WO2019085102A1 - Bcma特异性嵌合抗原受体t细胞及其应用 - Google Patents

Bcma特异性嵌合抗原受体t细胞及其应用 Download PDF

Info

Publication number
WO2019085102A1
WO2019085102A1 PCT/CN2017/113221 CN2017113221W WO2019085102A1 WO 2019085102 A1 WO2019085102 A1 WO 2019085102A1 CN 2017113221 W CN2017113221 W CN 2017113221W WO 2019085102 A1 WO2019085102 A1 WO 2019085102A1
Authority
WO
WIPO (PCT)
Prior art keywords
bcmascfv
chimeric antigen
antigen receptor
cd8a
cd3ζ
Prior art date
Application number
PCT/CN2017/113221
Other languages
English (en)
French (fr)
Inventor
龙飞
陈毓
胡金芳
李燕
张丽琴
Original Assignee
西安宇繁生物科技有限责任公司
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 西安宇繁生物科技有限责任公司 filed Critical 西安宇繁生物科技有限责任公司
Publication of WO2019085102A1 publication Critical patent/WO2019085102A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors

Definitions

  • the invention relates to the field of cellular immunotherapy of tumors, in particular to obtaining a chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ and the chimeric antigen receptor-modified T lymphocytes, which are modified by genetic engineering technology.
  • T lymphocytes can specifically recognize and kill tumor cells that express BCMA on the surface, and the tumor cells are preferably multiple myeloma cells.
  • Single-chain antibodies are relatively small in molecular mass, easy to construct and express, and have low immunogenicity and are not susceptible to hypersensitivity and rejection. Single-chain antibodies are among the most actively studied genetically engineered antibodies. Fusion proteins prepared by DNA recombinant engineering from single-chain antibodies and toxins or immune effector molecules, as well as intracellularly expressed single-chain antibodies, have shown good promise in disease research and treatment.
  • MM Multiple myeloma
  • MM is a common hematological malignancy in which malignant proliferation of mature plasma cells in the bone marrow leads to bone destruction and bone marrow failure.
  • MM accounts for about 1% of all human malignancies, accounting for 10% of hematological malignancies, MM in the United States (1/100000) is 3.5 to 4.5, China is about 1, and the median age of onset is in the United States. It is 68 years old and relatively low in China.
  • MM patients usually show symptoms such as bone pain, anemia, osteolytic lesions, hypercalcemia, renal dysfunction, cytopenia, and symptomatic plasma cells.
  • Traditional chemotherapy, hematopoietic stem cells, and proteasome inhibitors used in recent years have a certain effect, and the overall survival rate of patients has been improved, but it is still not completely cured.
  • BCMA B cell maturity antigen
  • TNF receptor family belongs to the TNF receptor family and is expressed in the differentiation of mature B cells into plasma cells. Due to its mRNA-restricting characteristics, BCMA is not expressed in the main organs of the human body. This is more specific and has fewer side effects than most of the monoclonal antibodies currently used in all B-lineage cells.
  • Chimeric antigen receptor-modified T cell (CAR-T) therapy utilizes genetic engineering technology to enable T cells to recognize tumor surface antigens and kill tumors by means of retrovirus or reverse viral vector or mRNA transduction. Chimeric antigen receptor. Compared with CAR-T treatment The traditional treatment method shows great advantages, which is reflected in the high precision of killing tumors.
  • the CAR-T cell therapy adopts the technology of antigen-antibody specific binding. Only the tumor cells with surface antigen expression are killed, and the damage to normal cells is small. The scope of killing tumors is wide. As long as the tumor-associated antigen is expressed, CAR-T cells can be cleared, which is effective for metastatic tumors and recurrent tumors.
  • CAR-T is not that perfect. Given the current limited number of cases, it is not possible to draw a systematic conclusion of efficacy. And the results of some cases have shown that some patients have more serious clinical side effects after CAR-T cell reinfusion treatment. What's more, some CAR-T recognition errors cause them to attack normal cells, the so-called "off-target effect" .
  • studies have confirmed that CAR-T cells with strong affinity and killing ability to target antigens also attack normal tissues and cause damage to tissues and organs, and in 2010, reported a case of colorectal cancer with liver. Patients with lung metastases die from off-target effects after CAR-T treatment with human epidermal growth factor receptor 2.
  • the present inventors have disclosed a specific chimeric antigen receptor directed against human BCMA which is capable of specifically binding to a target antigen and exerting a cytotoxic effect.
  • the present invention provides a chimeric antigen receptor (anti-BCMAscFv-CD8a-41BB-CD3 ⁇ ) targeting BCMA, which can specifically recognize and kill T cells.
  • a chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇
  • Tumor cells that express BCMA molecules on the surface, and preparation of therapeutic drugs for tumors.
  • a first aspect of the invention relates to a chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ comprising a single-chain antibody anti-BCMAscFv, a hinge region, a transmembrane region, and an intracellular region .
  • the single-chain antibody anti-BCMAscFv is formed by connecting a heavy chain variable region and a light chain variable region of an anti-human BCMA monoclonal antibody derived from BCMA-immunized mice. Hybridoma cells.
  • Both the hinge region and the transmembrane region are derived from the human CD8a region gene.
  • the intracellular region is composed of a co-stimulatory factor 4-1BB and a CD3 ⁇ series, that is, a human 4-1BB intracellular region and a human CD3 intracellular region.
  • the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ of the present invention further comprises a human CD8 ⁇ signal peptide region, and preferably, the gene sequence corresponding to the amino terminus of the CD8 ⁇ signal peptide region is introduced into the Kozak sequence and/or Enzyme cleavage site sequence.
  • the amino acid sequence of the complementarity determining region CDR-H1 to CDR-H3 of the heavy chain variable region of the anti-human BCMA monoclonal antibody is as shown in SEQ ID NO: 1-5, the anti-human BCMA monoclonal
  • the amino acid sequence of the complementarity determining regions of the light chain variable region of the antibody is shown in SEQ ID NOs: 6-8.
  • the amino acid sequence of the single-chain antibody anti-BCMAscFv of the present invention is shown as SEQ ID NO: 9 or SEQ ID NO: 10, or at least about 90%, 91% with SEQ ID NO: 9 or SEQ ID NO: 10.
  • the nucleotide sequence of the single-chain antibody anti-BCMAscFv is shown in SEQ ID NO: 11 or SEQ ID NO: 12.
  • the light chain and the heavy chain of the anti-BCMAscFv are linked by a linker peptide having an amino acid sequence of Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Gly Ser.
  • the hinge region and the transmembrane region are linked in series, the amino acid sequence is set forth in SEQ ID NO: 15, and the nucleotide sequence of the hinge region and the transmembrane region is SEQ ID NO: NO:16.
  • amino acid sequence of the intracellular region 4-1BB-CD3 ⁇ is shown in SEQ ID NO: 17
  • nucleotide sequence of the intracellular region 4-1BB-CD3 ⁇ is shown in SEQ ID NO: 18.
  • the amino acid sequence of the CD8 ⁇ signal peptide region is shown in SEQ ID NO: 13, and the nucleotide sequence of the CD8 ⁇ signal peptide region is shown in SEQ ID NO: 14.
  • the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ of the present invention is selected from the group consisting of One of them:
  • amino acid sequence of the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ is shown as part or all of the sequence shown in SEQ ID NO: 19 or 20;
  • the amino acid sequence of the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ has a degree of sequence identity with the amino acid of SEQ ID NO: 19 or 20 of at least about 90%, 91%, 92%, 93%. , 94%, 95%, 96%, 97%, 98% or at least 99%;
  • nucleic acid sequence encoding a chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ amino acid hybridizes under stringent conditions to the nucleotide sequence of the amino acid sequence set forth in SEQ ID NO: 19 or 20;
  • the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ amino acid has no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 Or no more than 1 amino acid;
  • a chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ amino acid sequence having the amino acid sequence of SEQ ID NO: 19 or 20, comprising substitutions, deletions and/or insertions of one or more amino acid residues;
  • nucleotide sequence encoding the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ is part or all of the sequence shown in SEQ ID NO: 21 or 22;
  • the nucleotide sequence encoding the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ has a degree of identity with the nucleotide sequence set forth in SEQ ID NO: 21 or 22 of at least about 90%, 91% , 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least 99%;
  • nucleotide sequence encoding the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ hybridizes under stringent conditions to the nucleotide sequence set forth in SEQ ID NO: 21 or 22;
  • nucleotide sequence encoding the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ differs from the sequence shown in SEQ ID NO: 21 or 22 by no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or no more than 1 nucleotide;
  • nucleotide sequence encoding the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ has the sequence shown in SEQ ID NO: 21 or 22, including substitutions, deletions and/or insertions of one or more nucleotides Nucleotide sequence.
  • amino acid sequence of the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ of the present invention is shown as SEQ ID NO: 19 or SEQ ID NO: 20, or with SEQ ID NO: 19.
  • SEQ ID NO: 20 is at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, Sequence of 99.4%, 99.5%, 99.7%, 99.8% or 99.9% amino acid sequence identity.
  • nucleotide sequence encoding the above chimeric antigen receptor chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ of the present invention is shown as SEQ ID NO: 21 or SEQ ID NO: 22.
  • a second aspect of the invention relates to a DNA encoding any of the chimeric antigen receptors anti-BCMAscFv-CD8a-41BB-CD3 ⁇ .
  • a third aspect of the invention relates to the use of any of the above chimeric antigen receptors anti-BCMAscFv-CD8a-41BB-CD3 ⁇ or the above DNA for the preparation of chimeric antigen receptor T cells.
  • a fourth aspect of the invention relates to a method for preparing an anti-BCMA CAR-T cell, wherein the method comprises the following steps:
  • step 4) Infecting T cells with anti-BCMA-CAR-containing lentiviral particles obtained in step 3) to obtain BCMA-specific CAR-T cells.
  • a fifth aspect of the invention relates to a cell, tissue, organ or construct comprising the chimeric antigen receptor or DNA described above, said cell, tissue, organ or construct expressing a specific chimeric antigen of BCMA body.
  • the cell is a T cell. More preferably, the T cells are prepared by the above method.
  • a sixth aspect of the present invention relates to the use of the above chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ or the above DNA or the above-mentioned cells, tissues, organs or constructs for the preparation of a therapeutic composition for treating tumors.
  • a seventh aspect of the invention relates to a pharmaceutical composition for treating a tumor, wherein the pharmaceutical composition comprises the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ of any of the above, the above DNA Or a cell, tissue, organ or construct as described above.
  • An eighth aspect of the invention relates to the use of any of the above-described chimeric antigen receptors anti-BCMAscFv-CD8a-41BB-CD3 ⁇ , DNA, or the above-described cells, tissues, organs or constructs for treating tumors.
  • a method of treating a tumor comprising administering to a patient a pharmaceutical composition as described above.
  • the tumor is any malignant tumor, including cancer of various organs, sarcoma, malignant tumor of the blood system, and the like.
  • malignant tumor including cancer of various organs, sarcoma, malignant tumor of the blood system, and the like.
  • the tumor is multiple myeloma.
  • the present invention employs a gene sequence of an anti-human BCMA single-chain antibody and optimizes its codon.
  • the signal sequence of human CD8 ⁇ , the hinge region, the transmembrane region and the intracellular region of 4-1BB and CD3 ⁇ were searched from the NCBI database. After segmentation synthesis, overlap extension PCR (SOE-PCR) was used for splicing and then cloned.
  • SOE-PCR overlap extension PCR
  • the expression vector and the packaging plasmids pMD2.G and psPAX2 were used to co-transfect 293T cells to obtain lentiviral particles, and then infected with human T lymphocytes to express T lymphocytes.
  • the chimeric antigen receptor was used to co-transfect 293T cells to obtain lentiviral particles, and then infected with human T lymphocytes to express T lymphocytes.
  • the obtained CAR-T cells were co-cultured with RPMI8226 cells in vitro, the cell infection efficiency was detected by flow cytometry, and the levels of cytokines (IL2, TNF ⁇ , ⁇ -IFN) in the co-culture supernatant were detected by ELISA.
  • the cytotoxicity of CAR-T cells to RPMI 8226 cells was examined by non-radioactive cytotoxicity assay to confirm the specific killing effect of the chimeric antigen receptor-modified T lymphocytes on tumor cells. Therefore, the chimeric antigen receptor anti-BCMAscFv-CD8a-41BB-CD3 ⁇ described in the present invention can be used in cell immunotherapy of tumors.
  • Figure 1 a schematic view of the structure of chimeric antigen receptor anti-BCMA-CD8a-41BB- CD3 ⁇ , wherein, SP is the CD8 ⁇ signal peptide region, V L is the light chain variable region, V H is a heavy chain variable region, CD8hinge of In the hinge region, CD8TM is a transmembrane region, 4-1BB is a human 4-1BB intracellular region, and CD3 ⁇ is a human CD3 intracellular region;
  • Figure 2 Anti-BCMA-CD8a-41BB-CD3 ⁇ lentiviral vector map
  • Figure 5 Flow cytometry to detect the proportion of CAR-T positive cells in total cells after 72 hours of Lentivirus-infected T cells.
  • Figure 5A shows uninfected T cells
  • Figure 5B shows T cells after 72 hours of infection.
  • Medium is a CAR-T positive cell population;
  • FIG. 6 ELISA detects IL-2 cytokine levels
  • FIG. 7 ELISA detects IFN ⁇ cytokine levels
  • FIG. 8 ELISA detects TNFa cytokine levels
  • Figure 9 Percentage of cleavage of BCMA positive cell line RPMI8226 by VEC-T and CAR-T cells at different potency: target ratios (1:4, 2:4, 4:4);
  • Figure 10 Percentage of cleavage of BCMA negative cell line K562 by VEC-T and CAR-T cells at different potency: target ratios (1:4, 2:4, 4:4);
  • Figure 11 Detection of the effect of anti-BCMA CAR-T cells on KMS-11 cells using a bioluminescence imaging system in which T cells transfected with empty vector were compared to PBS.
  • Example 1 Determination of anti-BCMA-CD8a-41BB-CD3 ⁇ gene sequence
  • the gene sequence of the anti-human BCMA single-chain antibody was derived from hybridoma cells of BCMA-immunized mice.
  • the human CD8 ⁇ signal peptide region, the hinge region and the transmembrane region, the human 4-1BB intracellular region, and the human CD3 intracellular region gene sequence information were searched from the NCBI website database.
  • the above sequences were codon optimized on the website http://www.jcat.de/ to ensure that the gene expression was more suitable for human gene expression without changing the amino acid sequence.
  • the Kozak sequence and the restriction site were introduced at the amino terminus of the signal peptide gene sequence, and each nucleotide sequence was sent to Shanghai Shenggong Biotechnology Co., Ltd. for synthesis.
  • anti-BCMA-CAR CAR molecule
  • SEQ ID NO: 21 or SEQ ID NO: 22 The nucleotide sequence of anti-BCMA-CAR is shown in SEQ ID NO: 21 or SEQ ID NO: 22, and the encoded amino acid sequence is shown in SEQ ID NO: 19 or SEQ ID NO: 20.
  • the nucleotide sequence of anti-BCMA-CAR was digested with EcoR I and BamH I, and inserted into the engineered lentiviral vector pLVX-EF1a-GFP-N1 (Addgene) by EcoT I and BamH I by T4 DNA ligase.
  • the site was transformed into competent DH5 ⁇ E. coli.
  • the obtained recombinant plasmid was sent to Shanghai Shenggong Biotechnology Co., Ltd. for sequencing, and the sequencing result was compared with the fitted anti-BCMA-CAR sequence to verify whether the sequence was correct.
  • Anti-BCMA-CD8a-41BB-CD3 ⁇ lentiviral vector map as shown in Figure 2 Shown.
  • Example 3 Packaging and titer determination with anti-BCMA-CAR lentivirus
  • the lentiviral packaging vectors pMD2.G, psPAX2 and anti-BCMA-CAR lentiviral vectors were co-transfected into 293T cells using the endotoxin-free plasmid (Tiangen Bio) operating instructions, 48 h after transfection.
  • the cell supernatant was collected at 72 h, centrifuged at 400 rcf for 10 min, and the cells and cell debris in the supernatant were removed.
  • the supernatant was filtered through a 0.45 ⁇ m filter and stored in duplicate for use.
  • the supernatant of the virus was diluted and then infected with 293T cells.
  • the positive rate of GFP was detected by fluorescence microscopy.
  • the titer of pLVX-CAR and pLVX-loaded virus stocks was estimated to be about 2 according to the formula. ⁇ 10 6 TU/mL and 5 ⁇ 10 6 TU/mL. After concentration by concentration, the titer can reach 1 ⁇ 10 8 TU/mL and 2 ⁇ 10 8 TU/mL.
  • Example 4 Lentivirus infection of human T cells
  • the first step Separate the pure CD3+ T cells with Ficoll separation solution (Tianjin Yuyang) and human T cell enriched antibody mixture, and adjust the cell density to 1 with 5% AB serum X-VIVO (LONZA) medium. ⁇ 10 6 /mL.
  • the cells were seeded at 1 mL/well into a 24-well plate pre-coated with anti-human 50 ng/m LCD3 antibody (Beijing Yiqiao Shenzhou Biotechnology Co., Ltd.) and 50 ng/mL CD28 antibody (Beijing Yiqiao Shenzhou Biotechnology Co., Ltd.) overnight.
  • 100 IU/mL of interleukin 2 (PeproTech) was added, and the virus obtained by the preparation of Example 3 was infected after 48 hours of stimulation;
  • the second step T cell activation was cultured every other day, and a new 24-well plate was coated with Retronectin (Takara) diluted to a final concentration of 15 ⁇ g/mL in PBS, 250 ⁇ L per well. Protect from light and store overnight at 4 °C.
  • Step 3 After two days of T cell activation, remove the 24-well plate of Retronectin coated in the previous step, aspirate the coating solution, add 500 ⁇ L of HBSS containing 2% BSA to each well, block for 30 min at room temperature, and aspirate the blocking solution. The plate was washed twice with HBSS containing 2.5% HEPES.
  • the fourth step the virus solution prepared in Example 3 was added to the above T cells, 500 ⁇ L per well, and then added to a final concentration of 8 ⁇ g/mL of polybrene, mixed at 32 ° C, 1500 g, and centrifuged for 2 h. Place in an incubator for 24 h.
  • Step 5 Centrifuge the cells after infection for 24 hours at 1000g for 10min, carefully aspirate the supernatant, and resuspend the cells in fresh RPMI1640 medium containing 10% FBS to a concentration of 1 ⁇ 10 6 /mL to 1 ⁇ 10 6 /well. Inoculate the well-packed 24-well plate in the third step, then add 100 IU/mL of interleukin 2, the same amount of virus stock solution and polybrene, 32 ° C, 1500 g, and centrifuge for 2 h. It is then placed in an incubator for cultivation.
  • Step 6 After cell infection, observe the density of the cells every day, and add T cell culture medium containing IL-2 100 IU/mL in time to maintain the density of T cells at about 5 ⁇ 10 5 /mL to expand the cells. After 72 hours of infection, the green fluorescence expression of T cells was observed by fluorescence microscopy and photographed. The results are shown in Figure 3. Thus, CAR-T cells infected with the lentivirus described in Example 3 were obtained, and named anti-BCMA CAR-T cells, that is, BCMA-specific CAR-T cells, and the anti-BCMA-CAR of Example 2 was expressed.
  • Example 4 The infected human T cells in Example 4 were collected and washed with PBS for 72 hours, and the cells were lysed by the cell protein extraction reagent (RIPA), and the proteins of the infected T cells were separated by 10% SDS-PAGE, and the constant flow was performed. 300 mA, 1 h) was transferred to a PVDF membrane, incubated with rabbit anti-CD3 ⁇ (1:400) antibody, and incubated overnight at 4 °C. After washing 3 times with TBST, the HRP goat anti-rabbit secondary antibody (1:10000) was incubated for 1 h at room temperature. After adding DAB color development, imaging was performed using Azure's C-300 multi-function imaging system, and the results are shown in FIG. It can be seen from Fig.
  • the recombinant plasmid constructed by the present invention can detect the expression of the target band, and the size is consistent with the expected CAR ( ⁇ 55 kDa), as shown in Figure 4, the band is 2-5; while the uninfected T cells have no band. See Figure 4 for strip 6.
  • Example 6 Flow cytometry to detect the proportion and surface expression of T lymphocytes after infection
  • Example 4 After 72 hours of infected T cells in Example 4, about 10 6 cells were collected, washed once with buffer (PBS containing 5% FBS), the supernatant was discarded by centrifugation, and resuspended by adding 100 ⁇ L of buffer. Add 1 ⁇ g of primary antibody BCMA-biotin and incubate for 40 min in a 37 ° C incubator. Centrifuge, discard the supernatant, and wash 1-2 times with 300 ⁇ L of buffer. Resuspend in 100 ⁇ L of buffer, add secondary anti-streptavidin-PE (concentration according to the instructions), incubate in a refrigerator at 20 ° C for 20-30 min. After centrifugation, the supernatant was discarded and washed 1-2 times with 300 ⁇ L of buffer. Resuspend with 300 ⁇ L of buffer and test with BD Calibur.
  • buffer PBS containing 5% FBS
  • the anti-BCMA + CAR-T cells accounted for about 50% of the total number of cells.
  • Example 7 Determination of tumor killing ability of CAR-T cells by in vitro co-culture
  • Example 4 The anti-BCMA CAR-T cells of Example 4 were collected by centrifugation one week after the infection and resuspended in fresh RPMI1640 medium containing 10% FBS at a concentration of 2 ⁇ 10 6 /mL.
  • T cells and target cells RPMI 8226 were co-cultured in a 6-well cell culture plate as follows:
  • VEC-T 2 ⁇ 10 6 / hole empty vector infected T cells
  • VECT + R 2 ⁇ 10 6 / hole VEC-T, 1 ⁇ 10 6 / hole RPMI 8226 (referred to as VECT + R in the legend)
  • Standard dilution Standards were diluted to 2400 ng/mL, 1200 ng/mL, 600 ng/mL, 300 ng/mL, 150 ng/mL with standard dilutions.
  • Color development Add 50 ⁇ L of the developer A to each well, then add 50 ⁇ L of the developer B, gently shake and mix, and develop at 37 ° C for 15 min in the dark.
  • Termination 50 ⁇ L of the stop solution was added to each well to terminate the reaction.
  • the IL2 detection value in CAR-T cells was 1876.0 pg/mL, while the other 3 groups were less than 50 pg/mL (see Figure 6); the ⁇ -IFN detection value was 4070.3. Pg/mL, while the remaining 3 groups were all less than 50 pg/mL (see Figure 7); the TNFa assay was 1236.8 pg/mL, while the remaining 3 groups were all less than 65 pg/mL (see Figure 8).
  • the secretion of each factor in CAR-T cells was significantly increased.
  • Target cell preparation Logarithmic growth phase RPMI 8226 cells were collected by centrifugation at 800 rpm for 5 min, and the cell concentration was adjusted to 1 ⁇ 10 6 /mL with RPMI1640 containing 5% FBS.
  • Lactate dehydrogenase (LDH) positive control preparation gently shake the LDH positive control to mix, then 2 ⁇ L diluted to 10 mL of PBS + 1% BSA (1:5000 dilution).
  • LDH positive control group 110 ⁇ L / well LDH diluted in step 2;
  • Maximum LDH release from target cells set the number of target cells to 0, 5 ⁇ 10 3 , 1 ⁇ 10 4 , 2 ⁇ 10 4 , 3 ⁇ 10 4 , 4 ⁇ 10 4 , and fill each well with RPMI1640 containing 5% FBS The volume was 100 ⁇ L and 10 ⁇ L of cell lysate (10 ⁇ ) was added to each well.
  • the microplate reader detects the absorbance at 490 nm.
  • the target cell number when the target cell absorbance value is at least twice the medium background control absorbance value is 5 ⁇ 10 4 .
  • Target cell preparation Logarithmic growth phase RPMI 8226 or K562 cells were collected by centrifugation at 800 rpm for 5 min, and the cell concentration was adjusted to 5 ⁇ 10 6 /mL with RPMI1640 containing 5% FBS.
  • CAR-T and VEC-T cells were collected one week after infection by centrifugation at 1000 rpm for 10 minutes, and the cell concentration was adjusted to 2.5 ⁇ 10 6 /mL with RPMI1640 containing 5% FBS.
  • volume corrected control well 100 ⁇ L 5% FBS of RPMI1640 + 10 ⁇ L cell lysate (10 ⁇ );
  • Target cell spontaneous release group 10 ⁇ L RPMI 8226 or K562 cells (5 ⁇ 10 4 cells) + 100 ⁇ L 5% FBS RPMI1640;
  • effector cells 5 ⁇ L, 10 ⁇ L, 20 ⁇ L/well of effector cells according to different effects: target ratio (1:4, 2:4, 4:4), all supplemented with 110 ⁇ L with RPMI1640 containing 5% FBS /hole;
  • CTL activity calculation Calculate the average value (A 490 ) of each group (3 complex wells), and calculate the percentage of CAR-T cell lysis target cells according to the following formula, and the formula is as follows.
  • the cleavage rates of anti-BCMA CAR-T cells to BCMA-positive tumor cells RPMI 8226 were 34%, 55%, and 62%, respectively.
  • the number of CAR-T cells increased and the lysis effect was significantly enhanced.
  • the cleavage rate of anti-BCMA CAR-T cells to BCMA-negative cell line K562 was less than 20%; as shown in Figure 9 and Figure 10, anti-BCMA CAR-T cells were positive for BCMA-positive tumor cells.
  • 8226 has a strong cleavage effect, but does not produce cleavage of the BCMA negative cell line K562.
  • Example 8 Evaluation of in vivo efficacy of anti-BCMACAR-T cells
  • KMS-11 is a human multiple myeloma cell line that expresses BCMA protein on the cell surface and can be grown as a xenograft in immunocompromised mice. KMS-11 cells were labeled with firefly luciferase and implanted in NSG mice to establish a multiple myeloma model that tested the efficacy of anti-BCMACAR-T.
  • luciferase-labeled KMS-11 cells were injected into NSG mice through the tail vein, and after 9 days of culture, 8 ⁇ 10 6 anti-BCMA CAR-T cells or transfected empty vector were injected through the tail vein. T cells or PBS.
  • the health status of the mice was measured daily, and tumor burden was monitored by a bioluminescence imaging system at 1, 7 days, 14 days, and 21 days, respectively.
  • D-fluorescein was injected intraperitoneally before the mice were anesthetized, and mice were anesthetized with Xenogen imaging 5 minutes after the injection, and the disease burden was evaluated by bioluminescence of the tumor cells. As a result, as shown in Fig.
  • the T cell group transfected with the empty vector and the mouse in the PBS group showed a strong bioluminescence signal, in contrast to the bioluminescence produced by the anti-BCMACAR-T cell group on the 14th day.
  • the signal was significantly reduced, and no biofluorescence signal was detected on day 21, indicating that anti-BCMACAR-T has a good clearance rate for BCMA-positive tumor cells in vivo.

Landscapes

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

Abstract

本发明公开了一种针对人BCMA的特异性嵌合抗原受体(anti-BCMA scFv-CD8a-41BB-CD3ζ)及其应用,该嵌合抗原受体由单链抗体anti-BCMA scFv,铰链区、跨膜区,胞内区串联构成,该嵌合抗原受体用于修饰人T淋巴细胞,修饰后的T淋巴细胞用于表面BCMA阳性肿瘤的预防与治疗以及肿瘤药物的制备。

Description

BCMA特异性嵌合抗原受体T细胞及其应用 技术领域
本发明涉及肿瘤的细胞免疫治疗领域,具体涉及利用基因工程技术获得了一种嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ和该嵌合抗原受体修饰的T淋巴细胞,被修饰的T淋巴细胞可以特异性的识别和杀伤表面表达BCMA的肿瘤细胞,所述肿瘤细胞优选为多发性骨髓瘤细胞。
背景技术
完整抗体的相对分子质量较大。仅由抗体重链V区(VH)与轻链V区(VL)通过人工合成的连接肽(linker)连接而成的小分子抗体称为单链抗体(single chain antibody fragment,scFv)。单链抗体相对分子质量小,易于构建和表达,而且其免疫原性弱,不易引起超敏反应和排斥反应。单链抗体是目前研究最活跃的基因工程抗体之一。由单链抗体与毒素或免疫效应分子通过DNA重组工程制备的融合蛋白以及胞内表达的单链抗体等在疾病研究与治疗中展现了良好的前景。
多发性骨髓瘤(multiple myeloma,MM)是一种由成熟浆细胞在骨髓中恶性增殖累积导致骨破坏和骨髓衰竭的常见的血液系统恶性肿瘤。据统计,MM约占所有人类恶性肿瘤的1%,占血液系统恶性肿瘤的10%,MM在美国的发病率(1/100000)为3.5~4.5,中国约为1,中位发病年龄在美国为68岁,在我国则相对较低。MM患者通常表现出骨疼痛、贫血、溶骨性病变、高钙血症、肾功能损害、血细胞减少症、症状性浆细胞产生等症状。传统的化疗、造血干细胞以及近年来使用的蛋白酶体抑制剂虽然都有一定的疗效,患者的总体生存率得到了提高,但仍无法完全治愈。
BCMA(B cell maturition antigen,B细胞成熟抗原)属于TNF受体家族成员,在成熟B细胞分化到浆细胞阶段表达,由于其具有mRNA限制性的特点,BCMA并不会表达于人体主要的器官中,这与目前所应用的大多数单抗类作用于所有B系细胞的特点相比,特异性更强,副作用更小。
嵌合抗原受体修饰的T细胞(chimeric antigen receptor,CAR-T)治疗是利用基因工程技术,借助逆转录病毒或者逆转病毒载体或者mRNA转导,使得T细胞获得可以识别肿瘤表面抗原,杀伤肿瘤的嵌合抗原受体。CAR-T治疗相比 于传统的治疗方法展现出巨大的优势,具体体现在杀伤肿瘤精准性高,CAR-T细胞治疗采用抗原抗体特异性结合的技术,只有表面表达抗原的肿瘤细胞被杀伤,对正常细胞的伤害小;杀伤肿瘤范围广泛,只要表达肿瘤相关抗原,CAR-T细胞就能清除,对于转移性肿瘤、复发性肿瘤均有效;对于患者而言避免了放化疗的痛苦,恢复健康迅速。首次临床应用的报道是2008年,在Nature medicine上报道了使用CAR-T细胞治疗11例儿童成纤维神经瘤,6例好转的病例。目前的临床应用,主要集中在白血病上,针对的是CD19、CD20、CD22三个靶向位点。在2011和2013年,先后在NEJM上发表了个案报道,描述了运用CAR-T细胞治疗慢性和急性白血病的病例。2014年7月FDA授予诺华公司开发的个性化CAR-T癌症疗法CTL019突破性药物认证,2017年8月,美国FDA核准该药(商品名Kyrmiah)上市。
但是,CAR-T也并不是那么完美。鉴于目前病例数量有限,所以无法得出一个系统的疗效结论。并且已有病例结果显示,部分患者CAR-T细胞回输治疗后有较为严重的临床副反应,更有甚者,有些CAR-T的识别失误导致其攻击正常细胞,即所谓的“脱靶效应”。截止目前,已经有研究证实对靶抗原亲和力和杀伤能力强的CAR-T细胞在清除肿瘤的同时也会攻击正常组织,造成组织器官的损伤,在2010年,报道了1例结直肠癌合并肝肺转移的患者在接受人表皮生长因子受体2的CAR-T治疗后因脱靶效应而死亡。
由此可见,本领域需要特异性的胞外靶向连接区,其能够特异性的对目标抗原进行识别,避免错误识别的影响。本发明人公开了一种针对人BCMA的特异性嵌合抗原受体及其应用,所述CAR能够特异性的结合到靶抗原上,并发挥细胞毒效应。
发明内容
为解决上述问题,本发明提供了一种靶向BCMA的嵌合抗原受体(anti-BCMAscFv-CD8a-41BB-CD3ζ),该嵌合抗原受体修饰的T淋巴细胞可以特异性的识别并杀伤表面表达BCMA分子的肿瘤细胞,以及制备治疗肿瘤药物。
本发明的第一方面,涉及一种嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,所述嵌合抗原受体包括单链抗体anti-BCMAscFv,铰链区、跨膜区,胞内区。
优选的,所述单链抗体anti-BCMAscFv由抗人BCMA单克隆抗体的重链可变区和轻链可变区串联而成,所述的抗人BCMA单克隆抗体来源于BCMA免疫小鼠的杂交瘤细胞。
所述铰链区和所述跨膜区均来源于人的CD8a区基因。
所述胞内区由共刺激因子4-1BB和CD3ζ串联构成,即为人4-1BB胞内区及人CD3ζ胞内区。
本发明所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ还包括人的CD8α信号肽区,优选的,在所述CD8α信号肽区氨基端对应的基因序列引入Kozak序列和/或酶切位点序列。
优选的,所述抗人BCMA单克隆抗体的重链可变区的互补性决定区CDR-H1~CDR-H3的氨基酸序列如SEQ ID NO:1-5所示,所述抗人BCMA单克隆抗体的轻链可变区的互补性决定区CDR-L1~CDR-L3的氨基酸序列如SEQ ID NO:6-8所示。本发明所述的单链抗体anti-BCMAscFv的氨基酸序列如SEQ ID NO:9或SEQ ID NO:10所示,或至少与SEQ ID NO:9或SEQ ID NO:10有约90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、99.1%、99.2%、99.3%、99.4%、99.4%、99.5%、99.7%、99.8%或99.9%的氨基酸序列一致性的序列。
所述的单链抗体anti-BCMAscFv的核苷酸序列如SEQ ID NO:11或SEQ ID NO:12所示。
所述的anti-BCMAscFv的轻链和重链之间由氨基酸序列为Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser的连接肽连接而成。
在本发明的一个实施例中,所述铰链区和跨膜区串联连接,所述氨基酸序列如SEQ ID NO:15所示,所述铰链区和跨膜区串联的核苷酸序列如SEQ ID NO:16所示。
所述胞内区4-1BB-CD3ζ的氨基酸序列如SEQ ID NO:17所示,所述胞内区4-1BB-CD3ζ的核苷酸序列如SEQ ID NO:18所示。
所述CD8α信号肽区的氨基酸序列如SEQ ID NO:13所示,所述的CD8α信号肽区的核苷酸序列如SEQ ID NO:14所示。
本发明所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,选自下列组 中的一种:
a)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的氨基酸序列如SEQ ID NO:19或20所示的序列的部分或全部所示;
b)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的氨基酸序列与SEQ ID NO:19或20所示氨基酸的序列同一性程度为至少大约为90%、91%、92%、93%、94%、95%、96%、97%、98%或至少99%;
c)编码嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸的核酸序列在严格条件下,与SEQ ID NO:19或20所示的氨基酸序列的核苷酸序列杂交;
d)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸与SEQ ID NO:19或20所示氨基酸的序列差异不超过10、9、8、7、6、5、4、3、2或不超过1个氨基酸;
e)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸序列具有SEQ ID NO:19或20所示的,包括取代、缺失和/或插入一个或多个氨基酸残基的氨基酸序列;或
f)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列为SEQ ID NO:21或22所示的序列的部分或全部;
g)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列与SEQ ID NO:21或22所示的核苷酸序列同一性程度为至少大约为90%、91%、92%、93%、94%、95%、96%、97%、98%或至少99%;
h)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列在严格条件下,与SEQ ID NO:21或22所示的核苷酸序列杂交;
i)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列与SEQ ID NO:21或22所示的序列差异不超过10、9、8、7、6、5、4、3、2或不超过1个核苷酸;
j)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列具有SEQ ID NO:21或22所示的,包括取代、缺失和/或插入一个或多个核苷酸的核苷酸序列。
优选的,本发明所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的氨基酸序列如SEQ ID NO:19或SEQ ID NO:20所示,或与SEQ ID NO:19 或SEQ ID NO:20至少约90%、91%、92%、93%、94%、95%、96%、97%、98%、99%、99.1%、99.2%、99.3%、99.4%、99.4%、99.5%、99.7%、99.8%或99.9%的氨基酸序列一致性的序列。编码上述嵌合抗原受体本发明所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列,如SEQ ID NO:21或SEQ ID NO:22所示。
本发明的第二方面,涉及一种编码上述任一嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的DNA。
本发明的第三方面,涉及上述任一嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ或上述DNA在制备嵌合抗原受体T细胞的应用。
本发明的第四方面,涉及一种anti-BCMA CAR-T细胞的制备方法,其中,所述方法包括以下步骤:
1)依次按信号肽、anti-BCMAscFv、人CD8α铰链区、人CD8α跨膜区、人4-1BB胞内区、人CD3ζ胞内区序列进行连接,获得anti-BCMA-CAR;
2)将anti-BCMA-CAR片段通过EcoRI-BamHI酶切连接至骨架载体pLVX-EF1a-GFP-N1上,测序验证正确后,获得pLVX-CAR;
3)将步骤2)获得的pLVX-CAR表达载体和包装质粒pMD2.G、psPAX2共同转染293T细胞,获得含有anti-BCMA-CAR的慢病毒颗粒;
4)将步骤3)获得的含有anti-BCMA-CAR的慢病毒颗粒感染T细胞,获得BCMA特异性CAR-T细胞。
本发明的第五方面,涉及包含上述所述的嵌合抗原受体或DNA的细胞、组织、器官或构建体,所述的细胞、组织、器官或构建体表达BCMA的特异性嵌合抗原受体。
优选的,所述细胞为T细胞。更优选的,所述T细胞由上述方法制备得到。
本发明的第六方面,涉及一种上述嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ或上述DNA或上述细胞、组织、器官或构建体在制备治疗肿瘤药物组合物中的应用。
本发明的第七方面,涉及一种治疗肿瘤的药物组合物,其中,所述药物组合物包括上述任一的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ、上述DNA 或上述的细胞、组织、器官或构建体。
本发明的第八方面,涉及一种上述任一的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ、DNA、或上述的细胞、组织、器官或构建体在治疗肿瘤中的应用。
以及一种治疗肿瘤的方法,所述方法包括向患者给予上述药物组合物。
优选的,所述肿瘤为任一恶性肿瘤,包括各器官的癌、肉瘤、血液系统的恶性肿瘤等等。例如白血病、淋巴瘤、多发性骨髓瘤、卵巢癌、乳腺癌、子宫内膜癌、结肠癌、直肠癌、胃癌、膀胱癌、肺癌、支气管癌、骨癌、前列腺癌、胰腺癌、肝和胆管癌、食管癌、肾癌、甲状腺癌、头颈部癌、睾丸癌、胶质母细胞瘤、星形细胞瘤、黑色素瘤、骨髓增生异常综合征、以及肉瘤。
更优选的,所述肿瘤为多发性骨髓瘤。
除非特别说明,本发明的实践将采取细胞生物学、细胞培养、分子生物学、转基因生物学、微生物学、重组DNA和免疫学的传统技术。这些技术在以下文献中进行了详细的解释。例如:MolecularCloningALaboratoryManual,2ndEd.,ed.BySambrook,FritschandManiatis(ColdSpringHarborLaboratoryPress:1989);DNACloning,VolumesIandII(D.N.Glovered.,1985);OligonucleotideSynthesis(M.J.Gaited.,1984);Mullisetal.U.S.Pat.No.4,683,195;NucleicAcidHybridization(B.D.Hames&S.J.Higginseds.1984);TranscriptionAndTranslation(B.D.Hames&S.J.Higginseds.1984);CultureOfAnimalCells(R.I.Freshney,AlanR.Liss,Inc.,1987);ImmobilizedCellsAndEnzymes(IRLPress,1986);B.Perbal,APracticalGuideToMolecularCloning(1984);theseries,MethodsInENZYMOLOGY(J.AbelsonandM.Simon,eds.-in-chief,AcademicPress,Inc.,NewYork),specifically,Vols.154and155(Wuetal.eds.)andVol.185,″ols.154and155(Wuetal.eds.)andVol.18,ed.);GeneTransferVectorsForMammalianCells(J.H.MillerandM.P.Caloseds.,1987,ColdSpringHarborLaboratory);ImmunochemicalMethodsInCellAndMolecularBiology(MayerandWalker,eds.,AcademicPress,London,1987);HandbookOfExperimentalImmunology,VolumesV(D.M.WeirandC.C.Blackwell,eds., 1986);andManipulatingtheMouseEmbryo,(ColdSpringHarborLaboratoryPress,ColdSpringHarbor,N.Y.,1986)。
本发明采用抗人BCMA单链抗体的基因序列,将其密码子优化。从NCBI数据库中搜索得到人CD8ɑ的信号区、铰链区、跨膜区和4-1BB及CD3ζ的胞内区基因序列,分段合成后应用重叠延伸PCR(SOE-PCR)进行拼接,然后克隆进入经改造的慢病毒表达载体pLVX-EF1a-GFP-N1中,利用该表达载体及包装质粒pMD2.G、psPAX2共同转染293T细胞获得慢病毒颗粒,然后感染人T淋巴细胞,使得T淋巴细胞表达该嵌合抗原受体。获得的CAR-T细胞在体外与RPMI8226细胞共培养,采用流式细胞仪检测细胞感染效率、ELISA法检测共培养上清中细胞因子(IL2、TNFα、γ-IFN)水平以及
Figure PCTCN2017113221-appb-000001
非放射性细胞毒性法检测CAR-T细胞对RPMI 8226细胞的杀伤活性,以证实该嵌合抗原受体修饰的T淋巴细胞对肿瘤细胞的特异性杀伤作用。因此本发明所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ可在肿瘤的细胞免疫治疗中得到应用。
附图说明
图1:嵌合抗原受体anti-BCMA-CD8a-41BB-CD3ζ的结构示意图,其中,SP为CD8α信号肽区,VL为轻链可变区,VH为重链可变区,CD8hinge为铰链区,CD8TM为跨膜区,4-1BB为人的4-1BB胞内区,CD3ζ为人的CD3ζ胞内区;
图2:anti-BCMA-CD8a-41BB-CD3ζ慢病毒载体图谱;
图3:慢病毒感染T细胞96h后,用荧光显微镜观察T细胞的绿色荧光表达情况;
图4:WesternBlotting检测CAR-T细胞中外源CAR(~55KDa)的表达,其中1为Marker,2为MOI=10,3为MOI=20,4为MOI=40,5为MOI=80,6为阴性对照,30KDa的条带为内参GAPDH;
图5:流式细胞仪检测慢病毒感染T细胞72h后,CAR-T阳性细胞占总细胞数的比例,其中图5A为未经感染的T细胞,图5B为感染72h后的T细胞,框中为CAR-T阳性细胞群;
图6:ELISA检测IL-2细胞因子水平;
图7:ELISA检测IFNγ细胞因子水平;
图8:ELISA检测TNFa细胞因子水平;
图9:在不同的效:靶比例(1:4,2:4,4:4)条件下,VEC-T和CAR-T细胞对BCMA阳性细胞系RPMI8226的裂解百分率;
图10:在不同的效:靶比例(1:4,2:4,4:4)条件下,VEC-T和CAR-T细胞对BCMA阴性细胞系K562的裂解百分率;
图11:采用生物发光成像系统检测anti-BCMA CAR-T细胞对KMS-11细胞的作用,其中,转染空载体的T细胞与PBS为对照。
具体实施方式
下面结合具体实施例来进一步描述本发明。
实施例1:anti-BCMA-CD8a-41BB-CD3ζ基因序列的确定
抗人BCMA单链抗体的基因序列来源于BCMA免疫小鼠的杂交瘤细胞。从NCBI网站数据库搜索到人的CD8α信号肽区、铰链区和跨膜区、人的4-1BB胞内区和人的CD3ζ胞内区基因序列信息。将上述序列在网站http://www.jcat.de/进行密码子优化,以保证在编码氨基酸序列不变的情况下更适合人类基因表达。在信号肽基因序列氨基端引入Kozak序列和酶切位点,各段核苷酸序列送上海生工生物技术有限公司分别进行合成。各基因序列信息见SEQUENCE LISTING(SEQ ID NO:1-22),构建成功的嵌合抗原受体的结构如图1所示,其中,SP为CD8α信号肽区,VL为轻链可变区,VH为重链可变区,CD8hinge为铰链区,CD8 TM为跨膜区,4-1BB为人的4-1BB胞内区,CD3ζ为人的CD3ζ胞内区。
实施例2:anti-BCMA-CD8a-41BB-CD3ζ慢病毒载体的构建
采用重叠PCR将上述合成的序列依次按信号肽、anti-BCMAscFv、人CD8α铰链区、人CD8α跨膜区、人4-1BB胞内区、人CD3ζ胞内区序列进行连接,形成完整的anti-BCMA-CAR基因序列信息,获得CAR分子(下文称为“anti-BCMA-CAR”)。anti-BCMA-CAR的核苷酸序列如SEQ ID NO:21或SEQ ID NO:22所示,编码的氨基酸序列如SEQ ID NO:19或SEQ ID NO:20所示。
用EcoR I和BamH I双酶切anti-BCMA-CAR的核苷酸序列,经T4DNA连接酶连接插入经改造的慢病毒载体pLVX-EF1a-GFP-N1(Addgene)的EcoR I和BamH I酶切位点,转化到感受态DH5α大肠杆菌。将所获得的重组质粒送上海生工生物技术有限公司进行测序,将测序结果与拟合成的anti-BCMA-CAR序列比对来验证序列是否正确。anti-BCMA-CD8a-41BB-CD3ζ慢病毒载体图谱如图2 所示。
实施例3:含anti-BCMA-CAR慢病毒的包装和滴度测定
用无内毒素质粒大提试剂盒内(天根生物)的操作说明书提取慢病毒包装载体pMD2.G、psPAX2和anti-BCMA-CAR慢病毒载体共转染到293T细胞中,转染后48h和72h收集细胞上清,400rcf离心10min,去除上清中的细胞及细胞碎片。用0.45μm的滤膜过滤上清液,分装冻存,备用。
按照预期的病毒滴度(MOI),将病毒上清液梯度稀释后感染293T细胞,荧光显微镜检测GFP阳性率,根据公式推算出pLVX-CAR和pLVX-空载病毒原液的滴度分别约为2×106TU/mL和5×106TU/mL。经浓缩液浓缩后,滴度可达1×108TU/mL和2×108TU/mL。
实施例4:慢病毒感染人的T细胞
第一步:用Ficoll分离液(天津灏洋)和人T细胞富集抗体混合物分离获得较纯的CD3+T细胞,用含5%AB血清X-VIVO(LONZA)培养基调整细胞密度为1×106/mL。将细胞以1mL/孔接种到预先用抗人50ng/mLCD3抗体(北京义翘神州生物技术有限公司)和50ng/mL CD28抗体(北京义翘神州生物技术有限公司)过夜包被过的24孔板,再加入100IU/mL的白细胞介素2(PeproTech),刺激培养48小时后用实施例3制备得到的病毒感染;
第二步:T细胞活化培养后隔天,用PBS稀释至终浓度为15μg/mL的Retronectin(Takara)包被新的24孔板,每孔250μL。避光,4℃过夜备用。
第三步:T细胞活化培养两天后,取出上一步中Retronectin包被好的24孔板,吸弃包被液,每孔加入500μL含2%BSA的HBSS,室温封闭30min后,吸弃封闭液,用含2.5%HEPES的HBSS洗板两次。
第四步:将实施例3制备得到的病毒液加入上述T细胞中,每孔500μL,再加入终浓度为8μg/mL的polybrene,混匀后32℃,1500g,离心2h。置于培养箱内培养24h。
第五步:将感染24h后的细胞1000g离心10min,小心吸去上清,加入新鲜的含10%FBS的RPMI1640培养基重悬细胞至浓度1×106/mL,以1×106/孔接种到第三步包被好的24孔板中,再加入100IU/mL的白细胞介素2,病毒原液和polybrene用量同前,32℃,1500g,离心2h。然后置于培养箱内培养。
第六步:细胞感染后,每天观察细胞的密度,适时补加含IL-2 100IU/mL的T细胞培养液,使T细胞的密度维持在5×105/mL左右,使细胞扩增。感染72h后,荧光显微镜观察T细胞的绿色荧光表达情况并拍照。结果见图3。由此获得感染了实施例3所述慢病毒的CAR-T细胞,命名为anti-BCMA CAR-T细胞,即BCMA特异性CAR-T细胞,表达实施例2的anti-BCMA-CAR。
实施例5:anti-BCMA特异性嵌合抗原受体表达鉴定
实施例4中感染的人T细胞72h收集后用PBS洗一遍,用细胞蛋白提取试剂(RIPA)裂解细胞,提取感染后的T细胞的蛋白经10%的SDS-PAGE进行分离后,恒流(300mA,1h)转印至PVDF膜上,用兔抗CD3ζ(1:400)抗体孵育,4℃孵育过夜。用TBST洗涤3遍后,用HRP山羊抗兔的二抗(1:10000)室温孵育1h。加入DAB显色后,用Azure公司的C-300多功能成像系统进行成像,其结果如图4所示。由图4可知:本发明所构建的重组质粒能够检测到目的条带的表达,大小与预期CAR(~55kDa)一致,见图4中条带2-5;而未感染的T细胞没有条带,见图4条带6。
实施例6:流式细胞仪检测感染后T淋巴细胞的比例及表面表达
实施例4中感染的T细胞于72h后,收集约106个细胞,用buffer(含5%FBS的PBS)洗涤1次,离心弃去上清,加100μL buffer重悬。加一抗BCMA-biotin 1μg,于37℃孵箱中孵育40min。离心,弃上清,用300μL buffer洗涤1-2次。用100μLbuffer重悬,加入二抗streptavidin-PE(按照说明书的浓度),4℃冰箱孵育20-30min。离心,弃去上清,用300μL buffer洗涤1-2次。用300μLbuffer重悬,上机(BD Calibur)检测。
结果如图5所示,使用实施例4制备得到的慢病毒感染T细胞72小时后,抗BCMA+的CAR-T细胞约占总细胞数的50%。
实施例7:体外共培养测定CAR-T细胞的肿瘤杀伤能力
7.1样品的准备
1)离心收集感染一周后实施例4的anti-BCMA CAR-T细胞并重新悬浮在新鲜的含10%FBS的RPMI1640培养基中,调整浓度为2×106/mL。
2)对于实施例5anti-BCMACAR-T细胞和NT细胞,按如下所示将T细胞和靶细胞RPMI 8226共培养于6孔细胞培养板进行培养:
A:2×106/孔空载体感染的T细胞,VEC-T(图例中简称VECT)
B:2×106/孔anti-BCMACAR-T(图例中简称CART)
C:2×106/孔VEC-T,1×106/孔RPMI 8226(图例中简称VECT+R)
D:2×106/孔anti-BCMACAR-T,1×106/孔RPMI 8226(图例中简称CART+R)每组设置4复孔,将共培养细胞置于37℃孵育16h。
7.2ELISA法测定细胞因子水平
ELISA试剂盒(购自上海科兴商贸有限公司,货号:F14003-A),按试剂盒说明操作。
1)待测样品A,B,C,D组培养上清液,从-20℃取出,室温融化。
2)将浓缩洗涤液、浓缩稀释液、标准品、密封板条从冰箱中取出室温解冻,然后将浓缩洗涤液稀释至工作浓度(1×)。
3)标准品稀释:用标准品稀释液稀释标准品至2400ng/mL、1200ng/mL、600ng/mL、300ng/mL、150ng/mL。
4)加样:分别设置空白孔、标准品孔、待测样品孔。在酶标包被板上标准品孔加样50μL,待测样品孔中先加样品稀释液40μL,然后再加待测样品10μL(样品最终稀释度为5倍)。将样品加于酶标板孔底部,尽量不要触及孔壁,轻轻晃动混匀。
5)温育:用封板膜封板后置于37℃温育30min。
6)洗涤:小心揭掉封板膜,弃去液体,甩干,每孔加满洗涤液,静置30秒后弃去,如此重复5次,甩干。
7)加酶:每孔加入酶标试剂50μL,空白孔除外。
8)温育:用封板膜封板后置于37℃温育30min。
9)洗涤:小心揭掉封板膜,弃去液体,甩干,每孔加满洗涤液,静置30秒后弃去,如此重复5次,甩干。
10)显色:每孔先加入显色剂A 50μL,再加入显色剂B 50μL,轻轻震荡混匀,37℃避光显色15min。
11)终止:每孔先加入终止液50μL,终止反应。
测定:以空白孔调零,450nm波长依序测量各孔的吸光度值(OD值),测定应在加终止液后15min内完成。测定结果绘出标准曲线,然后对照样品OD值查出 其浓度。结果见图6-8,其中,图6为ELISA检测IL-2细胞因子水平,图7为ELISA检测γ-IFN细胞因子水平,图8为ELISA检测TNFa细胞因子水平。anti-BCMA CAR-T和RPMI 8226共同孵育后,CAR-T细胞中的IL2检测值为1876.0pg/mL,而其余3组均小于50pg/mL(见图6);γ-IFN检测值为4070.3pg/mL,而其余3组均小于50pg/mL(见图7);TNFa检测值为1236.8pg/mL,而其余3组均小于65pg/mL(见图8)。anti-BCMA CAR-T和RPMI 8226共孵育后,CAR-T细胞中各因子的分泌量显著增高。
7.3CytoTox
Figure PCTCN2017113221-appb-000002
非放射性细胞毒性法检测CAR-T细胞对肿瘤细胞的杀伤作用
A.优化靶细胞数目
1)靶细胞准备:800rpm离心5min收集对数生长期RPMI 8226细胞,用含5%FBS的RPMI1640调整细胞浓度至1×106/mL。
2)乳酸脱氢酶(LDH)阳性对照配制:轻轻震荡LDH阳性对照以混匀,然后取2μL稀释到10mL的PBS+1%BSA(1:5000稀释)。
3)在U型底96孔板设置检验孔(三复孔):
a.培养基背景对照:110μL/孔含5%FBS的RPMI1640;
b.LDH阳性对照组:110μL/孔步骤2中稀释的LDH;
c.靶细胞最大LDH释放:设置靶细胞数目为0、5×103、1×104、2×104、3×104、4×104,用含5%FBS的RPMI1640补足每孔的体积为100μL,并在每孔加入10μL细胞裂解液(10×)。
4)将孔板置于37℃,5%CO2,饱和湿度培养箱孵育45min,250g离心4min。
5)LDH活性测定:
a.转移50μL步骤4中培养上清至另一个干净的96孔新酶标板;
b.解冻Assay Buffer,取12mL Buffer溶解LDH底物;
c.加LDH底物50μL至96孔酶标板,室温避光反应30min;
d.每孔加入50μL终止液酶促反应;
e.酶标仪检测490nm处吸光度值。
6)确定靶细胞的吸光度值至少是培养基背景对照吸光度值的两倍时的靶细胞数目为5×104
B.细胞毒性检测
1)靶细胞准备:800rpm离心5min收集对数生长期RPMI 8226或K562细胞,用含5%FBS的RPMI1640调整细胞浓度至5×106/mL。
2)效应细胞准备:1000rpm离心10分钟收集感染后一周的CAR-T和VEC-T细胞,用含5%FBS的RPMI1640调整细胞浓度至2.5×106/mL。
3)在U型底96孔板设置对照组和实验组(三复孔):
a.培养基背景对照:110μL/孔含5%FBS的RPMI1640;
b.体积校正对照孔:100μL 5%FBS的RPMI1640+10μL细胞裂解液(10×);
c.靶细胞自发释放组:10μL RPMI 8226或K562细胞(5×104细胞)+100μL 5%FBS的RPMI1640;
d.靶细胞最大释放:10μL RPMI 8226或K562细胞(5×104细胞)+10μL细胞裂解液(10×);
e.效应细胞自发释放:按不同的效:靶比例(1:4,2:4,4:4)加效应细胞5μL、10μL、20μL/孔,均用含5%FBS的RPMI1640补足体积为110μL/孔;
f.实验组:按照上述效:靶比例加入效应细胞和靶细胞,用含5%FBS的RPMI1640补足体积为110μL/孔;
4)将孔板置于37℃,5%CO2,湿润的培养箱孵育4小时。然后250g离心4min。在收集上清前45min,向靶细胞最大释放孔加入10μL细胞裂解液(10×)。
5)LDH活性测定:同前所述。
6)CTL活性计算:计算各组(3复孔)平均值(A490),按照下列公式计算CAR-T细胞裂解靶细胞的百分率,公式如下。
Figure PCTCN2017113221-appb-000003
如图9所示,随着E:T从1:4增加到4:4,anti-BCMA CAR-T细胞对BCMA阳性肿瘤细胞RPMI 8226的裂解率分别为34%、55%和62%,随着CAR-T细胞的数量增加,裂解效应显著增强。如图10所示,anti-BCMA CAR-T细胞对BCMA阴性细胞系K562裂解率均低于20%;综合如图9、图10所示,anti-BCMA CAR-T细胞对BCMA阳性肿瘤细胞RPMI 8226有很强的裂解效应,但对BCMA阴性细胞系K562不产生裂解。而空载体感染的T细胞对RPMI 8226和K562都不发生 裂解,表明anti-BCMA CAR-T细胞对BCMA阳性肿瘤细胞具有特异性识别和杀伤作用,也就是说anti-BCMA CAR-T细胞对其他正常细胞基本没有副作用。
实施例8:anti-BCMACAR-T细胞体内药效评价
KMS-11是人多发性骨髓瘤细胞系,细胞表面表达BCMA蛋白,并且可以作为免疫受损小鼠中的异种移植物生长。用萤火虫荧光素酶标记KMS-11细胞并植入NSG小鼠,建立起测试anti-BCMACAR-T疗效的多发性骨髓瘤模型。
将1×106个荧光素酶标记的KMS-11细胞通过尾静脉注射入NSG小鼠,培养9天后,通过尾静脉再注射8×106个anti-BCMA CAR-T细胞或转染空载体的T细胞或PBS。每天检测小鼠的健康状况,分别在1天,7天,14天,21天通过生物发光成像系统监测肿瘤负荷。小鼠麻醉前在腹膜内注射D荧光素,注射后5分钟用Xenogen成像麻醉小鼠,通过肿瘤细胞的生物发光评估疾病负担。结果如图11所示,转染空载体的T细胞组和PBS组的小鼠中显示很强的生物荧光信号,相比之下,anti-BCMACAR-T细胞组在第14天产生的生物荧光信号显著降低,在第21天几乎检测不到生物荧光信号,表明在体内anti-BCMACAR-T对BCMA阳性的肿瘤细胞有很好的清除率。
以上详细描述了本发明的优选实施例,但是,本发明并不限于上述实施例中的具体细节,在本发明的技术构思范围内,可以对本发明的技术方案进行多种简单变型,这些简单变型均属于本发明的保护范围。
另外需要说明的是,在上述具体实施方式中所描述的各个具体技术特征,在不矛盾的情况下,可以通过任何合适的方式进行组合,为了避免不必要的重复,本发明对各种可能的组合方式不再另行说明。
此外,本发明的各种不同的实施方式之间也可以进行任意组合,只要其不违背本发明的思想,其同样应当视为本发明所公开的内容。
Figure PCTCN2017113221-appb-000004
Figure PCTCN2017113221-appb-000005
Figure PCTCN2017113221-appb-000006
Figure PCTCN2017113221-appb-000007
Figure PCTCN2017113221-appb-000008
Figure PCTCN2017113221-appb-000009
Figure PCTCN2017113221-appb-000010
Figure PCTCN2017113221-appb-000011
Figure PCTCN2017113221-appb-000012
Figure PCTCN2017113221-appb-000013
Figure PCTCN2017113221-appb-000014
Figure PCTCN2017113221-appb-000015
Figure PCTCN2017113221-appb-000016
Figure PCTCN2017113221-appb-000017
Figure PCTCN2017113221-appb-000018
Figure PCTCN2017113221-appb-000019
Figure PCTCN2017113221-appb-000020

Claims (21)

  1. 一种嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述嵌合抗原受体包括单链抗体anti-BCMAscFv,铰链区、跨膜区和胞内区。
  2. 根据权利要求1所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述单链抗体anti-BCMAscFv由抗人BCMA单克隆抗体的重链可变区和轻链可变区串联而成,所述的抗人BCMA单克隆抗体来源于BCMA免疫小鼠的杂交瘤细胞。
  3. 根据权利要求1-2任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述铰链区和所述跨膜区均来源于人的CD8a区。
  4. 根据权利要求1-3任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述胞内区为人4-1BB胞内区及人CD3ζ胞内区。
  5. 根据权利要求1-4任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,还包括人的CD8α信号肽区。
  6. 根据权利要求5所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,在所述CD8α信号肽区氨基端对应的基因序列引入Kozak序列和/或酶切位点序列。
  7. 根据权利要求2所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述抗人BCMA单克隆抗体的重链可变区的互补性决定区CDR-H1~CDR-H3的氨基酸序列如SEQ ID NO:1-5所示,所述抗人BCMA单克隆抗体的轻链可变区的互补性决定区CDR-L1~CDR-L3的氨基酸序列如SEQ ID NO:6-8所示。
  8. 根据权利要求1-7任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述的单链抗体anti-BCMAscFv的氨基酸序列如SEQ ID NO:9或SEQ ID NO:10所示,所述的单链抗体anti-BCMAscFv的核苷酸序列如SEQ ID NO:11或SEQ ID NO:12所示。
  9. 根据权利要求1-8任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述铰链区和跨膜区串联连接,其氨基酸序列如SEQ ID NO:15所示,所述铰链区和跨膜区串联的核苷酸序列如SEQ ID NO:16所示。
  10. 根据权利要求4所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述胞内区4-1BB-CD3ζ的氨基酸序列如SEQ ID NO:17所示,编 码所述胞内区4-1BB-CD3ζ的核苷酸序列如SEQ ID NO:18所示。
  11. 根据权利要求5所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,所述的CD8α信号肽区的氨基酸序列如SEQ ID NO:13所示,编码所述CD8α信号肽区的核苷酸序列如SEQ ID NO:14所示。
  12. 根据权利要求1所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,其特征在于,选自下列组中的一种:
    a)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的氨基酸序列如SEQ ID NO:19或20所示的序列的部分或全部所示;
    b)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的氨基酸序列与SEQ ID NO:19或20所示氨基酸的序列同一性程度为至少大约为90%、91%、92%、93%、94%、95%、96%、97%、98%或至少99%;
    c)编码嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸的核酸序列在严格条件下,与编码SEQ ID NO:19或20所示的氨基酸序列的核苷酸序列杂交;
    d)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸与SEQ ID NO:19或20所示氨基酸的序列差异不超过10、9、8、7、6、5、4、3、2或不超过1个氨基酸;
    e)嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ氨基酸序列具有SEQ ID NO:19或20所示的,包括取代、缺失和/或插入一个或多个氨基酸残基的氨基酸序列;或
    f)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列为SEQ ID NO:21或22所示的序列的部分或全部;
    g)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列与SEQ ID NO:21或22所示的核苷酸序列同一性程度为至少大约为90%、91%、92%、93%、94%、95%、96%、97%、98%或至少99%;
    h)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列在严格条件下,与SEQ ID NO:21或22所示的核苷酸序列杂交;
    i)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列与SEQ ID NO:21或22所示的序列差异不超过10、9、8、7、6、5、4、3、2或不超过1个核苷酸;
    j)编码该嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的核苷酸序列具有SEQ ID NO:21或22所示的,包括取代、缺失和/或插入一个或多个核苷酸的核苷酸序列。
  13. 一种编码权利要求1-12任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ的DNA。
  14. 权利要求1-12任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ或权利要求13所述的DNA在制备嵌合抗原受体T细胞的应用。
  15. 一种anti-BCMA CAR-T细胞的制备方法,其特征在于,所述方法包括以下步骤:
    1)依次按信号肽、anti-BCMAscFv、人CD8α铰链区、人CD8α跨膜区、人4-1BB胞内区、人CD3ζ胞内区序列进行连接,获得anti-BCMA-CAR;
    2)将anti-BCMA-CAR片段通过EcoRI-BamHI酶切连接至骨架载体pLVX-EF1a-GFP-N1上,经测序验证,获得pLVX-CAR;
    3)将步骤2)获得的pLVX-CAR表达载体和包装质粒pMD2.G、psPAX2共同转染293T细胞,获得含有anti-BCMA-CAR的慢病毒颗粒;
    4)将步骤3)获得的含有anti-BCMA-CAR的慢病毒颗粒感染T细胞,获得BCMA特异性CAR-T细胞。
  16. 包含权利要求1-12任一所述的嵌合抗原受体或权利要求13所述的DNA的细胞、组织、器官或构建体,其特征在于,所述的细胞、组织、器官或构建体表达BCMA的特异性嵌合抗原受体。优选的,所述细胞为T细胞。更优选的,所述T细胞由权利要求15所述的方法制备得到。
  17. 权利要求1-12任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,或权利要求13所述的DNA,或权利要求16所述的细胞、组织、器官或构建体在制备治疗肿瘤药物组合物中的应用。
  18. 一种治疗肿瘤的药物组合物,其特征在于,所述药物组合物包括权利要求1-12任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,或权利要求13所述的DNA,或权利要求16所述的细胞、组织、器官或构建体。
  19. 一种治疗肿瘤的方法,其特征在于,向患者给予权利要求18所述的药物组合物。
  20. 根据权利要求19或20所述的方法,其特征在于,所述的肿瘤为多发性骨髓瘤。
  21. 权利要求1-12任一所述的嵌合抗原受体anti-BCMAscFv-CD8a-41BB-CD3ζ,或权利要求13所述的DNA,或权利要求16所述的细胞、组织、器官或构建体,或权利要求18所述的药物组合物在治疗肿瘤中的应用。
PCT/CN2017/113221 2017-11-03 2017-11-28 Bcma特异性嵌合抗原受体t细胞及其应用 WO2019085102A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711067947.4A CN109748968B (zh) 2017-11-03 2017-11-03 Bcma特异性嵌合抗原受体t细胞及其应用
CN201711067947.4 2017-11-03

Publications (1)

Publication Number Publication Date
WO2019085102A1 true WO2019085102A1 (zh) 2019-05-09

Family

ID=66332740

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/113221 WO2019085102A1 (zh) 2017-11-03 2017-11-28 Bcma特异性嵌合抗原受体t细胞及其应用

Country Status (2)

Country Link
CN (1) CN109748968B (zh)
WO (1) WO2019085102A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113061185A (zh) * 2020-01-02 2021-07-02 益科思特(北京)医药科技发展有限公司 一种bcma抗体的制备方法与应用

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114222758A (zh) * 2019-08-20 2022-03-22 武汉华大吉诺因生物科技有限公司 抗bcma抗体及其在car-t领域中的应用
CN112851815B (zh) * 2020-03-17 2023-08-08 西安宇繁生物科技有限责任公司 抗bcma抗体或其抗原结合片段及其制备方法和应用
CN111848820B (zh) * 2020-07-31 2022-05-17 广东昭泰体内生物医药科技有限公司 Cd19和bcma双靶点嵌合抗原受体及其应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104379179A (zh) * 2012-04-11 2015-02-25 美国卫生和人力服务部 靶向b-细胞成熟抗原的嵌合抗原受体
CN106687483A (zh) * 2014-07-21 2017-05-17 诺华股份有限公司 使用人源化抗‑bcma嵌合抗原受体治疗癌症
CN106795217A (zh) * 2014-07-24 2017-05-31 蓝鸟生物公司 Bcma嵌合抗原受体
WO2017130223A2 (en) * 2016-01-29 2017-08-03 Virocan Therapeutics Pvt. Ltd. A chimeric antigen receptor specific to b-cell maturation antigen, a recombinant expression vector and a method thereof
US20170281766A1 (en) * 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105837693A (zh) * 2016-05-30 2016-08-10 李斯文 一种基于bcma的抗原嵌合受体及其制备方法和应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104379179A (zh) * 2012-04-11 2015-02-25 美国卫生和人力服务部 靶向b-细胞成熟抗原的嵌合抗原受体
CN106687483A (zh) * 2014-07-21 2017-05-17 诺华股份有限公司 使用人源化抗‑bcma嵌合抗原受体治疗癌症
CN106795217A (zh) * 2014-07-24 2017-05-31 蓝鸟生物公司 Bcma嵌合抗原受体
WO2017130223A2 (en) * 2016-01-29 2017-08-03 Virocan Therapeutics Pvt. Ltd. A chimeric antigen receptor specific to b-cell maturation antigen, a recombinant expression vector and a method thereof
US20170281766A1 (en) * 2016-04-01 2017-10-05 Kite Pharma, Inc. Chimeric antigen and t cell receptors and methods of use

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113061185A (zh) * 2020-01-02 2021-07-02 益科思特(北京)医药科技发展有限公司 一种bcma抗体的制备方法与应用
CN113061185B (zh) * 2020-01-02 2023-08-08 益科思特(北京)医药科技发展有限公司 一种bcma抗体的制备方法与应用

Also Published As

Publication number Publication date
CN109748968A (zh) 2019-05-14
CN109748968B (zh) 2020-12-01

Similar Documents

Publication Publication Date Title
EP3503918B1 (en) Adenovirus armed with bispecific t cell engager (bite)
CN110818802B (zh) 一种嵌合t细胞受体star及其应用
US20160215261A1 (en) Nucleic Acid Of Coded GPC3 Chimeric Antigen Receptor Protein And T Lymphocyte Expressing GPC3 Chimeric Antigen Receptor Protein
TW201934574A (zh) 一種結合bcma的嵌合抗原受體(car)及其應用
WO2019085102A1 (zh) Bcma特异性嵌合抗原受体t细胞及其应用
JP2023052263A (ja) 腫瘍形質導入のための組成物及び方法
CN107841506A (zh) 靶向间皮素的嵌合抗原受体及其用途
WO2021254333A1 (zh) 一种合成t细胞受体抗原受体复合物及其应用
CN106117367B (zh) 一种her-3特异性嵌合抗原受体及其应用
WO2023104171A1 (zh) 分泌双特异性抗体的嵌合抗原受体t细胞及其制备方法和应用
CN112522208A (zh) 一种转基因肿瘤浸润淋巴细胞及其用途
WO2022135578A1 (zh) Claudin18.2嵌合抗原受体以及其用途
CN111378624B (zh) 一种靶向性抗肿瘤t细胞及其制备方法和应用
WO2022151959A1 (zh) 靶向b7-h3的car-t细胞及其在急性髓系白血病治疗中的应用
CN116064620A (zh) 一种增强向肿瘤部位浸润能力的car-nk细胞制备及应用
WO2021135178A1 (zh) 一种增强型t细胞受体star及其应用
WO2023125813A1 (zh) 抗间皮素纳米抗体嵌合抗原受体及其应用
WO2024119769A1 (zh) 增强向肿瘤部位浸润能力的car-nk细胞制备及应用
WO2023274355A1 (zh) 改造的间充质干细胞和免疫效应细胞联合治疗肿瘤
WO2023199068A1 (en) Mesothelin binders
WO2019014891A1 (zh) 一种用于治疗肿瘤的egfr单域抗体cart及其应用
CN118005794A (zh) 一种抗cldn18.2的重链抗体、相关产品和用途
CN115572715A (zh) 一种靶向folr1和her2双打靶点car t的制备及应用
TW202204419A (zh) 嵌合抗原受體
CN116376920A (zh) Ceacam5靶向的car-t细胞的制备方法及应用

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: 17930707

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: 17930707

Country of ref document: EP

Kind code of ref document: A1