体、或其他应用DNA重组技术合成的嵌合受体。The chimeric receptors include chimeric antigen receptors, chimeric switching receptors, or other chimeric receptors synthesized by recombinant DNA technology.
优选地,步骤(1)所述转座子系统包括PiggyBac转座子系统、Sleeping Beauty转座子系统、Tol2转座子系统或其他应用转座酶的转座子系统。Preferably, the transposon system of step (1) includes the PiggyBac transposon system, the Sleeping Beauty transposon system, the Tol2 transposon system or other transposon systems using transposase.
优选地,步骤(1)所述转座子系统包括转座子元件和转座酶元件;所述转座子元件包括转座子5’反向末端重复序列(5’ITR)和3’反向末端重复序列(3’ITR),在5’ITR和3’ITR之间设有两个绝缘子,在两个绝缘子之间包括启动子和嵌合受体的编码基因,所述转座子可以是质粒或线性化的核酸片段;所述转座酶元件可以由质粒或线性化的核酸片段编码,也可以由mRNA编码,也可以是蛋白。Preferably, the transposon system in step (1) includes a transposon element and a transposase element; the transposon element includes a transposon 5' inverted terminal repeat (5' ITR) and a 3' inverse terminal repeat. To the terminal repeat (3'ITR), there are two insulators between the 5'ITR and the 3'ITR, and between the two insulators include a promoter and a chimeric receptor encoding gene, the transposon can Is a plasmid or a linearized nucleic acid fragment; the transposase element can be encoded by a plasmid or a linearized nucleic acid fragment, or can be encoded by mRNA, or can be a protein.
优选地,所述转座酶的核酸序列和转座子的核酸序列可以在一个载体上,也可以在两个不同的载体上。Preferably, the nucleic acid sequence of the transposase and the nucleic acid sequence of the transposon may be on one vector or on two different vectors.
优选地,所述转座子元件中可以加入促进NK细胞存活的细胞因子编码基因,包括IL-15和/或IL-21。Preferably, genes encoding cytokines that promote the survival of NK cells, including IL-15 and/or IL-21, can be added to the transposon element.
所述细胞因子可以是分泌型的,膜结合型的,或细胞因子连接受体形成的组合型;所述细胞因子的编码基因可以与嵌合受体的编码基因在同一转座子表达载体中,也可以在不同的转座子表达载体中。The cytokine can be secreted, membrane-bound, or a combination of cytokine-linked receptors; the cytokine encoding gene can be in the same transposon expression vector with the chimeric receptor encoding gene , also in different transposon expression vectors.
更优选地,所述转座子元件中加入促进NK细胞存活的IL-15编码基因,且与嵌合受体的编码基因在同一转座子表达载体中。More preferably, the IL-15 encoding gene that promotes the survival of NK cells is added to the transposon element, and is in the same transposon expression vector as the encoding gene of the chimeric receptor.
优选地,所述应用转座子系统将嵌合受体的编码基因转入NK细胞的方法包括:电穿孔的方法、化学试剂转染的方法、以及其他非病毒转染的方法等。Preferably, the method for using the transposon system to transfer the encoding gene of the chimeric receptor into NK cells includes electroporation, chemical transfection, and other non-viral transfection methods.
更优选地,通过电穿孔的方法将转座子系统的相关载体转入原代NK细胞中。More preferably, the relevant vectors of the transposon system are transferred into primary NK cells by electroporation.
优选地,步骤(1)所述应用转座子系统将转基因转入NK细胞前,NK细胞可以预先进行活化,也可以预先不进行活化。Preferably, before the transposon system is used to transfer the transgene into the NK cells in step (1), the NK cells may or may not be activated in advance.
更优选地,步骤(1)中使用所述转座子系统将转基因转入NK细胞前,预先对NK细胞进行活化。More preferably, before using the transposon system to transfer the transgene into the NK cells in step (1), the NK cells are activated in advance.
优选地,若NK细胞预先进行活化,可以在NK细胞活化后的0~14天,例如1、2、3、4、5、6、7、8、9、10、11、12、13或14天,应用转座子系统将嵌合受体的编码基因转入NK细胞中。Preferably, if the NK cells are activated in advance, it can be 0 to 14 days after the activation of the NK cells, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14. Today, the gene encoding the chimeric receptor was transferred into NK cells using the transposon system.
更优选地,在NK细胞活化后1~7天,例如1、2、3、4、5、6或7天,应用转座子系统将嵌合受体的编码基因转入NK细胞中。More preferably, 1 to 7 days, eg, 1, 2, 3, 4, 5, 6 or 7 days after the activation of the NK cells, the gene encoding the chimeric receptor is transferred into the NK cells using a transposon system.
更优选地,在NK细胞活化后2天,应用转座子系统将嵌合受体的编码基因转入NK细胞中。More preferably, 2 days after the activation of the NK cells, the gene encoding the chimeric receptor is transferred into the NK cells using a transposon system.
优选地,若NK细胞预先进行活化,可以采用人工抗原呈递细胞、饲养细胞、细胞因子、抗体或化合物等进行活化。Preferably, if NK cells are activated in advance, artificial antigen-presenting cells, feeder cells, cytokines, antibodies or compounds can be used for activation.
本发明中,所述人工抗原呈递细胞包括以细胞为基础的、人工合成的或以外泌体为基础的人工抗原呈递细胞。In the present invention, the artificial antigen-presenting cells include cell-based, artificially synthesized or exosome-based artificial antigen-presenting cells.
所述以细胞为基础的人工抗原呈递细胞选自人髓系白血病K562细胞、人伯基特淋巴瘤Daudi细胞、EBV转化的B淋巴母细胞样细胞(EBV-LCL)细胞或小鼠胚胎成纤维细胞系NIH/3T3细胞。The cell-based artificial antigen presenting cells are selected from human myeloid leukemia K562 cells, human Burkitt lymphoma Daudi cells, EBV transformed B lymphoblastoid cells (EBV-LCL) cells or mouse embryonic fibroblasts Cell line NIH/3T3 cells.
优选地,所述以细胞为基础的人工抗原呈递细胞选自K562细胞。Preferably, the cell-based artificial antigen presenting cells are selected from K562 cells.
优选地,所述以细胞为基础的人工抗原呈递细胞为工程化的细胞,所述工程化细胞表达嵌合受体识别的抗原。Preferably, the cell-based artificial antigen presenting cell is an engineered cell that expresses an antigen recognized by a chimeric receptor.
优选地,所述工程化细胞表达NK细胞活化所需的配体分子、细胞因子等,其中,细胞因子可以是分泌型的,也可以是膜结合型的。Preferably, the engineered cells express ligand molecules, cytokines, etc. required for NK cell activation, wherein the cytokines may be secreted or membrane-bound.
更优选地,NK细胞活化所需配体分子包括CD137L和/或OX40L,NK活化所需细胞因子包括人IL-12、人IL-15、人IL-18或人IL-21中的任意一种或至少两种的组合。More preferably, the ligand molecules required for NK cell activation include CD137L and/or OX40L, and the cytokines required for NK activation include any one of human IL-12, human IL-15, human IL-18 or human IL-21 or a combination of at least two.
优选地,所述以细胞为基础的人工抗原呈递细胞和饲养细胞经γ射线(100Gy)或丝裂霉素C(20μg/mL)处理。Preferably, the cell-based artificial antigen presenting cells and feeder cells are treated with gamma radiation (100 Gy) or mitomycin C (20 μg/mL).
更优选地,NK细胞活化和CAR-NK细胞扩增都可以使用人工抗原呈递细胞进行。More preferably, both NK cell activation and CAR-NK cell expansion can be performed using artificial antigen presenting cells.
优选地,步骤(1)所述在应用转座子系统将嵌合受体的编码基因转入NK细胞后的0~14天,例如1、2、3、4、5、6、7、8、9、10、11、12、13或14天,利用人工抗原呈递细胞刺激嵌合受体修饰的NK细胞增殖。Preferably, in step (1), 0 to 14 days after the gene encoding the chimeric receptor is transferred into NK cells using the transposon system, for example, 1, 2, 3, 4, 5, 6, 7, 8 , 9, 10, 11, 12, 13, or 14 days, using artificial antigen-presenting cells to stimulate the proliferation of chimeric receptor-modified NK cells.
优选地,步骤(2)所述嵌合受体修饰的NK细胞扩增多次,每一轮扩增周期为3~14天,例如3、4、5、6、7、8、9、10、11、12、13或14天。Preferably, the chimeric receptor-modified NK cells in step (2) are expanded multiple times, and each expansion cycle is 3 to 14 days, such as 3, 4, 5, 6, 7, 8, 9, 10 , 11, 12, 13 or 14 days.
优选地,本发明所述方法中可以在步骤(1)前利用磁分选富集NK细胞或去除CD3+细胞;也可以在步骤(1)之后的0~14天,利用磁分选富集NK细胞或去除CD3+细胞;也可以在步骤(2)后利用磁分选富集NK细胞或去除CD3+细胞。Preferably, in the method of the present invention, magnetic sorting can be used to enrich NK cells or CD3+ cells can be removed before step (1); or 0 to 14 days after step (1), magnetic sorting can be used to enrich NK cells cells or remove CD3+ cells; magnetic sorting can also be used to enrich NK cells or remove CD3+ cells after step (2).
优选地,在步骤(1)之后,所述方法还包括CAR+细胞的分选的步骤。Preferably, after step (1), the method further comprises the step of sorting CAR+ cells.
优选地,在步骤(2)之后,所述方法还包括CAR+细胞的分选的步骤。Preferably, after step (2), the method further includes the step of sorting CAR+ cells.
更优选地,在步骤(2)之后,即与人工抗原呈递细胞共培养一轮后,进行CAR+细胞的分选。More preferably, after step (2), that is, after one round of co-culture with artificial antigen-presenting cells, sorting of CAR+ cells is performed.
优选地,在应用转座子系统制备嵌合受体修饰的NK细胞过程中,可以添加细胞因子组合物,例如hrIL-12和hrIL-18的组合或hrIL-12、hrIL-15和hrIL-18的组合。Preferably, a cytokine composition, such as a combination of hrIL-12 and hrIL-18 or a combination of hrIL-12, hrIL-15 and hrIL-18, can be added during the preparation of chimeric receptor-modified NK cells using the transposon system The combination.
优选地,所述细胞因子组合物可以在嵌合受体修饰的NK细胞制备过程中的任意一天添加。Preferably, the cytokine composition can be added on any day during the preparation of the chimeric receptor-modified NK cells.
更优选地,所述细胞因子组合物可以在嵌合受体修饰的NK细胞制备过程中添加一天后通过离心去除。More preferably, the cytokine composition can be removed by centrifugation one day after being added during the production of chimeric receptor-modified NK cells.
更优选地,所述hrIL-12的终浓度为1~100ng/mL,hrIL-15的终浓度为1~100ng/mL,hrIL-18的终浓度为1~100ng/mL,所述hrIL-12、hrIL-15或hrIL-18的终浓度相同或者不同,例如可以是1ng/mL、10ng/mL、20ng/mL、30ng/mL、40ng/mL、50ng/mL、60ng/mL、70ng/mL、80ng/mL、90ng/mL或100ng/mL。More preferably, the final concentration of hrIL-12 is 1-100 ng/mL, the final concentration of hrIL-15 is 1-100 ng/mL, the final concentration of hrIL-18 is 1-100 ng/mL, and the final concentration of hrIL-12 is 1-100 ng/mL. , the final concentration of hrIL-15 or hrIL-18 is the same or different, for example, it can be 1ng/mL, 10ng/mL, 20ng/mL, 30ng/mL, 40ng/mL, 50ng/mL, 60ng/mL, 70ng/mL, 80ng/mL, 90ng/mL or 100ng/mL.
本申请中,嵌合受体修饰的NK细胞培养采用液体培养基进行,所述液体培养基可以是本领域常用的细胞培养基,例如可以是AIM V TM(Gibco)、X-VIVO TM 15(Lonza)、SCGM TM(CellGenix)、RPMI、NK MACS TM(Miltenyi)、OpTmizer TM(Gibco)、ImmunoCult TM-XF T Cell Expansion Medium(STEMCELL)或StemSpan TM(STEMCELL)。 In this application, the chimeric receptor-modified NK cell culture is carried out using a liquid medium, and the liquid medium can be a cell culture medium commonly used in the art, such as AIM V (Gibco), X-VIVO 15 ( Lonza), SCGM (CellGenix), RPMI, NK MACS (Miltenyi), OpTmizer (Gibco), ImmunoCult -XF T Cell Expansion Medium (STEMCELL) or StemSpan (STEMCELL).
作为优选技术方案,本申请提供了一种应用PiggyBac转座子系统制备嵌合受体修饰的NK细胞的方法,所述方法包括:As a preferred technical solution, the present application provides a method for preparing chimeric receptor-modified NK cells using the PiggyBac transposon system, the method comprising:
(1)将分离的外周血单个核细胞(PBMC)与经γ射线处理的工程化K562细胞共培养;(1) Co-culturing the isolated peripheral blood mononuclear cells (PBMCs) with engineered K562 cells treated with γ-rays;
(2)2天后,利用Lonza 4D-Nucleofector device,将编码PiggyBac(PB)转座子系统的辅助质粒和编码嵌合受体的供体质粒电转至NK细胞中;(2) After 2 days, using the Lonza 4D-Nucleofector device, the helper plasmid encoding the PiggyBac (PB) transposon system and the donor plasmid encoding the chimeric acceptor were electroporated into NK cells;
(3)电转5~9天(例如5、6、7、8或9天)后,将嵌合受体修饰的NK细胞与经γ射线处理的工程化K562细胞共培养,培养起始时添加hrIL-12、hrIL-15和hrIL-18;(3) After electroporation for 5 to 9 days (eg, 5, 6, 7, 8, or 9 days), the chimeric receptor-modified NK cells were co-cultured with γ-ray-treated engineered K562 cells, and added at the beginning of the culture. hrIL-12, hrIL-15 and hrIL-18;
其中,所述细胞培养液为添加有1~5%人AB血清/人自体血浆的AIM V培养液,在培养过程中存在hrIL-2。Wherein, the cell culture medium is AIMV medium supplemented with 1-5% human AB serum/human autologous plasma, and hrIL-2 is present during the culture.
进一步优选地,本发明所述应用PiggyBac转座子系统制备嵌合受体修饰的NK细胞的方法为:Further preferably, the method for preparing chimeric receptor-modified NK cells using the PiggyBac transposon system according to the present invention is:
(1)利用CD3磁珠将单个核细胞(MNC)中的CD3+细胞去除;(1) Use CD3 magnetic beads to remove CD3+ cells from mononuclear cells (MNCs);
(2)将分选后的单个核细胞(MNC)与经γ射线处理的工程化K562细胞共培养;(2) co-culturing the sorted mononuclear cells (MNCs) with engineered K562 cells treated with γ-rays;
(3)2天后,利用Lonza 4D-Nucleofector device,将编码PiggyBac(PB)转座子系统的辅助质粒和编码嵌合受体的供体质粒电转至NK细胞中;(3) After 2 days, using the Lonza 4D-Nucleofector device, the helper plasmid encoding the PiggyBac (PB) transposon system and the donor plasmid encoding the chimeric acceptor were electroporated into NK cells;
(3)电转5~9天后,对CAR+细胞进行分选;(3) 5-9 days after electroporation, CAR+ cells were sorted;
(4)将分选后的嵌合受体修饰的NK细胞与经γ射线处理的工程化K562细胞共培养,培养起始时添加hrIL-12、hrIL-15和hrIL-18;(4) Co-culturing the sorted chimeric receptor-modified NK cells with γ-ray-treated engineered K562 cells, adding hrIL-12, hrIL-15 and hrIL-18 at the beginning of the culture;
其中,所述细胞培养液为添加有1~5%人AB血清/人自体血浆的AIM V培养液,在培养过程中存在hrIL-2。Wherein, the cell culture medium is AIMV medium supplemented with 1-5% human AB serum/human autologous plasma, and hrIL-2 is present during the culture.
与现有技术相比,本申请具有如下有益效果:Compared with the prior art, the present application has the following beneficial effects:
(1)目前对原代NK细胞进行嵌合受体的修饰主要依赖于慢病毒或逆转录病毒介导的基因整合或mRNA电穿孔介导的基因瞬时表达,由于病毒载体制备过程复杂、制备和使用过程中存在安全隐患、病毒质量参差不齐,mRNA瞬时表达、制备成本高,阻碍了嵌合受体修饰的NK细胞在肿瘤治疗中的临床探索,本申请提出应用转座子系统和人工抗原呈递细胞来制备嵌合受体修饰的原代NK细胞,其中转座子系统和人工抗原呈递细胞的结合是必须的,而且本申请还发现并提出利用转座子系统将目标基因转入NK细胞的时间点对于NK细胞的扩增效率、纯度、嵌合受体的阳性率非常重要,本申请通过理论和实验研究,优化了各个步骤的顺序、时间点、所用试剂的浓度或共培养的比例等,显著提升了嵌合受体在NK细胞中的阳性率、NK的扩增倍数、纯度和肿瘤杀伤能力等;(1) At present, the modification of chimeric receptors in primary NK cells mainly relies on lentivirus or retrovirus-mediated gene integration or mRNA electroporation-mediated gene transient expression. During use, there are potential safety hazards, uneven virus quality, transient mRNA expression, and high production costs, which hinder the clinical exploration of chimeric receptor-modified NK cells in tumor treatment. This application proposes the application of transposon systems and artificial antigens. Presenting cells to prepare chimeric receptor-modified primary NK cells, in which the combination of transposon system and artificial antigen presenting cells is necessary, and this application also discovers and proposes the use of transposon system to transfer target genes into NK cells The time point is very important for the expansion efficiency, purity, and positive rate of chimeric receptors of NK cells. Through theoretical and experimental research, this application optimizes the sequence of each step, time point, concentration of reagents used or the ratio of co-culture. Etc., significantly improved the positive rate of chimeric receptors in NK cells, the amplification fold, purity and tumor killing ability of NK cells;
(2)目前在制备嵌合受体修饰的NK细胞的过程中,目的基因主要依赖于慢病毒载体或逆转录病毒载体进行递送,然而慢病毒或逆转录病毒生产过程复杂、成本高,极大地增加了基因修饰的NK细胞的生产成本,昂贵的价格使得很多肿瘤病人无法承担,因此降低嵌合受体修饰的免疫细胞生产成本的需求显得格外迫切,本申请应用转座子系统制备嵌合受体修饰的原代NK细胞,转座子系统只需要GMP级别的质粒即可,原材料制备过程简单,大大降低了生产成本;(2) Currently, in the process of preparing chimeric receptor-modified NK cells, the target gene mainly relies on lentiviral vectors or retroviral vectors for delivery. The production cost of gene-modified NK cells is increased, and the high price makes many tumor patients unaffordable. Therefore, the need to reduce the production cost of chimeric receptor-modified immune cells is particularly urgent. This application uses a transposon system to prepare chimeric receptors. Body-modified primary NK cells, the transposon system only needs GMP-grade plasmids, the raw material preparation process is simple, and the production cost is greatly reduced;
(3)慢病毒载体或逆转录病毒载体的使用给操作人员也带来了一定的安全隐患,不仅如此,在使用慢病毒载体或逆转率病毒载体制备嵌合受体修饰的NK细胞的同时,也存在NK细胞产品被复制型逆转录病毒(RCR)或复制型慢病毒(RCL)污染的潜在风险,虽然病毒载体设计及生产体系在不断改进,这种风险已大大降低,但仍不能完全排除,本申请使用的转座子系统是一种非病毒的方法,十分安全,不会存在这些隐患;(3) The use of lentiviral vectors or retroviral vectors also brings certain safety hazards to operators. Not only that, when using lentiviral vectors or retroviral vectors to prepare chimeric receptor-modified NK cells, There is also a potential risk of contamination of NK cell products by replicating retrovirus (RCR) or replicating lentivirus (RCL). Although the viral vector design and production system are constantly improving, this risk has been greatly reduced, but it cannot be completely ruled out. , the transposon system used in this application is a non-viral method, which is very safe and will not have these hidden dangers;
(4)应用本申请的方法,嵌合受体的编码基因在NK细胞中的整合效率在40~80%之间,已达到现有技术采用逆转录病毒修饰原代NK细胞的同等水平。(4) By applying the method of the present application, the integration efficiency of the gene encoding the chimeric receptor in NK cells is between 40% and 80%, which has reached the same level as that of using retrovirus to modify primary NK cells in the prior art.
附图说明Description of drawings
图1A为质粒2的图谱,图1B为质粒4的图谱;FIG. 1A is the map of plasmid 2, and FIG. 1B is the map of plasmid 4;
图2A为K562-NK1细胞表面的膜结合的人白介素15的表达,图2B为K562-NK1细胞表面的膜结合的白介素21的表达,横坐标为膜结合的人白介素IL-15或IL-21的表达强度,纵坐标为细胞计数;Figure 2A is the expression of membrane-bound human interleukin 15 on the surface of K562-NK1 cells, Figure 2B is the expression of membrane-bound human interleukin 21 on the surface of K562-NK1 cells, and the abscissa is the membrane-bound human interleukin IL-15 or IL-21 The expression intensity of , the ordinate is the cell count;
图3为利用PiggyBac转座子系统制备CAR-NK的流程图;Fig. 3 is the flow chart that utilizes PiggyBac transposon system to prepare CAR-NK;
图4A为不同CAR-NK制备方法在培养第10、17、24天时,NK细胞在细胞群体中的比例,图4B为不同CAR-NK制备方法在培养第10、17、24天时,CAR+细胞在NK细胞中的比例;Figure 4A shows the proportion of NK cells in the cell population on the 10th, 17th, and 24th days of culture for different CAR-NK preparation methods, and Figure 4B shows the proportion of CAR+ cells in the cell population for different CAR-NK preparation methods on the 10th, 17th, and 24th days of culture. the proportion of NK cells;
图5为不同CAR-NK制备方法在培养第10、17、24天时,总细胞数的累积扩增倍数;Figure 5 shows the cumulative expansion fold of the total cell number on the 10th, 17th, and 24th days of culture for different CAR-NK preparation methods;
图6A为不同来源的单个核细胞(MNC)在第0天,第1、2、3、4轮共培养结束时,NK细胞在细 胞群体中的比例,图6B为不同来源的MNC在第1、2、3、4轮共培养结束时,CAR+细胞在NK细胞中的比例;Figure 6A shows the proportion of NK cells in the cell population of mononuclear cells (MNCs) from different sources at the end of the 1st, 2nd, 3rd, and 4th rounds of co-culture on day 0, and Figure 6B shows the ratios of MNCs from different sources in the first At the end of 2, 3, and 4 rounds of co-culture, the proportion of CAR+ cells in NK cells;
图7为不同来源的MNC在第1、2、3、4轮共培养结束时,总细胞数的累积扩增倍数;Figure 7 is the cumulative expansion fold of the total cell number of MNCs from different sources at the end of the first, second, third, and fourth rounds of co-culture;
图8A为第三轮共培养结束第21天的NK和NKG2D CAR-NK细胞对人结直肠癌细胞株HCT116的实时杀伤图谱;图8B为第三轮共培养结束第21天的NK和NKG2D CAR-NK细胞对人胃癌细胞株MGC803的实时杀伤图谱;其中,横坐标为肿瘤细胞铺板后(即实验开始后)的时间,以小时计,纵坐标为细胞指数;Figure 8A shows the real-time killing map of NK and NKG2D CAR-NK cells on the human colorectal cancer cell line HCT116 on the 21st day after the third round of co-culture; Figure 8B shows the NK and NKG2D CAR on the 21st day after the third round of co-culture - The real-time killing map of NK cells to human gastric cancer cell line MGC803; wherein, the abscissa is the time after tumor cells are plated (that is, after the experiment starts), in hours, and the ordinate is the cell index;
图9为第四轮共培养结束第28天的NK和NKG2D CAR-NK细胞与人结直肠癌细胞株HCT116、人卵巢癌细胞株SKOV3、人胃癌细胞株MGC803共培养过夜后(E:T=10:1)IFNγ的分泌;其中,纵坐标为共培养上清液中IFNγ的浓度(pg/mL),*:P<0.05,**:P<0.01;Figure 9 shows the overnight co-culture of NK and NKG2D CAR-NK cells with human colorectal cancer cell line HCT116, human ovarian cancer cell line SKOV3, and human gastric cancer cell line MGC803 on the 28th day after the fourth round of co-culture (E:T= 10:1) Secretion of IFNγ; wherein, the ordinate is the concentration of IFNγ in the co-culture supernatant (pg/mL), *: P<0.05, **: P<0.01;
图10A为pmax-GFP质粒电转1天后GFP在NK细胞中的表达,使用的NK细胞是扩增第17天的NK细胞,横坐标为GFP的表达强度,纵坐标为细胞计数,图10B为pmax-GFP质粒电转前和电转1天后NK细胞的数目,使用的NK细胞是扩增第17天的NK细胞,横坐标分别为电转前和电转1天后,纵坐标为细胞数目;Figure 10A shows the expression of GFP in NK cells 1 day after the pmax-GFP plasmid was electroporated. The NK cells used were NK cells on the 17th day of expansion. The abscissa is the expression intensity of GFP, the ordinate is the cell count, and Figure 10B is pmax -Number of NK cells before and after 1 day of electroporation of GFP plasmid, the NK cells used are NK cells on the 17th day of expansion, the abscissa is before and 1 day after electroporation, respectively, and the ordinate is the number of cells;
图11A为不同CAR-NK制备方法在培养第10、17、24、31天时,NK细胞在细胞群体中的比例,图11B为不同CAR-NK制备方法在培养第10、17、24、31天时,CAR+细胞在NK细胞中的比例;Figure 11A shows the proportion of NK cells in the cell population on the 10th, 17th, 24th and 31st days of culture by different CAR-NK preparation methods, and Figure 11B shows the ratio of NK cells in the cell population by different CAR-NK preparation methods on the 10th, 17th, 24th and 31st days of culture , the proportion of CAR+ cells in NK cells;
图12为不同CAR-NK制备方法在培养第10、17、24、31天时,总细胞数的累积扩增倍数;Figure 12 shows the cumulative expansion fold of the total cell number on the 10th, 17th, 24th, and 31st days of culture for different CAR-NK preparation methods;
图13A为第17天的NK、CAR-NK和eCAR-NK细胞对人卵巢癌细胞株SKOV3的实时杀伤图谱,横坐标为肿瘤细胞铺板后(即实验开始后)的时间,以小时计,纵坐标为细胞指数,图13B为在加入效应细胞24小时后,进行数据分析,计算肿瘤生长抑制率,横坐标为不同实验组,纵坐标为肿瘤生长抑制率;Figure 13A is the real-time killing map of human ovarian cancer cell line SKOV3 by NK, CAR-NK and eCAR-NK cells on the 17th day. The coordinate is the cell index, and Figure 13B shows the data analysis after adding effector cells for 24 hours to calculate the tumor growth inhibition rate, the abscissa is the different experimental groups, and the ordinate is the tumor growth inhibition rate;
图14A为检测培养第22天的NK、NKG2D CAR-NK和NKG2D eCAR-15NK在有IL-2或无IL-2的条件下体外培养一周的存活情况;图14B为检测培养第21天的NK、NKG2D eCAR-15NK和NKG2D eCAR-15 NK(sorted)在有IL-2或无IL-2的条件下体外培养一周的存活情况;图14C为检测培养第22天的NK、NKG2D eCAR-NK(sorted)和NKG2D eCAR-mbIL15 NK(sorted)在无IL-2的条件下体外培养一周的存活情况;图14D为检测培养第16天的NK、NKG2D CAR-mbIL15 NK和NKG2D eCAR-mbIL15 NK(sorted)在有IL-2或无IL-2的条件下体外培养一周的存活情况;其中,横坐标中“-IL2”为不添加IL-2培养组,“+IL2”为添加IL-2培养组,纵坐标为相对于培养一周前细胞数量的变化百分比;Figure 14A is the detection of the survival of NK, NKG2D CAR-NK and NKG2D eCAR-15NK on the 22nd day of culture in the presence or absence of IL-2 for one week in vitro; Figure 14B is the detection of the NK on the 21st day of culture , NKG2D eCAR-15NK and NKG2D eCAR-15 NK(sorted) were cultured in vitro for one week in the presence or absence of IL-2; Figure 14C is the detection of NK, NKG2D eCAR-NK ( sorted) and NKG2D eCAR-mbIL15 NK(sorted) in vitro cultured without IL-2 for one week; ) Survival of one week in vitro culture with or without IL-2; among them, "-IL2" in the abscissa is the culture group without IL-2, and "+IL2" is the culture group with IL-2 added , the ordinate is the percentage change relative to the number of cells before one week of culture;
图15A为第三轮共培养结束第24天的NK、NKG2D CAR-mbIL15 NK、NKG2D eCAR-mbIL15 NK(sorted)对人急性髓性白血病细胞株KG1的杀伤结果,横坐标为效靶比(E:T),纵坐标杀伤百分比;图15B为第三轮共培养结束第24天的NK、NKG2D eCAR-mbIL15 NK(sorted)对人结直肠癌细胞株HCT116 的杀伤结果,横坐标为肿瘤细胞铺板后(即实验开始后)的时间,以小时计,纵坐标为细胞指数;Figure 15A shows the killing results of NK, NKG2D CAR-mbIL15 NK, and NKG2D eCAR-mbIL15 NK (sorted) on the 24th day after the third round of co-culture on human acute myeloid leukemia cell line KG1, the abscissa is the effect-target ratio (E : T), the killing percentage on the ordinate; Figure 15B is the killing result of NK and NKG2D eCAR-mbIL15 NK (sorted) on the human colorectal cancer cell line HCT116 on the 24th day after the third round of co-culture, the abscissa is the tumor cell plating The time after (that is, after the start of the experiment), in hours, the ordinate is the cell index;
图16为K562-NK2细胞表面BCMA的表达;Figure 16 is the expression of BCMA on the surface of K562-NK2 cells;
图17A为一个健康捐献者来源的外周血单个核细胞(PBMC)在第0天,第1、2、3轮共培养结束时,NK细胞在细胞群体中的比例;图17B为一个健康捐献者来源的PBMC在第1、2、3轮共培养结束时,CAR+细胞在NK细胞中的比例;Figure 17A shows the proportion of NK cells in the cell population of peripheral blood mononuclear cells (PBMC) derived from a healthy donor at the end of the 1st, 2nd and 3rd rounds of co-culture on day 0; Figure 17B shows a healthy donor The proportion of CAR+ cells in NK cells at the end of the 1st, 2nd and 3rd rounds of co-culture of the derived PBMC;
图18为一个健康捐献者来源的PBMC在第1、2、3轮共培养结束时,每一轮总细胞数的扩增倍数;Figure 18 is the expansion fold of the total cell number in each round of PBMC derived from a healthy donor at the end of the 1st, 2nd, and 3rd rounds of co-culture;
图19为共培养第五轮结束时第36天的NK和BCMA eCAR-NK对人多发性骨髓瘤细胞株U266的杀伤结果,其中,横坐标为效靶比(E:T),纵坐标杀伤百分比。Figure 19 shows the killing results of human multiple myeloma cell line U266 by NK and BCMA eCAR-NK on the 36th day at the end of the fifth round of co-culture, wherein the abscissa is the effect-target ratio (E:T), and the ordinate is the killing percentage.
具体实施方式detailed description
为进一步阐述本申请所采取的技术手段及其效果,以下结合实施例和附图对本申请作进一步地说明。可以理解的是,此处所描述的具体实施方式仅仅用于解释本申请,而非对本申请的限定。In order to further illustrate the technical means adopted in the present application and its effects, the present application will be further described below with reference to the embodiments and the accompanying drawings. It should be understood that the specific embodiments described herein are only used to explain the present application, but not to limit the present application.
实施例中未注明具体技术或条件者,按照本领域内的文献所描述的技术或条件,或者按照产品说明书进行。所用试剂或仪器未注明生产厂商者,均为可通过正规渠道商购获得的常规产品。If no specific technique or condition is indicated in the examples, the technique or condition described in the literature in the field or the product specification is used. The reagents or instruments used without the manufacturer's indication are conventional products that can be purchased through regular channels.
除非特别说明,否则细胞培养在37℃、5%CO 2、潮化(相对湿度为95%)的条件下。 Unless otherwise stated, cells were cultured at 37 ℃, 5% CO 2, of the tide (95% relative humidity) conditions.
实施例中采用的实验材料如下:The experimental materials used in the examples are as follows:
PBMC提取自实体肿瘤患者或健康捐献者的外周血;脐带血来源的MNC购自迈顺生物科技有限公司;人慢性髓系白血病细胞株野生型K562细胞购自ATCC,人多发性骨髓瘤细胞株U266购自武汉普诺赛生命科技有限公司;其余未提及的试剂或耗材等均来自本领域常规试剂厂商或提供本领域常规手段制备;PBMC were extracted from peripheral blood of solid tumor patients or healthy donors; MNC derived from umbilical cord blood was purchased from Maishun Biotechnology Co., Ltd.; human chronic myeloid leukemia cell line wild-type K562 cells were purchased from ATCC, human multiple myeloma cell line U266 was purchased from Wuhan Proceeds Life Technology Co., Ltd.; the other reagents or consumables not mentioned were all from conventional reagent manufacturers in the field or prepared by conventional means in the field;
人卵巢癌细胞株SKOV3和人结直肠癌细胞株HCT116的培养基为McCoy 5A(Gibco)+10%FBS(Gibco),人胃癌细胞株MGC803和U266的培养基为RPMI(Gibco)+10%FBS(Gibco),K562和人AML细胞株KG1的培养基为IMDM(Gibco)+10%FBS(Gibco);The medium of human ovarian cancer cell line SKOV3 and human colorectal cancer cell line HCT116 is McCoy 5A (Gibco) + 10% FBS (Gibco), and the medium of human gastric cancer cell lines MGC803 and U266 is RPMI (Gibco) + 10% FBS (Gibco), the medium of K562 and human AML cell line KG1 is IMDM (Gibco) + 10% FBS (Gibco);
实施例中采用的K562-NK1细胞为经γ射线处理(100Gy)的基因工程改造的编码膜结合的人IL-15(mbIL15)、膜结合的人IL-21(mbIL21)、eGFP和Puromycin的K562细胞。由于K562细胞自然表达NKG2D配体,因此这里不再额外把NKG2D CAR识别的抗原装入K562-NK1细胞中。膜结合的人IL-15由GM-CSF Rα信号肽(UniprotKB P15509第1~22位氨基酸)、人IL-15(UniprotKB P40933第30~162位氨基酸)和人CD8的铰链跨膜区(UniprotKB P01732第128~213位氨基酸)融合在一起;膜结合的人IL-21是由GM-CSF Rα信号肽(UniprotKB P15509第1~22位氨基酸)、人IL-21(UniprotKB Q9HBE4第25~162位氨基酸)、人IGHG4的铰链恒定区(UniprotKB P01861第99~327位氨基酸)、人CD4的跨膜区(UniprotKB P01730第397~418位氨基酸)融合在一起,eGFP的氨基酸序列如SEQ ID No:3所示,Puromycin的氨基酸序列如SEQ ID No:4所示;The K562-NK1 cells used in the examples are γ-ray-treated (100 Gy) genetically engineered K562 encoding membrane-bound human IL-15 (mbIL15), membrane-bound human IL-21 (mbIL21), eGFP and Puromycin cell. Since K562 cells naturally express NKG2D ligands, the antigens recognized by NKG2D CAR are not additionally loaded into K562-NK1 cells here. Membrane-bound human IL-15 is composed of the GM-CSF Ra signal peptide (amino acids 1-22 of UniprotKB P15509), human IL-15 (amino acids 30-162 of UniprotKB P40933) and the hinge transmembrane region of human CD8 (UniprotKB P01732 Amino acids 128-213) are fused together; membrane-bound human IL-21 is composed of GM-CSF Rα signal peptide (amino acids 1-22 of UniprotKB P15509), human IL-21 (amino acids 25-162 of UniprotKB Q9HBE4) ), the hinge constant region of human IGHG4 (the 99th to 327th amino acids of UniprotKB P01861), the transmembrane region of human CD4 (the 397th to 418th amino acids of UniprotKB P01730) are fused together, and the amino acid sequence of eGFP is as shown in SEQ ID No:3 Shown, the amino acid sequence of Puromycin is shown in SEQ ID No:4;
Figure PCTCN2021103235-appb-000001
Figure PCTCN2021103235-appb-000001
实施例中采用的K562-NK2细胞为经γ射线处理(100Gy)的基因工程改造的编码膜结合的人IL-15(mbIL15)、膜结合的人IL-21(mbIL21)、短截的BCMA(tBCMA)、eGFP和Puromycin的K562细胞。tBCMA的序列为UniProt KB-Q02223第1-77位的氨基酸序列。The K562-NK2 cells used in the examples were genetically engineered to encode membrane-bound human IL-15 (mbIL15), membrane-bound human IL-21 (mbIL21), truncated BCMA ( tBCMA), eGFP and Puromycin K562 cells. The sequence of tBCMA is the amino acid sequence of positions 1-77 of UniProt KB-Q02223.
人重组IL-2(hrIL2)购自北京双鹭公司,人重组IL-12(hrIL12)、人重组IL-15(hrIL15)、人重组IL-18(hrIL18)购自近岸蛋白公司;Human recombinant IL-2 (hrIL2) was purchased from Beijing Shuanglu Company, and human recombinant IL-12 (hrIL12), human recombinant IL-15 (hrIL15), and human recombinant IL-18 (hrIL18) were purchased from Nearshore Protein Company;
NK细胞培养液:AIM培养基(Gibco公司)+5%人类AB血清(Gemini公司);NK cell culture medium: AIM medium (Gibco company) + 5% human AB serum (Gemini company);
P3 Primary Cell 4D-Nucleofector X Kit购自Lonza公司;P3 Primary Cell 4D-Nucleofector X Kit was purchased from Lonza Company;
PE-结合的抗人CD3抗体、APC-结合的抗人CD56抗体购自BD公司,FITC-结合的抗Strep-tag II抗体购自金斯瑞公司,Biotin结合的抗人IL-15抗体和APC结合的Streptavidin购自Biolegend公司,Biotin结合的抗人IL-21抗体购自eBioscience公司,anti-Biotin microbeads和Streptavidin micobeads购自Miltenyi公司;PE-conjugated anti-human CD3 antibody, APC-conjugated anti-human CD56 antibody were purchased from BD, FITC-conjugated anti-Strep-tag II antibody was purchased from GenScript, Biotin-conjugated anti-human IL-15 antibody and APC Conjugated Streptavidin was purchased from Biolegend Company, Biotin-conjugated anti-human IL-21 antibody was purchased from eBioscience Company, anti-Biotin microbeads and Streptavidin micobeads were purchased from Miltenyi Company;
流式细胞仪购自BD公司,型号为C6 Sampler;实时杀伤检测仪购自ACEA Bio公司,型号为xCELLigence RTCA DP;人IFNγELISA检测试剂盒购自Biolegend公司;Calcein-AM购自翌圣生物科技;pmax-GFP购自Lonza公司;Flow cytometer was purchased from BD Company, model C6 Sampler; real-time killing detector was purchased from ACEA Bio Company, model xCELLigence RTCA DP; human IFNγ ELISA detection kit was purchased from Biolegend Company; Calcein-AM was purchased from Yisheng Biotechnology; pmax-GFP was purchased from Lonza Company;
PiggyBac转座酶的氨基酸序列来自GenBank:AAA87375.2;NKG2D CAR为:NKG2D-CD28-4-1BB-CD3ζ,抗原结合结构域来自NKG2D(UniProtKB-P26718)的第83~216位氨基酸、铰链区来自IgG4(UniProtKB-P01861)的第99~110位氨基酸、跨膜区来自CD28(UniProtKB-P10747)的第153~179位氨基酸,胞内结构域来自4-1BB(UniProtKB-Q07011)的第209~255位氨基酸和CD3ζ(UniProtKB-P20963)的第52~164位氨基酸;BCMA CAR为:anti-BCMA ScFv-CD28-4-1BB-DAP12ζ,抗原结合结构域来自Anti-BCMA抗体C11D5.3的scFv、铰链区来自IGHG4(UniProtKB-P01861)的第99~110位氨基酸、跨膜区来自CD28(UniProtKB-P10747)的第153~179位氨基酸,胞内结构域来自4-1BB (UniProtKB-Q07011)的第209~255位氨基酸和DAP12(UniprotKB-O43914)的第62~133位氨基酸。The amino acid sequence of PiggyBac transposase is from GenBank: AAA87375.2; the NKG2D CAR is: NKG2D-CD28-4-1BB-CD3ζ, the antigen-binding domain is from amino acids 83-216 of NKG2D (UniProtKB-P26718), and the hinge region is from Amino acids 99-110 of IgG4 (UniProtKB-P01861), transmembrane region from amino acids 153-179 of CD28 (UniProtKB-P10747), intracellular domain from 209-255 of 4-1BB (UniProtKB-Q07011) BCMA CAR is: anti-BCMA ScFv-CD28-4-1BB-DAP12ζ, the antigen binding domain is from the scFv, hinge of Anti-BCMA antibody C11D5.3 The region is derived from amino acids 99-110 of IGHG4 (UniProtKB-P01861), the transmembrane region is derived from amino acids 153-179 of CD28 (UniProtKB-P10747), and the intracellular domain is derived from the 209th amino acid of 4-1BB (UniProtKB-Q07011). Amino acids ~255 and amino acids 62-133 of DAP12 (UniprotKB-O43914).
实施例1载体构建Example 1 Vector construction
(1)构建用于制备工程化K562-NK1细胞的载体(1) Construction of a vector for preparing engineered K562-NK1 cells
DNA片段1:CMV启动子核苷酸序列、puromycin的编码核苷酸序列、T2A、EGFP的编码核苷酸序列由本领域常规的生物技术服务公司合成;DNA fragment 1: CMV promoter nucleotide sequence, puromycin coding nucleotide sequence, T2A, EGFP coding nucleotide sequence were synthesized by conventional biotechnology service companies in the field;
DNA片段2:CMV启动子核苷酸序列、mbIL-15的编码核苷酸序列、P2A的编码核苷酸列、mbIL-21的编码核苷酸序列由本领域常规的生物技术服务公司合成;DNA fragment 2: CMV promoter nucleotide sequence, mbIL-15 coding nucleotide sequence, P2A coding nucleotide sequence, mbIL-21 coding nucleotide sequence are synthesized by conventional biotechnology service companies in the field;
将DNA片段1和DNA片段2克隆进pFastBac1质粒(购自Thermofisher),命名为质粒1。 DNA Fragment 1 and DNA Fragment 2 were cloned into pFastBac1 plasmid (purchased from Thermofisher) and designated as plasmid 1 .
(2)构建PiggyBac转座酶载体(辅助载体)(2) Construction of PiggyBac transposase vector (auxiliary vector)
为了构建PiggyBac转座酶编码质粒,在PiggyBac转座酶基因(GenBank:EF587698.1)的两端分别添加AgeI和SacI酶切位点,序列由外包的服务公司合成,并通过两个酶切位点克隆进pmaxCloning载体,命名为质粒2,如图1A所示,pmaxCloning载体中的CMV启动子控制PiggyBac转座酶的表达。In order to construct a plasmid encoding PiggyBac transposase, AgeI and SacI restriction sites were added to both ends of the PiggyBac transposase gene (GenBank: EF587698.1). Point-cloned into the pmaxCloning vector, named plasmid 2, as shown in Figure 1A, the CMV promoter in the pmaxCloning vector controls the expression of the PiggyBac transposase.
(3)构建piggyBac转座子载体(供体载体)(3) Construction of piggyBac transposon vector (donor vector)
将PiggyBac转座子的5’反向末端重复序列(SEQ ID NO:1)、鸡β-珠蛋白染色质绝缘子cHS4(GenBank:AY040835.1)、EF1α启动子、EcoRI和SalI酶切位点序列、SV40 polyA序列、反向互补的cHS4序列和3’反向末端重复序列(SEQ ID NO:2)融合,并由外包的服务公司合成,通过BsaI克隆进pmaxCloning载体(购自Lonza公司),命名为pZTS4,融合基因的表达由EF1α启动子控制;The 5' inverted terminal repeat of the PiggyBac transposon (SEQ ID NO: 1), the chicken β-globin chromatin insulator cHS4 (GenBank: AY040835.1), the EF1α promoter, the EcoRI and SalI restriction site sequences , SV40 polyA sequence, reverse complementary cHS4 sequence and 3' reverse terminal repeat sequence (SEQ ID NO: 2) were fused, and synthesized by an outsourced service company, cloned into pmaxCloning vector (purchased from Lonza company) through BsaI, named For pZTS4, the expression of the fusion gene is controlled by the EF1α promoter;
SEQ ID NO:1:5’-ccctagaaagataatcatattgtgacgtacgttaaagataatcatgtgtaaaattgacgcatg-3’;SEQ ID NO: 1: 5'-ccctagaaagataatcatattgtgacgtacgttaaagataatcatgtgtaaaattgacgcatg-3';
SEQ ID NO:2:5’-catgcgtcaattttacgcagactatctttctaggg-3’;SEQ ID NO: 2: 5'-catgcgtcaattttacgcagactatctttctaggg-3';
(4)构建含有NKG2D CAR的转座子供体载体(4) Construction of transposon donor vector containing NKG2D CAR
为了构建NKG2D CAR载体,将NKG2D的胞外抗原结合结构域(ED)与IGHG4铰链区、CD28跨膜区、4-1BB和CD3ζ信号传导结构域融合,生成二代NKG2D CAR载体;为了便于通过流式细胞术检测CAR的表达,在CAR的序列中均加入3个Strept-tag II(ST2)的重复;CAR片段由本领域常规的生物技术服务公司合成,序列的5’端和3’端包括限制性酶切位点EcoRI和SalI;合成的DNA片段通过EcoRI和SalI酶切后,克隆进pFastBac1质粒(购自Thermofisher),命名为质粒3。In order to construct the NKG2D CAR vector, the extracellular antigen-binding domain (ED) of NKG2D was fused with the IGHG4 hinge region, CD28 transmembrane region, 4-1BB and CD3ζ signaling domain to generate the second-generation NKG2D CAR vector; CAR expression was detected by cytometry, and 3 repeats of Strept-tag II (ST2) were added to the sequence of the CAR; the CAR fragment was synthesized by a conventional biotechnology service company in the field, and the 5' and 3' ends of the sequence included restriction Sexual restriction sites EcoRI and SalI; after the synthesized DNA fragment was digested by EcoRI and SalI, it was cloned into pFastBac1 plasmid (purchased from Thermofisher) and named as plasmid 3.
为了构建含有NKG2D CAR基因的转座子供体载体,将质粒2中的CAR序列通过EcoRI和SalI酶切后克隆进pZTS4,命名为质粒4,如图1B所示,CAR基因的表达由EF1α启动子控制。In order to construct a transposon donor vector containing the NKG2D CAR gene, the CAR sequence in plasmid 2 was digested with EcoRI and SalI and cloned into pZTS4, named plasmid 4. As shown in Figure 1B, the expression of the CAR gene is controlled by the EF1α promoter. control.
为了构建含有人IL-15基因的NKG2D CAR转座子供体载体,将IRES和人IL-15(UniprotKB:P40933)的序列融合,序列的5'端和3'端包括限制性酶切位点SalI。合成的DNA片段通过SalI酶切后,克隆进质粒4,命名为质粒5。In order to construct the NKG2D CAR transposon donor vector containing the human IL-15 gene, the sequence of IRES and human IL-15 (UniprotKB: P40933) was fused, and the 5' and 3' ends of the sequence included the restriction site SalI . The synthesized DNA fragment was digested with SalI and cloned into plasmid 4, which was named plasmid 5.
为了构建含有膜结合的人IL-15(mbIL-15)基因的NKG2D CAR转座子供体载体,将IRES和膜结合的人IL-15的序列融合,序列的5'端和3'端包括限制性酶切位点SalI。合成的DNA片段通过SalI酶切后,克隆进质粒4,命名为质粒6。To construct the NKG2D CAR transposon donor vector containing the membrane-bound human IL-15 (mbIL-15) gene, the IRES and the sequence of membrane-bound human IL-15 were fused, including restriction at the 5' and 3' ends of the sequence The cleavage site SalI. The synthesized DNA fragment was digested with SalI and cloned into plasmid 4, which was named plasmid 6.
(5)构建含有anti-BCMA CAR的转座子供体载体(5) Construction of transposon donor vector containing anti-BCMA CAR
为了构建anti-BCMA CAR载体,将anti-BCMA的scFv(C11D5.3)与IGHG44铰链区、CD28跨膜区、4-1BB和CD3ζ信号传导结构域融合,生成二代anti-BCMA CAR载体;为了便于通过流式细胞术检测CAR的表达,在CAR的序列中均加入3个Strept-tag II(ST2)的重复;CAR片段由外包的服务公司合成,序列的5’端和3’端包括限制性酶切位点EcoRI和SalI;合成的DNA片段通过EcoRI和SalI酶切后,克隆进pFastBac1质粒(购自Thermofisher),命名为质粒7。In order to construct an anti-BCMA CAR vector, the anti-BCMA scFv (C11D5.3) was fused with the IGHG44 hinge region, CD28 transmembrane region, 4-1BB and CD3ζ signaling domains to generate a second-generation anti-BCMA CAR vector; in order to To facilitate the detection of CAR expression by flow cytometry, 3 repeats of Strept-tag II (ST2) are added to the CAR sequence; the CAR fragment is synthesized by an outsourced service company, and the 5' and 3' ends of the sequence include restriction Sex restriction sites EcoRI and SalI; the synthesized DNA fragment was digested by EcoRI and SalI, and then cloned into pFastBac1 plasmid (purchased from Thermofisher), named plasmid 7.
为了构建含有anti-BCMA CAR基因的转座子供体载体,将质粒7中的CAR序列通过EcoRI和SalI酶切后克隆进pZTS4,命名为质粒8,CAR基因的表达由EF1α启动子控制。In order to construct a transposon donor vector containing the anti-BCMA CAR gene, the CAR sequence in plasmid 7 was digested with EcoRI and SalI and cloned into pZTS4, which was named plasmid 8. The expression of the CAR gene was controlled by the EF1α promoter.
(6)构建用于制备工程化K562-NK2细胞的载体(6) Construction of a vector for preparing engineered K562-NK2 cells
为了构建含有tBCMA基因的转座子供体载体,由外包公司将tBCMA的DNA序列合成,并通过EcoRI和SalI克隆进pZTS4,命名为质粒9。In order to construct a transposon donor vector containing the tBCMA gene, the DNA sequence of tBCMA was synthesized by an outsourcing company, and cloned into pZTS4 by EcoRI and SalI, named plasmid 9.
实施例2制备工程化K562-NK1细胞Example 2 Preparation of engineered K562-NK1 cells
将野生型K562细胞重悬于10mL Opti-MEM中,200×g离心5min,将细胞沉淀重悬于100μL P3 buffer中,加入10μg质粒1,混匀后转移至Lonza电击杯;The wild-type K562 cells were resuspended in 10 mL Opti-MEM, centrifuged at 200 × g for 5 min, the cell pellet was resuspended in 100 μL P3 buffer, 10 μg plasmid 1 was added, and after mixing, it was transferred to the Lonza electric shock cup;
将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转,电转结束后,将一个cuvette中的K562细胞悬液缓慢转移至6孔板的一个孔中,孔里预先加入K562培养基(IMDM+10%FBS);Place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (single electroporation cup module) for electroporation. After electroporation, slowly transfer the K562 cell suspension in a cuvette to a well of a 6-well plate. K562 medium (IMDM+10%FBS);
电穿孔后的第5天,在200μg/mL浓度下进行为期1个月的puromycin筛选,培养基每2天更换一次;On the 5th day after electroporation, puromycin screening was performed at a concentration of 200 μg/mL for 1 month, and the medium was changed every 2 days;
1个月后,使用细胞分选仪BD FACSAria(BD Biosciences公司)将单细胞分选到96孔板,对扩增的单细胞克隆进行流式细胞术分析,检测融合基因的表达情况,检测抗体分别为APC-结合的Streptavidin、biotin-结合的抗人IL-15抗体和biotin-结合的抗人IL-21抗体,筛选到的单细胞克隆命名为K562-NK1。One month later, single cells were sorted into 96-well plates using a cell sorter BD FACSAria (BD Biosciences), and the amplified single cell clones were analyzed by flow cytometry to detect fusion gene expression and antibody detection. APC-conjugated Streptavidin, biotin-conjugated anti-human IL-15 antibody and biotin-conjugated anti-human IL-21 antibody, respectively, and the screened single cell clone was named K562-NK1.
如图2A和图2B所示,K562-NK1细胞表面表达了高水平的人IL-15和人-IL21,其阳性率分别为100.0%和78.9%。As shown in Figure 2A and Figure 2B, K562-NK1 cells expressed high levels of human IL-15 and human-IL21 on the surface, with positive rates of 100.0% and 78.9%, respectively.
实施例3利用piggyBac转座子系统制备NKG2D CAR-NKExample 3 Preparation of NKG2D CAR-NK using piggyBac transposon system
制备流程如图3所示:The preparation process is shown in Figure 3:
第0天,将5×10 6个PBMC和5×10 6K562-NK1(100Gyγ射线辐照)重悬于10mL NK细胞培养基,接种于T25细胞培养瓶中,加入hrIL-2使其终浓度为100IU/mL; On day 0, 5×10 6 PBMCs and 5×10 6 K562-NK1 (100Gy γ-irradiated) were resuspended in 10 mL of NK cell culture medium, seeded into T25 cell culture flasks, and hrIL-2 was added to make the final concentration is 100IU/mL;
第2天,取出悬浮的细胞进行计数,300×g离心10min;用10mL Opti-MEM重悬细胞沉淀,300×g 离心10min;将细胞沉淀重悬于100μL P3 buffer中,加入5μg质粒2和10μg质粒4,混匀后转移至电Lonza电击杯;将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转,电转结束后,将一个电击杯中的NK细胞悬液缓慢转移至一个新的T25细胞培养瓶中,加入含有终浓度为100IU/mL的hrIL-2的10mL NK细胞培养液,轻轻混匀,将T25细胞培养瓶置于37℃细胞培养箱培养;On the second day, the suspended cells were taken out and counted, and centrifuged at 300 × g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 × g for 10 min; resuspended the cell pellet in 100 μL P3 buffer, and added 5 μg plasmid 2 and 10 μg Plasmid 4, mix well and transfer to the electroporation Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module), and perform electroporation. After the electroporation, slowly transfer the NK cell suspension in one electroporation cup Transfer to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing a final concentration of 100IU/mL hrIL-2, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
第3天至第9天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 3rd day to the 9th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第10天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度100IU/mL; On the 10th day, all cells were collected and counted, and 2 × 10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2 × 10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-2 to make the final concentration 100IU/mL;
第10天至第16天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 10th day to the 16th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第17天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度100IU/mL; On the 17th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2×10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-2 to make the final concentration 100IU/mL;
第17天至第23天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 17th day to the 23rd day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第24天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度100IU/mL; On the 24th day, all cells were collected and counted, and 2 × 10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2 × 10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-2 to make the final concentration 100IU/mL;
第24天至第30天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 24th day to the 30th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第31天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体)。 On the 31st day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, and anti-Strep-tag II antibody).
实施例4不同电转条件对CAR-NK细胞的扩增倍数和细胞表型的影响Example 4 Effects of different electroporation conditions on the expansion fold and cell phenotype of CAR-NK cells
本实施例比较了在NK细胞活化前进行电转或在NK细胞活化后的第2或第4天进行电转,获得的CAR-NK中的NK细胞比例、CAR在NK细胞中的表达比例以及总细胞的扩增倍数。This example compares the ratio of NK cells in CAR-NK, the ratio of CAR expression in NK cells, and the total cells obtained by electroporation before NK cell activation or on the 2nd or 4th day after NK cell activation. the amplification fold.
在NK细胞活化后的第2天进行电转即为实施例3的方法记为Day2 EP;在NK细胞活化前进行电转的方法与实施例3的不同之处在于将5×10 6个PBMC按照实施例3的方法电转,电转完成后立刻与5×10 6个K562-NK1(100Gyγ射线辐照)共培养,记为Day0 EP;在NK细胞活化后的第4天进行电转与实施例3的不同之处在于将5×10 6PBMC和5×10 6K562-NK1(100Gyγ射线辐照)共培养4天后,再将悬浮细胞取出进行电转,记为Day4 EP。 Electrically At day 2 activated NK cells transfected is the procedure of Example 3 is referred to as Day2 EP; method of electroporation prior to activation of NK cells is different from the embodiment 3 in that the 5 × 10 6 PBMC were as described in embodiment The method of Example 3 was electroporated. Immediately after electroporation, it was co-cultured with 5×10 6 K562-NK1 (100Gy γ-ray irradiation), which was recorded as Day0 EP; electroporation was performed on the 4th day after the activation of NK cells, which was different from that in Example 3. The point is that after 5×10 6 PBMC and 5×10 6 K562-NK1 (100 Gy γ-ray irradiation) were co-cultured for 4 days, the suspended cells were taken out and electroporated, which was recorded as Day4 EP.
采用以上三种方法从1个正常捐献者PBMC中制备CAR-NK细胞,对获得的NK细胞纯度(CD3-CD56+)进行比较,结果如图4A所示,实施例3制备的CAR-NK细胞中NK细胞的纯度在培养第10、17、24天时分别为68%、77%、85%,而在NK细胞活化前进行电转制备的CAR-NK细胞中NK 细胞的纯度在培养第10、17天分别为11%和13%,在NK细胞活化后4天进行电转制备的CAR-NK细胞中NK细胞的纯度在培养第10、17、24天分别为46%、81%、88%。说明在NK细胞活化后进行电转比在NK活化前进行电转制备的CAR-NK细胞中,NK细胞的纯度更高。The above three methods were used to prepare CAR-NK cells from a normal donor PBMC, and the obtained NK cell purity (CD3-CD56+) was compared. The results are shown in Figure 4A. The purity of NK cells was 68%, 77%, and 85% on the 10th, 17th, and 24th days of culture, respectively, while the purity of NK cells in the CAR-NK cells prepared by electroporation before NK cell activation was on the 10th, 17th day of culture. were 11% and 13%, respectively, and the purity of NK cells in CAR-NK cells prepared by electroporation 4 days after NK cell activation were 46%, 81%, and 88% on culture days 10, 17, and 24, respectively. It shows that the purity of NK cells is higher in the CAR-NK cells prepared by electroporation after NK cell activation than that prepared by electroporation before NK activation.
进一步地,对获得的CAR-NK细胞中CAR的表达比例进行比较,结果如图4B所示,实施例3制备的CAR-NK细胞中CAR+的NK细胞比例在培养第10、17、24天时分别为14%、30%、45%,而在NK细胞活化前进行电转制备的CAR-NK细胞中CAR+的NK细胞比例在培养第10、17天分别为14%和27%,在NK细胞活化后4天进行电转制备的CAR-NK细胞中CAR+的NK细胞比例在培养第10、17、24天分别为2%、15%、19%。说明越早进行电转,CAR+的NK细胞比例越高。Further, the expression ratio of CAR in the obtained CAR-NK cells was compared, and the results were shown in Figure 4B. were 14%, 30%, and 45%, while the proportion of CAR+ NK cells in CAR-NK cells prepared by electroporation before NK cell activation was 14% and 27% on the 10th and 17th days of culture, respectively. The proportion of CAR+ NK cells in the CAR-NK cells prepared by electroporation on day 4 was 2%, 15%, and 19% on the 10th, 17th, and 24th days of culture, respectively. It shows that the earlier electroporation is performed, the higher the proportion of CAR+ NK cells.
本实施例还对获得的细胞总数扩增倍数进行了比较,结果如图5所示,实施例3的方法扩增24天得到的细胞总数扩增倍数为3022倍;在NK细胞活化前电转、共扩增17天,细胞总数扩增倍数为133倍;在NK细胞活化后4天电转、共扩增24天,细胞总数扩增倍数为736倍。说明在NK细胞活化后第2天进行电转所得细胞的扩增倍数最高。This example also compares the amplification multiples of the total number of cells obtained. The results are shown in Figure 5. The amplification multiples of the total number of cells obtained by the method of Example 3 for 24 days is 3022 times; After a total of 17 days of expansion, the total number of cells was expanded by 133 times; 4 days after the activation of NK cells, electroporated for 24 days, the total number of cells was expanded by 736 times. This indicated that the cells obtained by electroporation on the second day after the activation of NK cells had the highest expansion fold.
综合这三面的参数比较,实施例3的方法,即在NK细胞活化后2天进行电转,所制备的CAR-NK细胞,其NK细胞纯度、CAR+的NK细胞比例以及总细胞数的扩增倍数相对最好。Based on the comparison of these three parameters, the method of Example 3, that is, electroporation was performed 2 days after the activation of NK cells, the prepared CAR-NK cells, the purity of NK cells, the proportion of CAR+ NK cells, and the amplification fold of the total number of cells. Relatively best.
实施例5多个不同来源的单个核细胞均可利用PiggyBac转座子系统制备NKG2D CAR-NKExample 5 Mononuclear cells from multiple sources can use the PiggyBac transposon system to prepare NKG2D CAR-NK
本实施例中将7个不同来源的单个核细胞(MNC)按照实施例3所述的方法制备NKG2D CAR-NK,其中HD001、HD002、HD003、HD004、HD005、HD006来自健康捐献者外周血提取的PBMC,CB001来自脐带血提取的MNC。In this example, 7 mononuclear cells (MNCs) from different sources were used to prepare NKG2D CAR-NK according to the method described in Example 3, wherein HD001, HD002, HD003, HD004, HD005, and HD006 were extracted from peripheral blood of healthy donors. PBMC, CB001 was derived from MNC extracted from umbilical cord blood.
如图6A所示,第一轮共培养结束,CD3-CD56+的NK细胞比例达到90%左右,且在接下来几轮与K562-NK1(100Gyγ射线辐照)共培养后,NK比例基本不变,个别供体略有下降。如图6B所示,第一轮共培养结束,CAR+的NK细胞比例在18%~50%的范围内(中位数:19.5%),随着共培养次数的增加,CAR+的NK细胞比例也逐渐增加,在第四轮共培养结束,CAR+的NK细胞比例在30%~90%的范围内(中位数:64.7%)。As shown in Figure 6A, at the end of the first round of co-culture, the proportion of CD3-CD56+ NK cells reached about 90%, and after the next few rounds of co-culture with K562-NK1 (100Gy γ-ray irradiation), the proportion of NK cells remained basically unchanged. , the individual donors decreased slightly. As shown in Figure 6B, at the end of the first round of co-culture, the proportion of CAR+ NK cells ranged from 18% to 50% (median: 19.5%). With the increase of the number of co-cultures, the proportion of CAR+ NK cells also increased. Gradually increased, and at the end of the fourth round of co-culture, the proportion of CAR+ NK cells ranged from 30% to 90% (median: 64.7%).
如图7所示,第四轮共培养结束,总细胞数的累积扩增倍数在3382~24257的范围内(中位数:13580)。As shown in Figure 7, the fourth round of co-culture was completed, and the cumulative expansion fold of the total number of cells was in the range of 3382-24257 (median: 13580).
通过RTCA实时杀伤检测仪检测HD006 PBMC制备的NKG2D CAR-NK对肿瘤细胞株的体外杀伤情况。RTCA实验开始第0小时,将人结直肠癌细胞株HCT116或人胃癌细胞株MGC803细胞铺于16孔电极板(ACEA Bio公司)中,5000个细胞/孔;约24小时后,将NKG2D CAR-NK细胞和NK细胞(效应细胞)按效应细胞:靶细胞(E:T)=1:1的比例接种于电极板中,总体积200μL,每组实验设置2个复孔;其中NK细胞采用实施例3的方法制备,但是无需进行电转;用xCELLigence RTCA检测NKG2D CAR-NK细胞对HCT116和MGC803体外杀伤作用,HD006 PBMC制备的NKG2D CAR-NK在体外对HCT116和 MGC803也体现了显著的肿瘤杀伤作用(如图8A和图8B)。不仅如此,NKG2D CAR-NK在体外与HCT116细胞、MGC803细胞和人SKOV3细胞(E:T=10:1)共培养过夜后,培养液上清中的IFNγ含量显著增加(如图9)。The in vitro killing of tumor cell lines by NKG2D CAR-NK prepared from HD006 PBMC was detected by RTCA real-time killing detector. At the 0th hour of the RTCA experiment, human colorectal cancer cell line HCT116 or human gastric cancer cell line MGC803 cells were plated in a 16-well electrode plate (ACEA Bio Company), 5000 cells/well; about 24 hours later, NKG2D CAR- NK cells and NK cells (effector cells) were seeded in the electrode plate at a ratio of effector cells:target cells (E:T)=1:1, with a total volume of 200 μL, and each group of experiments set 2 replicate wells; The method of Example 3 was prepared, but electroporation was not required; xCELLigence RTCA was used to detect the killing effect of NKG2D CAR-NK cells on HCT116 and MGC803 in vitro, and NKG2D CAR-NK prepared by HD006 PBMC also showed significant tumor killing effect on HCT116 and MGC803 in vitro (Fig. 8A and Fig. 8B). Not only that, after overnight co-culture of NKG2D CAR-NK with HCT116 cells, MGC803 cells and human SKOV3 cells (E:T=10:1) in vitro, the IFNγ content in the culture supernatant was significantly increased (Figure 9).
实施例6直接对NK细胞进行电转,转基因的表达效率及NK细胞存活率Example 6 Direct electroporation of NK cells, transgene expression efficiency and NK cell survival rate
本实施例以绿色荧光蛋白GFP基因为报告基因,直接用pmax-GFP质粒对扩增17天的NK细胞进行电转,测试电转后1天NK细胞中GFP的表达比例及NK细胞的存活率。NK细胞扩增流程如下:第0天,将2×10 6个PBMC和2×10 6K562-NK1(100Gyγ射线辐照)重悬于10mL NK细胞培养基,接种于T75细胞培养瓶中(竖着),加入hrIL-2使其终浓度为100IU/mL;第2天至第10天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;第10天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度100IU/mL;第10天至第17天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;第17天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体)。取扩增第17天的NK细胞,采用实施例3中电转的方法,电转的NK细胞数为2×10 6,并将质粒替换为pmax-GFP质粒。电转后第二天,进行细胞计数和流式细胞术检测NK细胞中GFP的比例。如图10A和图10B所示,电转第二天,虽然GFP的表达比例有17.99%,NK细胞的存活率仅有3.2%。 In this example, the green fluorescent protein GFP gene was used as the reporter gene, and the pmax-GFP plasmid was directly used to electroporate the NK cells amplified for 17 days. The NK cell expansion procedure is as follows: On day 0, 2×10 6 PBMCs and 2×10 6 K562-NK1 (100Gy γ-ray irradiated) were resuspended in 10 mL of NK cell culture medium, and seeded in T75 cell culture flasks (vertical). hrIL-2 was added to make the final concentration 100IU/mL; from the 2nd day to the 10th day, according to the cell growth, an appropriate amount of fresh NK cell culture medium containing hrIL-2 was added; on the 10th day, all cells were collected Counting, taking 2 × 10 6 cells for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody); 2 × 10 6 cells and 2 × 10 6 K562-NK1 (100Gy γ-ray irradiation) ) was resuspended in 10 mL of medium, and hrIL-2 was added to make its final concentration 100 IU/mL; from the 10th day to the 17th day, according to the cell growth, an appropriate amount of fresh NK cell culture medium containing hrIL-2 was added; the 17th day On the next day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody). The NK cells on the 17th day of expansion were taken, and the electroporation method in Example 3 was used. The number of electroporated NK cells was 2×10 6 , and the plasmid was replaced by the pmax-GFP plasmid. On the second day after electroporation, cell counting and flow cytometry were performed to detect the proportion of GFP in NK cells. As shown in Figure 10A and Figure 10B, on the second day of electroporation, although the expression ratio of GFP was 17.99%, the survival rate of NK cells was only 3.2%.
上述结果表明,如果单纯只是对NK细胞进行电转,不用人工抗原呈递细胞富集扩增,NK细胞的活率和转基因的表达率都很低。因此转座子系统和人工抗原呈递细胞的结合是必须的,可以显著提高NK细胞的存活及扩增倍数,并且显著提高嵌合受体修饰的NK细胞的比例。The above results show that if only electroporation of NK cells is performed without artificial antigen-presenting cells enrichment and expansion, the viability of NK cells and the expression rate of transgenes are very low. Therefore, the combination of transposon system and artificial antigen-presenting cells is necessary, which can significantly improve the survival and expansion fold of NK cells, and significantly increase the proportion of NK cells modified by chimeric receptors.
实施例7利用piggyBac转座子系统制备NKG2D eCAR-NKExample 7 Preparation of NKG2D eCAR-NK using piggyBac transposon system
第0天,将5×10 6个PBMCs和5×10 6K562-NK1(100Gyγ射线辐照)重悬于10mL NK细胞培养基,接种于T25细胞培养瓶中,加入hrIL-2使其终浓度为100IU/mL; On day 0, 5×10 6 PBMCs and 5×10 6 K562-NK1 (100 Gy γ-irradiated) were resuspended in 10 mL of NK cell culture medium, seeded into T25 cell culture flasks, and hrIL-2 was added to make the final concentration is 100IU/mL;
第2天,取出悬浮的细胞进行计数,300×g离心10min;用10mL Opti-MEM重悬细胞沉淀,300×g离心10min;将细胞沉淀重悬于100μL P3 buffer中,加入5μg质粒2和10μg质粒4,混匀后转移至Lonza电击杯;将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转,电转结束后,将一个电击杯中的NK细胞悬液缓慢转移至一个新的T25细胞培养瓶中,加入含有终浓度为100IU/mL的hrIL-2的10mL NK细胞培养液,轻轻混匀,将T25细胞培养瓶置于37℃细胞培养箱培养;On the second day, the suspended cells were taken out for counting, and centrifuged at 300 × g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 × g for 10 min; resuspended the cell pellet in 100 μL P3 buffer, and added 5 μg plasmid 2 and 10 μg Plasmid 4, mix well and transfer to the Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module) and perform electroporation. After the electroporation, slowly transfer the NK cell suspension in one electroporation cup To a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 100IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
第3天至第9天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 3rd day to the 9th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第10天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培 养基中,加入hrIL-12、hrIL-15、hrIL-18使其终浓度分别为10ng/mL、50ng/mL和50ng/mL; On the 10th day, all cells were collected and counted, and 2 × 10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2 × 10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-12, hrIL-15, hrIL-18 to make the final concentrations of 10ng/mL, 50ng/mL and 50ng, respectively /mL;
第11天,收集全部细胞300×g离心10min,用20mL新鲜的NK细胞培养基重悬,添加hrIL-2使其终浓度100IU/mL;On the 11th day, all cells were collected and centrifuged at 300 × g for 10 min, resuspended in 20 mL of fresh NK cell culture medium, and hrIL-2 was added to make the final concentration 100 IU/mL;
第12天至第16天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 12th day to the 16th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第17天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度为100IU/mL; On the 17th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2×10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-2 to make the final concentration 100IU/mL;
第17天至第23天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 17th day to the 23rd day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第24天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度为100IU/mL; On the 24th day, all cells were collected and counted, and 2 × 10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2 × 10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-2 to make the final concentration 100IU/mL;
第24天至第30天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 24th day to the 30th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第31天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体)。 On the 31st day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, and anti-Strep-tag II antibody).
实施例8不同细胞因子组合物对CAR-NK细胞的扩增倍数和细胞表型的影响Example 8 Effects of different cytokine compositions on the expansion fold and cell phenotype of CAR-NK cells
本实施例比较了在CAR-NK制备第10天,与K562-NK1第二次共培养时,是否添加细胞因子组合物(IL-12、IL-15、IL-18)对CAR-NK细胞生长的影响。This example compares the effect of adding cytokine compositions (IL-12, IL-15, IL-18) on the growth of CAR-NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation. Impact.
CAR-NK组,即CAR-NK制备第10天与K562-NK1第二次共培养时,只添加hrIL-2(实施例3);CAR-NK group, that is, when CAR-NK was co-cultured with K562-NK1 for the second time on the 10th day of preparation, only hrIL-2 was added (Example 3);
eCAR-NK组,即CAR-NK制备第10天与K562-NK1第二次共培养时,添加细胞因子组合物IL-12、IL-15、IL-18(实施例7)。In the eCAR-NK group, that is, the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation, cytokine compositions IL-12, IL-15, and IL-18 were added (Example 7).
采用以上两种方法从同一个正常外周血捐献者的PBMC中制备CAR-NK细胞,对获得的NK细胞纯度(CD3-CD56+)进行比较,结果如图11A所示,实施例3制备的CAR-NK细胞中NK细胞的纯度在培养第10、17、24、31天时分别为79%、81%、90%、93%,而实施例4制备的eCAR-NK细胞中NK细胞的纯度在培养第10、17、24、31天时分别为79%、87%、93%、93%。说明在CAR-NK制备第10天与K562-NK1第二次共培养时,添加细胞因子组合物IL-12、IL-15、IL-18可以略微提高NK细胞的比例。The above two methods were used to prepare CAR-NK cells from the PBMC of the same normal peripheral blood donor, and the obtained NK cell purity (CD3-CD56+) was compared. The purity of NK cells in NK cells was 79%, 81%, 90%, and 93% on the 10th, 17th, 24th, and 31st days of culture, respectively, while the purity of NK cells in the eCAR-NK cells prepared in Example 4 was on the 1st day of culture. At 10, 17, 24, and 31 days, they were 79%, 87%, 93%, and 93%, respectively. This indicates that the addition of cytokines IL-12, IL-15 and IL-18 can slightly increase the proportion of NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation.
进一步地,对获得的细胞中CAR+的NK细胞比例进行比较,结果如图11B所示,实施例3制备的CAR-NK细胞中CAR+的NK细胞比例在培养第10、17、24、31天时分别为34%、47%、49%、74%,而实施例7制备的eCAR-NK细胞中CAR+的NK细胞比例在培养第10、17、24、31天时分别为34%、57%、58%、85%。说明在CAR-NK制备第10天与K562-NK1第二次共培养时,添加细胞因子组合物IL-12、IL-15、IL-18可以显著提高CAR在NK细胞中的表达比例。Further, the ratio of NK cells of CAR+ in the obtained cells was compared. The results are shown in Figure 11B. The ratio of NK cells of CAR+ in the CAR-NK cells prepared in Example 3 was on the 10th, 17th, 24th, and 31st days of culture, respectively. were 34%, 47%, 49%, and 74%, while the proportion of CAR+ NK cells in the eCAR-NK cells prepared in Example 7 was 34%, 57%, and 58% on the 10th, 17th, 24th, and 31st days of culture, respectively. , 85%. This indicated that the addition of cytokine compositions IL-12, IL-15 and IL-18 could significantly increase the expression ratio of CAR in NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation.
本实施例同时对CAR-NK和eCAR-NK细胞制备过程所获得的细胞总数扩增倍数进行比较,结果如图12所示,实施例3制备的CAR-NK细胞,其总扩增倍数在第0-10天、10-17天、17-24天和24-31天分别为27、21、9、8倍;而实施例7制备的eCAR-NK细胞,其总扩增倍数在第0-10天、10-17天、17-24天和24-31天分别为27、27、9、7倍。说明在CAR-NK制备第10天与K562-NK1第二次共培养时,添加细胞因子组合物IL-12、IL-15、IL-18可提高NK细胞的扩增倍数。In this example, the total cell expansion folds obtained in the preparation process of CAR-NK and eCAR-NK cells are compared. The results are shown in Figure 12. The total expansion fold of the CAR-NK cells prepared in Example 3 is in 0-10 days, 10-17 days, 17-24 days and 24-31 days were 27, 21, 9, and 8 times respectively; while the eCAR-NK cells prepared in Example 7 had a total expansion fold at the 0- 10 days, 10-17 days, 17-24 days and 24-31 days were 27, 27, 9, and 7 times, respectively. This indicated that the addition of cytokine compositions IL-12, IL-15 and IL-18 could increase the expansion fold of NK cells during the second co-culture with K562-NK1 on the 10th day of CAR-NK preparation.
综合比较以上三方面的数据,实施例7所述方法,即在CAR-NK制备第10天与K562-NK1第二次共培养时,添加细胞因子组合物IL-12、IL-15、IL-18,有助于CAR+的NK细胞的扩增。Comprehensively comparing the data of the above three aspects, the method described in Example 7, that is, adding the cytokine compositions IL-12, IL-15, IL- 18. Contribute to the expansion of CAR+ NK cells.
实施例9 CAR-NK制备第10天添加细胞因子组合物,对人卵巢癌细胞株SKOV3体外杀伤情况的作用(RTCA)Example 9 The effect of adding cytokine composition on the 10th day of CAR-NK preparation on the killing of human ovarian cancer cell line SKOV3 in vitro (RTCA)
本实施例比较了在NKG2D CAR-NK制备第10天,与K562-NK1第二次共培养时,是否添加细胞因子组合物(IL-12、IL-15、IL-18)对CAR-NK体外杀伤能力的影响。This example compares the effect of adding cytokine compositions (IL-12, IL-15, IL-18) on the in vitro effects of CAR-NK on the 10th day of NKG2D CAR-NK preparation, when it was co-cultured with K562-NK1 for the second time. impact on lethality.
RTCA实验开始第0小时,将SKOV3细胞铺于16孔电极板(ACEA Bio公司)中,5000个细胞/孔;约24小时后,将培养第17天的NKG2D CAR-NK细胞、NKG2D eCAR-NK细胞和NK细胞(效应细胞)分别按效应细胞:靶细胞=1:1或2:1的比例接种于电极板中,总体积200μL,每组实验设置2个复孔;其中NK细胞采用实施例3的方法制备,但是无需进行电转;用xCELLigence RTCA检测NKG2D CAR-NK细胞对SKOV3细胞体外杀伤作用。At the 0th hour of the RTCA experiment, SKOV3 cells were plated in a 16-well electrode plate (ACEA Bio Company), 5000 cells/well; about 24 hours later, the NKG2D CAR-NK cells, NKG2D eCAR-NK cells and NKG2D eCAR-NK cells were cultured on the 17th day. Cells and NK cells (effector cells) were seeded in the electrode plate at the ratio of effector cells:target cells=1:1 or 2:1 respectively, with a total volume of 200 μL, and 2 duplicate wells were set for each group of experiments; NK cells were used in the example 3, but without electroporation; the cytotoxic effect of NKG2D CAR-NK cells on SKOV3 cells in vitro was detected by xCELLigence RTCA.
如图13A和图13B所示,未加入效应细胞组(图中显示为“SKOV3”),肿瘤细胞持续生长至70个小时;当效应细胞:肿瘤细胞=1:1时,加入NK细胞(图中显示为“SKOV3:NK 1:1”)或CAR-NK细胞(图中显示为“SKOV3:CAR-NK 1:1”)后,可显著抑制肿瘤细胞的生长,其中CAR-NK细胞的杀瘤作用显著优于NK细胞,这表明了NKG2D CAR-NK细胞中CAR的作用;而加入eCAR-NK细胞(图中显示为“SKOV3:eCAR-NK 1:1”)或CAR-NK细胞(图中显示为“SKOV3:CAR-NK 1:2”)后,肿瘤细胞几乎完全被杀死。As shown in Figure 13A and Figure 13B, without adding effector cells (shown as "SKOV3" in the figure), tumor cells continued to grow for 70 hours; when effector cells:tumor cells=1:1, NK cells were added (Figure 13B). (shown as "SKOV3:NK 1:1" in the figure) or CAR-NK cells (shown as "SKOV3:CAR-NK 1:1" in the figure), the growth of tumor cells can be significantly inhibited, in which the killing of CAR-NK cells The tumor effect was significantly better than that of NK cells, which indicated the effect of CAR in NKG2D CAR-NK cells; while adding eCAR-NK cells (shown as "SKOV3:eCAR-NK 1:1" in the figure) or CAR-NK cells (Fig. shown as "SKOV3:CAR-NK 1:2"), the tumor cells were almost completely killed.
对加入效应细胞后24h的数据进行分析(图8A竖虚线所示处),根据以下公式计算肿瘤生长抑制率:The data 24h after adding effector cells (indicated by the vertical dotted line in Figure 8A) were analyzed, and the tumor growth inhibition rate was calculated according to the following formula:
100%×(SKOV3 cell index-实验组cell index)/SKOV3 cell index100%×(SKOV3 cell index-experimental group cell index)/SKOV3 cell index
可以发现加入eCAR-NK细胞组(图中显示为“SKOV3:eCAR-NK 1:1”)与加入CAR-NK细胞组(图中显示为“SKOV3:CAR-NK 1:2”),其肿瘤抑制率都在80%左右,这表明在NKG2D CAR-NK制备第10天,与K562-NK1第二次共培养时,添加细胞因子组合物(IL-12、IL-15、IL-18)可以显著提高CAR-NK细胞对肿瘤细胞的生长抑制作用,原因可能是NK细胞中CAR+的NK细胞比例的提升以及细胞因子组合物(IL-12、IL-15、IL-18)增强了NK细胞的本身杀伤作用。It can be found that adding the eCAR-NK cell group (shown as "SKOV3:eCAR-NK 1:1" in the figure) and adding the CAR-NK cell group (shown as "SKOV3:CAR-NK 1:2" in the figure), the tumor The inhibition rates were all around 80%, which indicated that the addition of cytokine compositions (IL-12, IL-15, IL-18) during the second co-culture with K562-NK1 on the 10th day of NKG2D CAR-NK preparation could Significantly improved the growth inhibitory effect of CAR-NK cells on tumor cells, which may be due to the increase in the proportion of NK cells with CAR+ in NK cells and the enhancement of NK cells by cytokine compositions (IL-12, IL-15, IL-18). its own killing effect.
实施例10利用PiggyBac转座子系统制备高纯度的NKG2D eCAR-NKExample 10 Preparation of high-purity NKG2D eCAR-NK using the PiggyBac transposon system
具体步骤包括:第0天,取2×10 7个PBMC细胞,用CD3磁珠(Miltenyi)按照厂家的推荐步骤进行CD3+细胞去除;将5×10 6个分选后的PBMC和5×10 6K562-NK1(100Gyγ射线辐照)重悬于10mL NK细胞培养基,接种于T25细胞培养瓶中,加入hrIL-2使其终浓度为50IU/mL; The specific steps include: on the 0th day, 2×10 7 PBMC cells were taken, and CD3+ cells were removed with CD3 magnetic beads (Miltenyi) according to the manufacturer’s recommended procedure; 5×10 6 sorted PBMCs and 5×10 6 cells were removed K562-NK1 (100Gy γ-ray irradiation) was resuspended in 10mL of NK cell culture medium, inoculated into a T25 cell culture flask, and hrIL-2 was added to make the final concentration 50IU/mL;
第2天,取出悬浮的细胞进行计数,300×g离心10min;用10mL Opti-MEM重悬细胞沉淀,300×g离心10min;将细胞沉淀重悬于100μL P3 buffer中,加入5μg质粒2和10μg质粒4,混匀后转移至Lonza电击杯;将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转,电转结束后,将一个电击杯中的NK细胞悬液缓慢转移至一个新的T25细胞培养瓶中,加入含有终浓度为50IU/mL的hrIL-2的10mL NK细胞培养液,轻轻混匀,将T25细胞培养瓶置于37℃细胞培养箱培养;On the second day, the suspended cells were taken out for counting, and centrifuged at 300 × g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 × g for 10 min; resuspended the cell pellet in 100 μL P3 buffer, and added 5 μg plasmid 2 and 10 μg Plasmid 4, mix well and transfer to the Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module) and perform electroporation. After the electroporation, slowly transfer the NK cell suspension in one electroporation cup To a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 50IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
第3天至第9天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 3rd day to the 9th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第10天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);取出1×10 7的NKG2D CAR-NK细胞,重悬于80μL DPBS+1%AB血清的缓冲液中,加入20μL anti-Strep tag II抗体,混匀后于2~8℃孵育20min;孵育完成后加入5mL缓冲液,300g离心10min,弃上清并重悬于80μL DPBS+1%AB血清的缓冲液中,加入20μL anti-Biotin磁珠,混匀后于2~8℃孵育15min;孵育完成后加入5mL缓冲液,300g离心10min,弃上清并重悬于500μL缓冲液,接下来按照Miltenyi厂家的推荐步骤进行磁珠标记的阳性细胞的分选。取2×10 6分选后磁珠标记的CAR-NK细胞与4×10 6个K562-NK1(100Gyγ射线辐照)重悬于20mL培养基中,加入hrIL-2使其终浓度为50IU/mL。 On the 10th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 1×10 7 NKG2D was taken out CAR-NK cells were resuspended in 80 μL of DPBS+1% AB serum buffer, added with 20 μL of anti-Strep tag II antibody, mixed and incubated at 2-8°C for 20 min; after incubation, 5 mL of buffer was added and centrifuged at 300g 10min, discard the supernatant and resuspend in 80μL DPBS+1% AB serum buffer, add 20μL anti-Biotin magnetic beads, mix well, incubate at 2-8°C for 15min; add 5mL buffer after incubation, centrifuge at 300g for 10min , discard the supernatant and resuspend in 500 μL of buffer, and then follow the steps recommended by the Miltenyi manufacturer to sort the positive cells labeled with magnetic beads. Take 2×10 6 sorted magnetic bead-labeled CAR-NK cells and 4×10 6 K562-NK1 (100Gy γ-ray irradiated) and resuspend in 20 mL of medium, and add hrIL-2 to make the final concentration 50IU/ mL.
第11天至第16天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 11th day to the 16th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第17天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-12、hrIL-15、hrIL-18使其终浓度分别为10ng/mL、50ng/mL和50ng/mL; On the 17th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2×10 6 cells were Resuspend with 2×10 6 K562-NK1 (100Gy γ-ray irradiation) in 10mL medium, add hrIL-12, hrIL-15, hrIL-18 to make the final concentrations of 10ng/mL, 50ng/mL and 50ng, respectively /mL;
第18天,收集全部细胞收集300×g离心10min,用20mL新鲜的NK细胞培养基重悬,添加hrIL-2使其终浓度50IU/mL;On the 18th day, all cells were collected, centrifuged at 300 × g for 10 min, resuspended in 20 mL of fresh NK cell culture medium, and hrIL-2 was added to make the final concentration 50 IU/mL;
第18天至第23天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 18th day to the 23rd day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第24天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体)。 On the 24th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, and anti-Strep-tag II antibody).
实施例11 CAR+细胞分选可显著提高CAR+NK细胞的比例Example 11 CAR+ cell sorting can significantly increase the proportion of CAR+ NK cells
本实施例中比较了对CAR+细胞进行分选以及不同的分选方法对CAR-NK细胞制备的影响。In this example, the sorting of CAR+ cells and the effect of different sorting methods on the preparation of CAR-NK cells are compared.
取第一轮共培养结束第10天的NKG2D CAR-NK细胞,分为三组:第一组取2×10 6个细胞与2×10 6个K562-NK1(100Gyγ射线辐照)共培养1周,命名为未分选组;第二组取1×10 7个细胞按实施例10所 述方法进行分选,分选后取剩余细胞按NK细胞:人工抗原呈递细胞=1:2与K562-NK1(100Gyγ射线辐照)共培养1周,命名为Biotin Anti-Strep tag II Ab+anti-Biotin microbeads组;第三组取1×10 7的细胞,重悬于90μL DPBS+1%AB血清的缓冲液中,加入10μL Streptavidin microbeads,混匀后于2~8℃孵育20min;孵育完成后加入5mL缓冲液,300×g离心10min,弃上清并重悬于500μL缓冲液,接下来进行磁珠标记的阳性细胞的分选,分选后取剩余细胞按NK细胞:人工抗原呈递细胞=1:2与K562-NK1(100Gyγ射线辐照)共培养1周,命名为Streptavidin microbeads组。 The NKG2D CAR-NK cells on the 10th day after the first round of co-culture were taken and divided into three groups: in the first group, 2 × 10 6 cells were co-cultured with 2 × 10 6 K562-NK1 (100Gy γ-ray irradiation) cells1 Week, named as the unsorted group; in the second group, 1×10 7 cells were sorted according to the method described in Example 10. After sorting, the remaining cells were selected as NK cells:artificial antigen-presenting cells=1:2 and K562 -NK1 (100Gy γ-ray irradiation) was co-cultured for 1 week, named Biotin Anti-Strep tag II Ab+anti-Biotin microbeads group; 1×10 7 cells were taken from the third group and resuspended in 90 μL DPBS+1% AB serum Add 10 μL of Streptavidin microbeads to the buffer, mix well and incubate at 2-8 °C for 20 min; after incubation, add 5 mL of buffer, centrifuge at 300 × g for 10 min, discard the supernatant and resuspend in 500 μL of buffer, and then proceed with magnetic beads The labeled positive cells were sorted, and the remaining cells were taken after sorting and co-cultured with K562-NK1 (100Gy γ-ray irradiation) for 1 week according to NK cells:artificial antigen-presenting cells=1:2, named Streptavidin microbeads group.
结果如下表1所示,未分选组第二轮共培养结束第17天,NK细胞中CAR+细胞比例为33.1%,第二轮扩增倍数为10.2倍;Biotin Anti-Strep tag II Ab+anti-Biotin microbeads组分选后细胞得率为66%(100%×分选后得到的细胞/(1×10 7×CAR+%)),第二轮共培养结束第17天,NK细胞中CAR+细胞比例为95.3%,第二轮扩增倍数为44.64倍;Streptavidin microbeads组分选后细胞得率为10.6%,第二轮共培养结束第17天,NK细胞中CAR+细胞比例为97.8%,第二轮扩增倍数为20.65倍。 The results are shown in Table 1 below. On the 17th day after the end of the second round of co-culture in the unsorted group, the proportion of CAR+ cells in the NK cells was 33.1%, and the expansion fold of the second round was 10.2 times; Biotin Anti-Strep tag II Ab+anti -Biotin cell yield of 66% after the component is selected from microbeads (100% × after sorting the resulting cell / (1 × 10 7 × CAR +%)), the end of the second round of co-culture on day 17, NK cells are CAR + cells The ratio was 95.3%, and the expansion fold in the second round was 44.64 times; the cell yield of the Streptavidin microbeads group was 10.6%, and on the 17th day after the second round of co-culture, the proportion of CAR+ cells in the NK cells was 97.8%. The round amplification factor was 20.65 times.
表1Table 1
Figure PCTCN2021103235-appb-000002
Figure PCTCN2021103235-appb-000002
根据上述结果,利用Biotin Anti-Strep tag II Ab+anti-Biotin microbeads分选的得率较高,且可以使扩增一轮后NK细胞中CAR+的细胞比例提高至90%以上。According to the above results, the sorting yield of Biotin Anti-Strep tag II Ab+anti-Biotin microbeads is higher, and the proportion of CAR+ cells in NK cells after one round of expansion can be increased to more than 90%.
实施例12在CAR的载体中加入分泌型或膜结合型的人IL-15可促进CAR-NK细胞在体外的存活Example 12 Adding secreted or membrane-bound human IL-15 to the CAR carrier can promote the survival of CAR-NK cells in vitro
本实施例在含有NKG2D CAR的转座子供体中加入人IL-15的基因,构建了质粒5,并以PBMC为来源分别制备了NK、NKG2D CAR-NK和可以分泌IL-15的NKG2D eCAR-NK(NKG2D eCAR-IL15 NK)。In this example, the gene of human IL-15 was added to the transposon donor containing NKG2D CAR, plasmid 5 was constructed, and NK, NKG2D CAR-NK and NKG2D eCAR-NK that could secrete IL-15 were prepared from PBMC. NK (NKG2D eCAR-IL15 NK).
其中NKG2D CAR-NK的制备参考实施例3;NKG2D eCAR-IL15 NK的制备参考实施例7,将质粒4替换为质粒5,由于CAR载体中含有IL-15,第17天的细胞因子组合物中去除IL-15。取第三轮共培养结束第22天的细胞每组各2×10 6,分别在添加和不添加hrIL-2(50IU/mL)的条件下体外培养1周后计数,计算细胞的变化百分比(100%×培养1周后的细胞数/2×10 6)。 The preparation of NKG2D CAR-NK refers to Example 3; the preparation of NKG2D eCAR-IL15 NK refers to Example 7, and plasmid 4 is replaced with plasmid 5. Since the CAR vector contains IL-15, the cytokine composition on the 17th day IL-15 was removed. Take 2 × 10 6 cells in each group on the 22nd day after the third round of co-culture, and count them after 1 week of in vitro culture with and without addition of hrIL-2 (50 IU/mL), and calculate the percentage change of cells ( 100%×the number of cells after 1 week of culture/2×10 6 ).
如图14A所示,在不添加IL-2的情况下,NK和NKG2D CAR-NK的细胞变化百分比为3%和7%;加入IL-15后,CAR-NK的细胞变化百分比提高至37%;在添加IL-2后,所有组的细胞存活都提高。As shown in Figure 14A, the percentage of cell change in NK and NKG2D CAR-NK was 3% and 7% without the addition of IL-2; after the addition of IL-15, the percentage of cell change in CAR-NK increased to 37% ; After the addition of IL-2, cell survival was improved in all groups.
本实施例中以脐带血为来源分别制备了NK、NKG2D eCAR-IL15 NK和NKG2D eCAR-IL15 NK (sorted)。其中,NKG2D eCAR-IL15 NK的制备参考实施例7,将质粒4替换为质粒5,由于CAR载体中含有IL-15,第17天的细胞因子组合物中去除IL-15;NKG2D eCAR-IL15 NK(sorted)的制备参考实施例10,将质粒4替换为质粒5,由于CAR载体中含有IL-15,第17天的细胞因子组合物中去除IL-15。In this example, NK, NKG2D eCAR-IL15 NK and NKG2D eCAR-IL15 NK (sorted) were prepared from umbilical cord blood, respectively. Among them, for the preparation of NKG2D eCAR-IL15 NK, refer to Example 7, and replace plasmid 4 with plasmid 5. Since the CAR vector contains IL-15, IL-15 was removed from the cytokine composition on day 17; NKG2D eCAR-IL15 NK (sorted) Preparation Refer to Example 10, replace plasmid 4 with plasmid 5, since IL-15 is contained in the CAR vector, IL-15 is removed from the cytokine composition on day 17.
取第三轮共培养结束第21天的细胞每组各5×10 5,分别在添加和不添加hrIL-2(50IU/mL)的条件下体外培养1周后计数,计算细胞的变化百分比(100%×培养1周后的细胞数/5×10 5)。 Take 5 × 10 5 cells in each group on the 21st day after the third round of co-culture, and count them after 1 week of in vitro culture with and without hrIL-2 (50 IU/mL), and calculate the percentage change of cells ( 100%×the number of cells after 1 week of culture/5×10 5 ).
如图14B所示,在不添加IL-2的情况下,NK、NKG2D eCAR-IL15 NK、NKG2D eCAR-IL15 NK(sorted)的细胞变化百分比分别为25%、47%和316%,含有IL-15并经分选的高纯度的CAR-NK细胞甚至呈现了增殖。在添加IL-2的情况下,所有组的细胞存活都提高。As shown in Figure 14B, in the absence of IL-2 addition, the percentage of cell change in NK, NKG2D eCAR-IL15 NK, and NKG2D eCAR-IL15 NK(sorted) were 25%, 47%, and 316%, respectively, with IL- 15 and sorted high-purity CAR-NK cells even exhibited proliferation. With the addition of IL-2, cell survival was improved in all groups.
本实施例也尝试在含有NKG2D CAR的转座子供体加入膜结合的人IL-15的基因,构建了质粒6,并以脐带血为来源分别制备了NK、NKG2D eCAR-NK(sorted)和NKG2D eCAR-mbIL15 NK(sorted)。其中NKG2D eCAR-NK(sorted)和NKG2D eCAR-mbIL15 NK(sorted)的制备均参考实施例10。NKG2D eCAR-mbIL15 NK(sorted)的制备中将质粒4替换为质粒6,由于CAR载体中含有mbIL-15,第17天的细胞因子组合物中去除IL-15。In this example, we also tried to add the membrane-bound human IL-15 gene to the transposon donor containing NKG2D CAR, constructed plasmid 6, and prepared NK, NKG2D eCAR-NK(sorted) and NKG2D respectively from umbilical cord blood eCAR-mbIL15 NK(sorted). The preparations of NKG2D eCAR-NK (sorted) and NKG2D eCAR-mbIL15 NK (sorted) refer to Example 10. Plasmid 4 was replaced with plasmid 6 in the preparation of NKG2D eCAR-mbIL15 NK (sorted), since the CAR vector contained mbIL-15, IL-15 was removed from the cytokine composition on day 17.
取第三轮共培养结束第21天的细胞每组各2×10 6,在不添加hrIL-2(50IU/mL)的条件下体外培养1周后计数,计算细胞的变化百分比(100%×培养1周后的细胞数/2×10 6)。 Take 2×10 6 cells in each group on the 21st day after the third round of co-culture, culture them in vitro for 1 week without adding hrIL-2 (50 IU/mL), and count the percentage of cell change (100%× The number of cells/2×10 6 after culturing for 1 week).
如图14C所示,在不添加IL-2的情况下,NK、NKG2D eCAR-NK(sorted)、NKG2D eCAR-mbIL15 NK(sorted)的细胞变化百分比分别为6%、13%和140%,膜结合IL-15(mbIL-15)同样可以促进CAR-NK的体外存活。As shown in Figure 14C, in the absence of IL-2 addition, the percentage of cell change in NK, NKG2D eCAR-NK(sorted), NKG2D eCAR-mbIL15 NK(sorted) were 6%, 13%, and 140%, respectively, and the membrane Binding of IL-15 (mbIL-15) can also promote the survival of CAR-NK in vitro.
本实施例同时以PBMC为来源分别制备了NK、NKG2D CAR-mbIL15 NK和NKG2D eCAR-mbIL15 NK(sorted)。其中NKG2D CAR-mbIL15 NK的制备参考实施例3,将质粒4替换为质粒6,由于CAR载体中含有mbIL-15,D17天的细胞因子组合物中去除IL-15;NKG2D eCAR-mbIL15 NK(sorted)的制备参考实施例10,将质粒4替换为质粒6,由于CAR载体中含有mbIL-15,第17天的细胞因子组合物中去除IL-15。In this example, NK, NKG2D CAR-mbIL15 NK and NKG2D eCAR-mbIL15 NK (sorted) were prepared from PBMCs, respectively. The preparation of NKG2D CAR-mbIL15 NK refers to Example 3, and plasmid 4 is replaced with plasmid 6. Since the CAR vector contains mbIL-15, IL-15 is removed from the cytokine composition on day 17; NKG2D eCAR-mbIL15 NK (sorted ) Preparation of ) Refer to Example 10, replace plasmid 4 with plasmid 6, since mbIL-15 is contained in the CAR vector, IL-15 is removed from the cytokine composition on day 17.
取第二轮共培养结束第16天的细胞每组各2×10 6,在不添加hrIL-2(50IU/mL)的条件下体外培养1周后计数,计算细胞的变化百分比(100%×培养1周后的细胞数/2×10 6)。 Take 2×10 6 cells in each group on the 16th day after the second round of co-culture, and count them after 1 week of in vitro culture without adding hrIL-2 (50 IU/mL), and calculate the percentage change of cells (100%× The number of cells/2×10 6 after culturing for 1 week).
如图14D所示,在不添加IL-2的情况下,NK、NKG2D CAR-mbIL15 NK、NKG2D eCAR-mbIL15 NK(sorted)的细胞变化百分比分别为27%、62%和114%,膜结合IL-15(mbIL-15)同样可以促进CAR-NK的体外存活,含有mbIL-15并经分选的高纯度CAR-NK在体外甚至呈现了少量的增殖。As shown in Figure 14D, in the absence of IL-2, the percentage of cell change in NK, NKG2D CAR-mbIL15 NK, NKG2D eCAR-mbIL15 NK(sorted) was 27%, 62%, and 114%, respectively, and membrane-bound IL -15 (mbIL-15) can also promote the survival of CAR-NK in vitro, and the sorted high-purity CAR-NK containing mbIL-15 even showed a small amount of proliferation in vitro.
实施例13分选后高纯度的NKG2D CAR-mbIL15 NK在体外对肿瘤细胞株展现了较强的杀伤作用Example 13 High-purity NKG2D CAR-mbIL15 NK after sorting showed strong killing effect on tumor cell lines in vitro
本实施例以PBMC为来源分别制备了NK、NKG2D eCAR-mIL15 NK和NKG2D eCAR-mbIL15 NK (sorted),其中NKG2D eCAR-mbIL15 NK的制备参考实施例7,将质粒4替换为质粒6,由于CAR载体中含有mbIL-15,第17天的细胞因子组合物中去除IL-15;NKG2D eCAR-mbIL15 NK(sorted)的制备参考实施例10,将质粒4替换为质粒6,由于CAR载体中含有mbIL-15,第17天的细胞因子组合物中去除IL-15。In this example, NK, NKG2D eCAR-mIL15 NK and NKG2D eCAR-mbIL15 NK (sorted) were prepared respectively with PBMC as the source, wherein the preparation of NKG2D eCAR-mbIL15 NK refers to Example 7, and plasmid 4 was replaced with plasmid 6, because CAR The vector contains mbIL-15, and IL-15 is removed from the cytokine composition on day 17; the preparation of NKG2D eCAR-mbIL15 NK (sorted) refers to Example 10, and plasmid 4 is replaced with plasmid 6, because the CAR vector contains mbIL -15, Depletion of IL-15 in the cytokine composition on day 17.
取第二轮共培养结束第16天的细胞进行体外杀伤试验。取人AML细胞株KG1细胞按1×10 6个细胞/ml重悬于DPBS中,加入Calcein-AM(终浓度0.2μM)于37℃进行染色15min。染色结束后按等体积加入FBS终止染色,并用DPBS洗3遍。将染色后的KG1重悬于培养基中,调整细胞密度至2×10 5个细胞/ml,于U形96孔板中每孔100μL进行KG1铺板。将效应细胞按照E:T=2:1、1:1和0.5:1接种于96孔板,每孔100μL,每孔总体积200μL。于37℃孵育过夜后,第二天通过流式检测每孔中GFP+的细胞数。根据以下公式计算肿瘤生长抑制率: The cells on the 16th day after the second round of co-culture were taken for in vitro killing test. The human AML cell line KG1 cells were taken and resuspended in DPBS at 1×10 6 cells/ml, and Calcein-AM (final concentration 0.2 μM) was added for staining at 37° C. for 15 min. After staining, FBS was added in an equal volume to stop the staining, and washed three times with DPBS. The stained KG1 was resuspended in the medium, the cell density was adjusted to 2×10 5 cells/ml, and KG1 was plated in 100 μL per well in a U-shaped 96-well plate. Effector cells were seeded in 96-well plates according to E:T=2:1, 1:1 and 0.5:1, 100 μL per well, and the total volume per well was 200 μL. After overnight incubation at 37°C, the number of GFP+ cells per well was determined by flow cytometry the next day. The tumor growth inhibition rate was calculated according to the following formula:
100%×(单KG1细胞组的GFP+细胞数-实验组GFP+细胞数)/单KG1细胞组的GFP+细胞数100%×(the number of GFP+ cells in the single KG1 cell group - the number of GFP+ cells in the experimental group)/the number of GFP+ cells in the single KG1 cell group
如图15A所示,对比于NK,NKG2D eCAR-mIL15 NK和NKG2D eCAR-mbIL15 NK(sorted)都对KG1展示出显著的肿瘤杀伤作用,且NKG2D eCAR-mbIL15 NK(sorted)对KG1的杀伤略强。As shown in Figure 15A, compared with NK, both NKG2D eCAR-mIL15 NK and NKG2D eCAR-mbIL15 NK(sorted) showed significant tumor killing effect on KG1, and NKG2D eCAR-mbIL15 NK(sorted) was slightly stronger on KG1 .
本实施例还通过RTCA实时杀伤检测仪(具体实验信息参考实施例5)检测了NKG2D eCAR-mbIL15 NK(sorted)在体外对人结直肠癌细胞株HCT116的杀伤作用;In this example, the killing effect of NKG2D eCAR-mbIL15 NK (sorted) on human colorectal cancer cell line HCT116 in vitro was also detected by RTCA real-time killing detector (refer to Example 5 for specific experimental information);
如图15B所示,在E:T=0.5:1时,NK只能减缓肿瘤的生长,NKG2D eCAR-mbIL15 NK(sorted)对HCT116展示出非常显著的杀伤作用。As shown in Figure 15B, when E:T=0.5:1, NK could only slow down the growth of tumor, and NKG2D eCAR-mbIL15 NK(sorted) showed a very significant killing effect on HCT116.
实施例14制备工程化K562-NK2细胞Example 14 Preparation of engineered K562-NK2 cells
将K562-NK1细胞重悬于10mL Opti-MEM中,300×g离心10min,将细胞沉淀重悬于100μL P3 buffer中,加入5μg质粒2和10μg质粒9混匀后转移至Lonza电击杯;将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转。The K562-NK1 cells were resuspended in 10 mL Opti-MEM, centrifuged at 300 × g for 10 min, the cell pellet was resuspended in 100 μL P3 buffer, 5 μg plasmid 2 and 10 μg plasmid 9 were added, and the cells were transferred to the Lonza electric shock cup; The cups were placed in a Lonza 4D-NucleofectorTM X Unit (in a single shock cup module) for electroporation.
电转结束后,将一个电击杯中的K562细胞悬液缓慢转移至6孔板的一个孔中,孔里预先加入K562培养基(IMDM+10%FBS);电穿孔后的第7天,使用细胞分选仪BD Fusion(BD Biosciences公司)将单细胞分选到96孔板,对扩增的单细胞克隆进行流式细胞术分析,检测BCMA的表达情况,检测抗体为APC-结合的抗人BMCA抗体,筛选到的单细胞克隆命名为K562-NK2。After the electroporation, the K562 cell suspension in an electroporation cup was slowly transferred to a well of a 6-well plate, and K562 medium (IMDM+10% FBS) was pre-added to the well; on the 7th day after electroporation, the cells were used The sorting instrument BD Fusion (BD Biosciences) sorts single cells into 96-well plates, and analyzes the amplified single cell clones by flow cytometry to detect the expression of BCMA. The detection antibody is APC-conjugated anti-human BMCA. Antibody, the screened single cell clone was named K562-NK2.
如图16所示,K562-NK2细胞表面表达了高水平BCMA,其阳性率为99.4%。As shown in Figure 16, K562-NK2 cells expressed high levels of BCMA on the surface, with a positive rate of 99.4%.
实施例15利用piggyBac转座子系统制备BCMA eCAR-NKExample 15 Preparation of BCMA eCAR-NK using piggyBac transposon system
第0天,取2×10 7个PBMC细胞,用CD3磁珠(Miltenyi)按照厂家的推荐步骤进行CD3+细胞去除,剩余约1×10 7个PBMC;将5×10 6个分选后的PBMC和5×10 6K562-NK2(100Gyγ射线辐照)重悬于10mL NK细胞培养基,接种于T25细胞培养瓶中,加入hrIL-2使其终浓度为50IU/mL; On day 0, 2 × 10 7 PBMC cells were taken, and CD3+ cells were removed using CD3 magnetic beads (Miltenyi) according to the manufacturer's recommended procedure, and about 1 × 10 7 PBMCs remained; 5 × 10 6 sorted PBMCs were removed and 5×10 6 K562-NK2 (100Gy γ-ray irradiation) were resuspended in 10mL NK cell culture medium, inoculated into T25 cell culture flask, and hrIL-2 was added to make the final concentration 50IU/mL;
第2天,取出悬浮的细胞进行计数,300×g离心10min;用10mL Opti-MEM重悬细胞沉淀,300×g离心10min;将细胞沉淀重悬于100μL P3 buffer中,加入5μg质粒2和10μg质粒8,混匀后转移至电Lonza电击杯;将电击杯置于Lonza 4D-NucleofectorTM X Unit(单电击杯模块中),进行电转,电转结束后,将一个电击杯中的NK细胞悬液缓慢转移至一个新的T25细胞培养瓶中,加入含有终浓度为50IU/mL的hrIL-2的10mL NK细胞培养液,轻轻混匀,将T25细胞培养瓶置于37℃细胞培养箱培养;On the second day, the suspended cells were taken out for counting, and centrifuged at 300 × g for 10 min; resuspended the cell pellet with 10 mL Opti-MEM and centrifuged at 300 × g for 10 min; resuspended the cell pellet in 100 μL P3 buffer, and added 5 μg plasmid 2 and 10 μg Plasmid 8, after mixing, transfer to the electroporation Lonza electroporation cup; place the electroporation cup in the Lonza 4D-NucleofectorTM X Unit (in a single electroporation cup module), and perform electroporation. After the electroporation, slowly transfer the NK cell suspension in one electroporation cup Transfer to a new T25 cell culture flask, add 10 mL of NK cell culture medium containing hrIL-2 with a final concentration of 50IU/mL, mix gently, and place the T25 cell culture flask in a 37°C cell incubator for cultivation;
第3天至第9天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 3rd day to the 9th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第10天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);取出1×10 7的BCMA CAR-NK细胞,重悬于80μL DPBS+1%AB血清的缓冲液中,加入20μL anti-Strep tag II抗体,混匀后于2~8℃孵育20min;孵育完成后加入5mL缓冲液,300×g离心10min,弃上清并重悬于80μL DPBS+1%AB血清的缓冲液中,加入20μL anti-Biotin磁珠,混匀后于2~8℃孵育15min;孵育完成后加入5mL缓冲液,300×g离心10min,弃上清并重悬于500μL缓冲液按照(Miltenyi)厂家的推荐步骤进行磁珠标记的阳性细胞的分选。取2×10 6分选后磁珠标记CAR-NK细胞与4×10 6个K562-NK2(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-2使其终浓度为50IU/mL。 On the 10th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 1×10 7 BCMA was taken out CAR-NK cells were resuspended in 80 μL DPBS+1% AB serum buffer, 20 μL anti-Strep tag II antibody was added, mixed and incubated at 2-8 °C for 20 min; after incubation, 5 mL buffer was added, 300× Centrifuge at g for 10 min, discard the supernatant and resuspend in 80 μL DPBS+1% AB serum buffer, add 20 μL anti-Biotin magnetic beads, mix well and incubate at 2-8 °C for 15 min; after the incubation, add 5 mL buffer, 300 After centrifugation at ×g for 10 min, the supernatant was discarded and resuspended in 500 μL buffer to sort the positive cells labeled with magnetic beads according to the procedure recommended by the manufacturer (Miltenyi). Take 2 × 10 6 magnetic beads-labeled CAR-NK cells and 4 × 10 6 K562-NK2 (100Gy γ-ray irradiated) and resuspend them in 10 mL of medium after sorting, and add hrIL-2 to make the final concentration 50IU/mL .
第11天至第16天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 11th day to the 16th day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第17天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体);将2×10 6个细胞与2×10 6个K562-NK2(100Gyγ射线辐照)重悬于10mL培养基中,加入hrIL-12、hrIL-15和hrIL-18使其终浓度分别为10ng/mL、50ng/mL和50ng/mL; On the 17th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, anti-Strep-tag II antibody); 2×10 6 cells were Resuspend in 10 mL medium with 2×10 6 K562-NK2 (100 Gy γ-irradiated), and add hrIL-12, hrIL-15 and hrIL-18 to make the final concentrations 10ng/mL, 50ng/mL and 50ng, respectively /mL;
第18天,收集全部细胞收集300×g离心10min,用20mL新鲜的NK细胞培养基重悬,添加hrIL-2使其终浓度50IU/mL;On the 18th day, all cells were collected, centrifuged at 300 × g for 10 min, resuspended in 20 mL of fresh NK cell culture medium, and hrIL-2 was added to make the final concentration 50 IU/mL;
第18天至第23天,根据细胞生长情况,加入适量新鲜的含有hrIL-2的NK细胞培养液;From the 18th day to the 23rd day, according to the cell growth, add an appropriate amount of fresh NK cell culture medium containing hrIL-2;
第24天,收集全部细胞进行计数,取2×10 6个细胞进行流式细胞表型分析(抗人CD3抗体、抗人CD56抗体、抗Strep-tag II抗体)。 On the 24th day, all cells were collected and counted, and 2×10 6 cells were taken for flow cytometric phenotype analysis (anti-human CD3 antibody, anti-human CD56 antibody, and anti-Strep-tag II antibody).
如图17A所示,第一轮共培养结束,CD3-CD56+的NK细胞比例达到90%左右,且在接下来几轮与K562-NK2(100Gyγ射线辐照)共培养后,NK比例基本不变。如图17B所示,第一轮共培养结束,CAR+的细胞比例为16%;第二轮共培养结束,CAR+的细胞比例为57.6%;第三轮共培养结束,CAR+的细胞比例为72.6%。如图17C所示,第一轮共培养的扩增倍数为13倍,分选后第二轮共培养的扩增倍数为60倍,第三轮共培养的扩增倍数为46.5倍。As shown in Figure 17A, after the first round of co-culture, the proportion of CD3-CD56+ NK cells reached about 90%, and after the next few rounds of co-culture with K562-NK2 (100Gy γ-ray irradiation), the proportion of NK cells remained basically unchanged. . As shown in Figure 17B, the first round of co-culture ended, the proportion of CAR+ cells was 16%; the second round of co-culture ended, the proportion of CAR+ cells was 57.6%; the third round of co-culture ended, the proportion of CAR+ cells was 72.6% . As shown in Figure 17C, the expansion fold of the first round of co-culture was 13 times, the second round of co-culture after sorting was 60 times, and the third round of co-culture was 46.5 times.
图18为一个健康捐献者来源的PBMC制备BCMA eCAR-NK的过程中,在第1、2、3轮共培养结束时,每一轮总细胞数的扩增倍数,第一轮共培养的扩增倍数为13倍,第二轮共培养的扩增倍数为60倍,第三轮共培养的扩增倍数为46倍。Figure 18 shows the expansion fold of the total number of cells in each round at the end of the first, second, and third rounds of co-culture during the preparation of BCMA eCAR-NK from a healthy donor-derived PBMC, and the expansion of the first round of co-culture. The multiplication factor was 13 times, the second round of co-culture was 60 times, and the third round of co-culture was 46 times.
本实施例中还检测了第三轮共培养结束第22天的BCMA eCAR-NK细胞体外对人MM细胞株U266的杀伤。In this example, the killing of human MM cell line U266 in vitro by BCMA eCAR-NK cells on the 22nd day after the third round of co-culture was also detected.
取U266细胞按1×10 6个细胞/mL重悬于DPBS中,加入Calcein-AM(终浓度0.2μM)于37℃进行染色15min。染色结束后按等体积加入FBS终止染色,并用DPBS洗3遍。将染色后的U266细胞重悬于培养基中,调整细胞密度至2×10 5个细胞/mL,于U形96孔板中每孔100μL进行U266铺板。将效应细胞按照E:T=5:1、2.5:1和1.25:1接种于96孔板,每孔100μL,每孔总体积200μL。于37℃孵育过夜后,第二天通过流式检测每孔中GFP+的细胞数。根据以下公式计算肿瘤生长抑制率: U266 cells were taken and resuspended in DPBS at 1×10 6 cells/mL, and Calcein-AM (final concentration 0.2 μM) was added for staining at 37° C. for 15 min. After staining, FBS was added in an equal volume to stop the staining, and washed three times with DPBS. The stained U266 cells were resuspended in the medium, the cell density was adjusted to 2×10 5 cells/mL, and 100 μL per well of U266 was plated in a U-shaped 96-well plate. Effector cells were seeded in 96-well plates according to E:T=5:1, 2.5:1 and 1.25:1, 100 μL per well, and the total volume per well was 200 μL. After overnight incubation at 37°C, the number of GFP+ cells per well was detected by flow cytometry the next day. Tumor growth inhibition rate was calculated according to the following formula:
100%×(单U266细胞组的GFP+细胞数-实验组GFP+细胞数)/单U266细胞组的GFP+细胞数100%×(the number of GFP+ cells in the single U266 cell group - the number of GFP+ cells in the experimental group)/the number of GFP+ cells in the single U266 cell group
如图19所示,对比于NK,BCMA eCAR-NK(sorted)对U266展示出显著的肿瘤杀伤作用。As shown in Figure 19, compared to NK, BCMA eCAR-NK (sorted) exhibited significant tumor killing effect on U266.
综上所述,本申请采用非病毒的方法,利用转座子系统在NK细胞中引入稳定高表达的转基因,实现对NK细胞的高效遗传修饰,生产周期短、生产成本低,制备的转基因修饰NK细胞纯度高、安全性好,对肿瘤细胞具有显著的杀伤和/或抑制作用。To sum up, the present application adopts a non-viral method and uses a transposon system to introduce a stable and highly expressed transgene into NK cells to achieve efficient genetic modification of NK cells. The production cycle is short and the production cost is low. The prepared transgenic modification NK cells have high purity and good safety, and have significant killing and/or inhibitory effects on tumor cells.
申请人声明,本申请通过上述实施例来说明本申请的详细方法,但本申请并不局限于上述详细方法,即不意味着本申请必须依赖上述详细方法才能实施。所属技术领域的技术人员应该明了,对本申请的任何改进,对本申请产品各原料的等效替换及辅助成分的添加、具体方式的选择等,均落在本申请的保护范围和公开范围之内。The applicant declares that the present application illustrates the detailed method of the present application through the above-mentioned embodiments, but the present application is not limited to the above-mentioned detailed method, which does not mean that the present application must rely on the above-mentioned detailed method for implementation. Those skilled in the art should understand that any improvement to the application, the equivalent replacement of each raw material of the product of the application, the addition of auxiliary components, the selection of specific methods, etc., all fall within the scope of protection and disclosure of the application.
本文所涉及基因序列的列表List of gene sequences covered in this paper
Figure PCTCN2021103235-appb-000003
Figure PCTCN2021103235-appb-000003

Claims (27)

  1. 一种利用非病毒转座子系统和人工抗原呈递细胞制备嵌合受体修饰的自然杀伤细胞的方法,其包括:A method for preparing natural killer cells modified by chimeric receptors using a non-viral transposon system and artificial antigen-presenting cells, comprising:
    (1)利用转座子系统将嵌合受体的编码基因转入自然杀伤细胞,得到初始的嵌合受体修饰的自然杀伤细胞;(1) using the transposon system to transfer the encoding gene of the chimeric receptor into natural killer cells to obtain the original natural killer cells modified by the chimeric receptor;
    (2)利用人工抗原呈递细胞扩增所述初始的嵌合受体修饰的自然杀伤细胞。(2) Using artificial antigen-presenting cells to expand the original chimeric receptor-modified natural killer cells.
  2. 根据权利要求1所述的方法,其中,所述自然杀伤细胞包括人原代自然杀伤细胞;The method of claim 1, wherein the natural killer cells comprise human primary natural killer cells;
    所述人原代自然杀伤细胞来源于外周血、脐带血、胎盘组织或诱导性多能干细胞。The primary human natural killer cells are derived from peripheral blood, umbilical cord blood, placental tissue or induced pluripotent stem cells.
  3. 根据权利要求1所述的方法,其中,所述嵌合受体包括嵌合抗原受体和/或嵌合转换受体。The method of claim 1, wherein the chimeric receptor comprises a chimeric antigen receptor and/or a chimeric switch receptor.
  4. 根据权利要求1所述的方法,其中,步骤(1)所述转座子系统包括PiggyBac转座子系统、Sleeping Beauty转座子系统或Tol2转座子系统。The method according to claim 1, wherein the transposon system of step (1) comprises a PiggyBac transposon system, a Sleeping Beauty transposon system or a Tol2 transposon system.
  5. 根据权利要求1所述的方法,其中,步骤(1)所述转座子系统包括转座子元件和转座酶元件。The method according to claim 1, wherein the transposon system in step (1) comprises a transposon element and a transposase element.
  6. 根据权利要求5所述的方法,其中,所述转座子元件包括转座子5’反向末端重复序列和3’反向末端重复序列;The method of claim 5, wherein the transposon element comprises a transposon 5' inverted terminal repeat and a 3' inverted terminal repeat;
    所述5’反向末端重复序列和3’反向末端重复序列之间含有两个绝缘子;There are two insulators between the 5' inverted terminal repeat and the 3' inverted terminal repeat;
    所述两个绝缘子之间含有启动子和嵌合受体的编码基因;A gene encoding a promoter and a chimeric receptor is contained between the two insulators;
    所述转座子元件为质粒或线性化核酸片段。The transposon element is a plasmid or a linearized nucleic acid fragment.
  7. 根据权利要求5所述的方法,其中,所述转座酶元件包括转座酶蛋白或编码转座酶的核酸分子;The method of claim 5, wherein the transposase element comprises a transposase protein or a nucleic acid molecule encoding a transposase;
    所述编码转座酶的核酸分子包括质粒、线性化核酸片段或mRNA。The nucleic acid molecules encoding transposases include plasmids, linearized nucleic acid fragments, or mRNA.
  8. 根据权利要求5所述的方法,其中,所述转座子元件和转座酶元件连接在同一表达载体上。The method of claim 5, wherein the transposon element and the transposase element are linked to the same expression vector.
  9. 根据权利要求5所述的方法,其中,所述转座子元件和转座酶元件在不同的表达载体上。6. The method of claim 5, wherein the transposon element and the transposase element are on different expression vectors.
  10. 根据权利要求6所述的方法,其特征在于,所述转座子元件还包括促进NK细胞存活的细胞因子的编码基因,所述细胞因子包括IL-15或IL-21;The method of claim 6, wherein the transposon element further comprises a gene encoding a cytokine that promotes NK cell survival, the cytokine comprising IL-15 or IL-21;
    所述细胞因子包括分泌型、膜结合型或细胞因子连接受体形成的组合型;The cytokines include secreted, membrane-bound or a combination of cytokine-linked receptors;
    所述细胞因子的编码基因与嵌合受体的编码基因在同一转座子表达载体中或在不同的转座子表达载体中。The encoding gene of the cytokine and the encoding gene of the chimeric receptor are in the same transposon expression vector or in different transposon expression vectors.
  11. 根据权利要求1所述的方法,其中,步骤(1)所述转座子系统通过电穿孔和/或化学试剂转入自然杀伤细胞。The method according to claim 1, wherein the transposon system in step (1) is transferred into natural killer cells by electroporation and/or chemical reagents.
  12. 根据权利要求1所述的方法,其中,在步骤(1)之前还包括活化自然杀伤细胞的步骤。The method according to claim 1, further comprising a step of activating natural killer cells before step (1).
  13. 根据权利要求12所述的方法,其中,所述自然杀伤细胞活化0~14天后,将所述转座子系统转入自然杀伤细胞。The method according to claim 12, wherein the transposon system is transferred into natural killer cells after 0-14 days of activation of the natural killer cells.
  14. 根据权利要求12所述的方法,其中,所述活化自然杀伤细胞采用人工抗原呈递细胞、饲养细胞、细胞因子、抗体或化合物进行。The method of claim 12, wherein the activation of natural killer cells is performed using artificial antigen presenting cells, feeder cells, cytokines, antibodies or compounds.
  15. 根据权利要求1或14所述的方法,其中,所述人工抗原呈递细胞包括以细胞为基础的人工抗原呈递细胞、人工合成的人工抗原呈递细胞或以外泌体为基础的人工抗原呈递细胞。The method according to claim 1 or 14, wherein the artificial antigen-presenting cells comprise cell-based artificial antigen-presenting cells, artificially synthesized artificial antigen-presenting cells or exosome-based artificial antigen-presenting cells.
  16. 根据权利要求15所述的方法,其中,所述以细胞为基础的人工抗原呈递细胞包括人髓系白血病K562细胞、人伯基特淋巴瘤Daudi细胞、EBV转化的B淋巴母细胞样细胞或小鼠胚胎成纤维细胞系NIH/3T3细胞。The method of claim 15, wherein the cell-based artificial antigen presenting cells comprise human myeloid leukemia K562 cells, human Burkitt lymphoma Daudi cells, EBV transformed B lymphoblastoid cells or small cells The mouse embryonic fibroblast cell line NIH/3T3 cells.
  17. 根据权利要求16所述的方法,其中,所述以细胞为基础的人工抗原呈递细胞为工程化细胞;The method of claim 16, wherein the cell-based artificial antigen presenting cell is an engineered cell;
    所述工程化细胞表达嵌合受体识别的抗原;the engineered cells express an antigen recognized by the chimeric receptor;
    所述工程化细胞表达用于活化自然杀伤细胞的配体分子和/或细胞因子;The engineered cells express ligand molecules and/or cytokines for activating natural killer cells;
    所述配体分子包括CD137L和/或OX40L;The ligand molecule includes CD137L and/or OX40L;
    所述细胞因子包括人IL-21、人IL-15或人IL-18中的任意一种或至少两种的组合;The cytokine comprises any one or a combination of at least two of human IL-21, human IL-15 or human IL-18;
    所述细胞因子包括分泌型细胞因子和/或膜结合细胞因子;The cytokines include secreted cytokines and/or membrane-bound cytokines;
    所述工程化细胞经γ射线或丝裂霉素C处理。The engineered cells were treated with gamma rays or mitomycin C.
  18. 根据权利要求1所述的方法,其中,将嵌合受体的编码基因转入自然杀伤细胞0~14天后,利用人工抗原呈递细胞扩增所述初始的嵌合受体修饰的自然杀伤细胞。The method according to claim 1, wherein the initial chimeric receptor-modified natural killer cells are expanded by using artificial antigen-presenting cells 0 to 14 days after the gene encoding the chimeric receptor is transferred into the natural killer cells.
  19. 根据权利要求1所述的方法,其中,所述扩增进行多次,每一轮扩增周期为3~14天。The method according to claim 1, wherein the amplification is performed multiple times, and each round of amplification cycle is 3-14 days.
  20. 根据权利要求1所述的方法,其中,步骤(1)还包括磁分选富集NK细胞或去除CD3+细胞的步骤。The method according to claim 1, wherein step (1) further comprises a step of magnetic sorting to enrich NK cells or remove CD3+ cells.
  21. 根据权利要求20所述的方法,其中,所述磁分选的步骤在将嵌合受体的编码基因转入自然杀伤细胞之前和/或在所述初始的嵌合受体修饰的自然杀伤细胞培养0~14天后进行。The method according to claim 20, wherein the step of magnetic sorting is before the transfer of the chimeric receptor-encoding gene into natural killer cells and/or before the initial chimeric receptor-modified natural killer cells The culture was carried out after 0 to 14 days.
  22. 根据权利要求1所述的方法,其中,在步骤(1)之后还包括CAR+细胞分选的操作。The method according to claim 1, wherein after step (1), the operation of CAR+ cell sorting is further included.
  23. 根据权利要求1所述的方法,其中,步骤(2)之后还包括利用磁分选富集NK细胞或去除CD3+细胞的步骤。The method according to claim 1, wherein after step (2), it further comprises the step of enriching NK cells or removing CD3+ cells by magnetic sorting.
  24. 根据权利要求1所述的方法,其中,在步骤(2)之后还包括CAR+细胞分选的操作。The method according to claim 1, wherein after step (2), the operation of sorting CAR+ cells is further included.
  25. 根据权利要求1所述的方法,其中,所述嵌合受体修饰的自然杀伤细胞的制备过程中,培养基中添加有细胞因子组合物,所述细胞因子组合物为hrIL-12和hrIL-18的组合,或者为hrIL-12、hrIL-15和hrIL-18的组合。The method according to claim 1, wherein, during the preparation of the chimeric receptor-modified natural killer cells, a cytokine composition is added to the culture medium, and the cytokine composition is hrIL-12 and hrIL- 18, or a combination of hrIL-12, hrIL-15 and hrIL-18.
  26. 嵌合受体修饰的自然杀伤细胞,其采用权利要求1-25任一项所述的方法制备得到。Chimeric receptor-modified natural killer cells, which are prepared by the method described in any one of claims 1-25.
  27. 权利要求26所述的嵌合受体修饰的自然杀伤细胞在制备肿瘤治疗药物和/或肿瘤预防药物中的应 用。The application of the chimeric receptor-modified natural killer cell according to claim 26 in the preparation of a tumor therapeutic drug and/or a tumor preventive drug.
PCT/CN2021/103235 2020-07-09 2021-06-29 Non-viral method for preparing stable nk cells with high chimeric receptor expression WO2022007665A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010657827.5 2020-07-09
CN202010657827.5A CN113913458A (en) 2020-07-09 2020-07-09 Non-viral method for preparing NK (natural killer) cells of stable high-expression chimeric receptor

Publications (1)

Publication Number Publication Date
WO2022007665A1 true WO2022007665A1 (en) 2022-01-13

Family

ID=79231926

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/103235 WO2022007665A1 (en) 2020-07-09 2021-06-29 Non-viral method for preparing stable nk cells with high chimeric receptor expression

Country Status (2)

Country Link
CN (1) CN113913458A (en)
WO (1) WO2022007665A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0814838A1 (en) * 1995-03-08 1998-01-07 The Scripps Research Institute Antigen presenting system and methods for activation of t-cells
CN106414748A (en) * 2014-02-14 2017-02-15 得克萨斯州大学系统董事会 Chimeric antigen receptors and methods of making
CN107523545A (en) * 2016-06-20 2017-12-29 上海细胞治疗研究院 A kind of lethal cell of high efficiency stable expression antibody and application thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0814838A1 (en) * 1995-03-08 1998-01-07 The Scripps Research Institute Antigen presenting system and methods for activation of t-cells
CN106414748A (en) * 2014-02-14 2017-02-15 得克萨斯州大学系统董事会 Chimeric antigen receptors and methods of making
CN107523545A (en) * 2016-06-20 2017-12-29 上海细胞治疗研究院 A kind of lethal cell of high efficiency stable expression antibody and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
RAN YUE, ZIYANG LIU, YA ZHENG, XIAODAN LU, SHANSHAN HU, BINGYONG ZHANG, XIULING LI, JINGGUO LI, SHUANGYIN HAN: "Nanocarrier-mediated PiggyBac transposon system for preparation of CAR-NK cells", CHINESE JOURNAL OF CANCER BIOTHERAPY, vol. 27, no. 2, 25 February 2020 (2020-02-25), Publishing House of Chinese Journal of Cancer Biotherapy, CN, pages 109 - 114, XP055886315, ISSN: 1007-385X, DOI: 10.3872/j.issn.1007-385x.2020.02.002 *
YOU FENGTAO: "CD7-CAR-NK-92MI Cell Targeted Therapy for T Cell Tumor and A Bispecific CAR-T Cell Targeted Therapy for B Cell Tumor", CHINESE DOCTORAL DISSERTATIONS FULL-TEXT DATABASE-SOOCHOW UNIVERSITY, 1 June 2019 (2019-06-01), XP055886320, [retrieved on 20220202] *

Also Published As

Publication number Publication date
CN113913458A (en) 2022-01-11

Similar Documents

Publication Publication Date Title
AU2016206868B2 (en) Modified hepatitis post-transcriptional regulatory elements
US20210154231A1 (en) Method for producing t cells modified by chimeric antigen receptor
CN111566221B (en) Methods for NK cell transduction
CN112204133A (en) CAR NK cells
CN112210539A (en) Fourth-generation CAR-T cell and application thereof
US11912987B2 (en) Methods for screening for cancer targets
CN112063653A (en) Method for preparing NK (natural killer) sample cells based on electrotransfer reprogramming plasmid
WO2022007665A1 (en) Non-viral method for preparing stable nk cells with high chimeric receptor expression
EP4349969A1 (en) Composition containing feeder cell for proliferating natural killer cell
CN112592898B (en) Reprogrammed NK feeder cells and preparation method and application thereof
CN111378046A (en) Immune effector cell conversion receptor
CN112626028B (en) Engineering cell for activating NK-like cells and preparation method and application thereof
US20230142916A1 (en) All-in one vector for car and therapeutic effector molecule
EP2267118A1 (en) Method for production of transfected cell
CN114269929A (en) Methods of producing cell populations containing CAR-expressing immune cells
CN112048480A (en) Method for preparing NK (natural killer) sample cells based on electrotransformation
CN116410336B (en) Chimeric antigen receptor encoding nucleotide, CAR-NK cell, construction method and application thereof
CN117467619B (en) Self-disintegrating induced artificial antigen presenting cell and preparation method and application thereof
WO2023125860A1 (en) Preparation technique for universal car-t cell, and application of universal car-t cell
US20220228164A1 (en) Engineered antigen presenting cells
US11359012B1 (en) Specific chimeric antigen receptor cells targeting human CLDN18A2, preparation method and application thereof
WO2023082640A1 (en) Method for enhancing durability of immune cell
CN113913384A (en) Method for preparing target specific NK cells and application thereof
CN113913459A (en) Method for preparing high-purity chimeric antigen receptor modified T cell by non-viral method and application thereof
CN112608901A (en) Artificial antigen presenting cell and preparation method and application thereof

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: 21837744

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21837744

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 21837744

Country of ref document: EP

Kind code of ref document: A1