WO2020025039A1 - Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée - Google Patents

Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée Download PDF

Info

Publication number
WO2020025039A1
WO2020025039A1 PCT/CN2019/099000 CN2019099000W WO2020025039A1 WO 2020025039 A1 WO2020025039 A1 WO 2020025039A1 CN 2019099000 W CN2019099000 W CN 2019099000W WO 2020025039 A1 WO2020025039 A1 WO 2020025039A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
target
expression vector
expression
Prior art date
Application number
PCT/CN2019/099000
Other languages
English (en)
Chinese (zh)
Inventor
周剑峰
徐皓
胡广
杨永坤
Original Assignee
南京驯鹿医疗技术有限公司
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 南京驯鹿医疗技术有限公司 filed Critical 南京驯鹿医疗技术有限公司
Priority to CN201980049205.XA priority Critical patent/CN112469734A/zh
Publication of WO2020025039A1 publication Critical patent/WO2020025039A1/fr

Links

Images

Classifications

    • 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
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes

Definitions

  • the present invention relates to T cells (CAR T cells) expressing a Chimeric Antigen Receptor (CAR), expression vectors for transforming T cells, and their use in the treatment of multiple myeloma.
  • CAR T cells CAR T cells
  • CAR Chimeric Antigen Receptor
  • SLAMF7 also known as CD319, CRACC, or CS1
  • SLAMF7 is a member of the transmembrane receptor signal transduction lymphocyte activation molecule family, which is expressed at high levels in myeloma cells and is involved in regulating the mutual adhesion of myeloma cells and bone marrow stromal cells effect.
  • Immunohistochemical analysis of a series of lymphomas and leukemias revealed that CS1, although present in all myeloma cases, is not expressed in most acute leukemias, B-cell lymphomas, and classic Hodgkin lymphomas.
  • CS1 Although researchers have investigated the feasibility of CS1 as a CAR T target.
  • CS1-CAR T cells target CS1-expressing multiple myeloma cell lines (such as NCI-H929, IM9, MM1S) and primary tumor cells isolated from patients with multiple myeloma. Compared with untransduced CAR control T cells, CS1-CAR T cells had significantly enhanced cytokine IFN- ⁇ and IL-2 secretion, and significantly increased the number of killed multiple myeloma cell lines or multiple myeloma patients. Generation of tumor cells 4 . These results demonstrate that the effect of CS1-CAR T cells is CS1-dependent. Importantly, CS1-CAR T cells can prolong the survival time of NSG mice transplanted with MM1S cells.
  • multiple myeloma cell lines such as NCI-H929, IM9, MM1S
  • CS1 has great potential as a CAR target for the treatment of multiple myeloma
  • data on CS1 as a CAR target in vitro and in vivo function data have been reported in the literature, but some researchers have pointed out that CS1 as the subject of technical difficulties CAR target and there is concern aspects 5.
  • CS1 is highly expressed on multiple myeloma cells, but on NK cells, T cells, B cells, and mature dendritic cells also expressed 6,7.
  • T cells are divided into helper T cells (CD4 +) and cytotoxic (CD8 +) T cells.
  • CD4 + T cells can proliferate to activate other types of immune cells that produce a direct immune response.
  • CD8 + T cells can kill target cells that produce antigenic responses.
  • CS1 in NK cells also play an important role in the interaction of activated NK cell function 9 by the same tropism of CS1.
  • NK cells express various activating and inhibiting receptors that recognize ligands on potential target cells. The balance between the signals from these receptors determines whether NK cells will be activated, killing target cells and secreting cytokines.
  • Antibodies and ligand-mediated stimulation experiments have demonstrated that CS1 plays an activating role in NK cells9,10,11 . The ability of CS1-deficient NK cells to kill CS1 + target cells is impaired.
  • CS1-deficient NK cells When encountering target cells, the ability of CS1-deficient NK cells to secrete interferon (IFN) also decreased. At the same time, the cytotoxicity of CS1-deficient NK cells to CS1-target cells also decreased.
  • IFN interferon
  • CS1 is involved in the interaction between NK cells and NK cells, and has the potential to promote NK cell function.
  • CS1 may have beneficial functions in T cells and NK cells, the expression of CS1 in T cells may cause difficulties in the preparation and application of CS1-CAR T cells.
  • CS1 protein expression level is lower than the amount of expression in CD8 + T cells in 5. Since CS1 is also expressed in some T cells and other immune cells, CS1-CAR T may cause these cells to be selectively killed and eliminated. Some researchers analyzed the ability of CS1-CAR T cells to recognize normal lymphocytes and found that they selectively kill CS1 + / high NK cells, CD4 + and CD8 + T cells and B cells.
  • CS1-CAR T cells showed a significant reduction in CS1 protein-positive CD4 + and CD8 + cells compared to control T cells after several days of culture.
  • the study also found that when CS1-CAR T cells were co-cultured with a normal lymphocyte population, compared to CD19-CAR T cells, CD4 + T cells, CD8 + T cells, and NK cells co-cultured with CS1-CAR T cells. The percentage of viable cells decreased, with the most significant decrease in the rate of viable cells of NK cells 5 .
  • CS1-CAR T cells caused only CS1-negative or low-expressing cells to survive in CS1-CAR T cells, which made it difficult for CS1-CAR T cells to expand; and because CD8 + T cells had more than CD4 + T cells The expression of CS1 will be more cleared, resulting in an abnormal ratio of CD4: CD8 in CS1-CAR T cells, reducing its cytotoxic activity in vitro and in vivo.
  • the invention provides, in one aspect, a T cell expressing a chimeric antigen receptor, the chimeric antigen receptor comprising an extracellular domain that recognizes a target antigen on the surface of a target cell, thereby mediating Killing of the target cell by the T cell; the T cell itself also expresses the target antigen, and in order to prevent the T cell from killing each other, the expression of the target antigen by the T cell is down-regulated.
  • the extracellular domain of the chimeric antigen receptor includes a single chain antibody derived from an antibody against the target antigen.
  • the target cells are tumor cells, especially multiple myeloma cells.
  • the target antigen is CS1.
  • the T cell down-regulates expression of the target antigen by expressing siRNA.
  • the siRNA is produced from a shRNA expressed by the T cell.
  • the target nucleic acid sequence of the shRNA includes a nucleotide sequence as shown in SEQ ID NO: 29.
  • the coding sequence of the shRNA includes a nucleotide sequence as shown in SEQ ID NO: 28.
  • the single chain antibody is derived from an anti-CS1 antibody and has an amino acid sequence as shown in SEQ ID NO: 20.
  • the amino acid sequence of the chimeric antigen receptor includes the CD8 ⁇ signal peptide, the single chain antibody, the CD8 hinge region, the CD28 transmembrane region, and the CD28 intracellular co-stimulatory domain in order from the N-terminus to the C-terminus. And 4-1BB intracellular co-stimulatory domain and CD3 ⁇ intracellular signaling domain.
  • the T cells are transformed with an expression vector including the coding sequence of the chimeric antigen receptor and an expression vector including the coding sequence of the shRNA, or are encoded by the chimeric antigen receptor.
  • the expression vector of the sequence and the coding sequence of the shRNA is transformed.
  • the present invention provides an expression vector for expression in T cells, comprising a coding sequence for a chimeric antigen receptor and a shRNA coding sequence, wherein the chimeric antigen receptor recognizes on the surface of a target cell And the shRNA down-regulates the expression of the target antigen in the T cells through the siRNA produced by the shRNA.
  • the chimeric antigen receptor includes an extracellular domain, a transmembrane domain, and an intracellular domain, the extracellular domain comprising a single chain antibody derived from an antibody against the target antigen.
  • the coding sequence of the shRNA is under the control of the H1 promoter.
  • the expression vector is selected from a lentiviral expression vector, a DNA plasmid expression vector, or a viral expression vector.
  • the target cells are tumor cells, especially multiple myeloma cells.
  • the expression vector uses pLVX-EF1 ⁇ -IRES-Puro as a backbone vector.
  • the target antigen is CS1.
  • the target nucleic acid sequence of the shRNA includes a nucleotide sequence as shown in SEQ ID NO: 29.
  • the coding sequence of the shRNA includes a nucleotide sequence as shown in SEQ ID NO: 28.
  • the single chain antibody is derived from an anti-CS1 antibody and has an amino acid sequence as shown in SEQ ID NO: 20.
  • the present invention provides a method for preparing a T cell expressing a chimeric antigen receptor, which comprises transforming the T cell with the expression vector described above.
  • the present invention provides a method for preventing mutual killing of T cells expressing a chimeric antigen receptor, comprising down-regulating the expression of a target antigen targeted by the chimeric antigen receptor in the T cells.
  • the target antigen is CS1.
  • the down-regulating comprises allowing the T cells to express shRNA, and the siRNA produced by the shRNA inhibits expression of the target antigen in the T cells.
  • the method is achieved by transforming the T cell with an expression vector comprising a coding sequence of the shRNA.
  • the present invention provides a method for treating multiple myeloma in a subject, comprising administering to the subject T cells expressing a chimeric antigen receptor, the chimeric antigen receptor targets CS1 on the surface of multiple myeloma cells, and the T cells also express shRNAs for inhibiting the expression of CS1 in the T cells.
  • the present invention provides the use of the above T cell or expression vector in the manufacture of a medicament for treating a disease caused by the proliferation of CS1-positive cells.
  • the disease is multiple myeloma or plasma cell leukemia.
  • the T cells provided by the present invention can be used to treat diseases caused by abnormal proliferation of CS1-positive cells (such as multiple myeloma) by targeting the CS1 antigen, and at the same time prevent the T cells from down-regulating the CS1 expression of the T cells to prevent The T cells kill each other, which is conducive to the expansion and survival of the T cells in vitro and in vivo.
  • CS1-positive cells such as multiple myeloma
  • FIG. 1 is a schematic diagram of the composition of mock-5.3-CAR elements.
  • the CAR consists of the following components: CD8 ⁇ signal peptide, BCMA-specific scFv (BCMA scFv), CD8 hinge region, CD28 transmembrane region (TM), CD28 intracellular costimulatory domain, 4-1BB intracellular costimulatory domain, and CD3 ⁇ cell Inner signal domain.
  • FIG. 2 is a schematic diagram of the composition of the mock-CS1-CAR element.
  • the CAR consists of the following components: CD8 ⁇ signal peptide, CS1-specific scFv (CS1scFv), CD8 hinge region, CD28 transmembrane domain (TM), CD28 intracellular costimulatory domain, 4-1BB intracellular costimulatory domain, and CD3 ⁇ intracellular Signal domain.
  • FIG. 3 is a schematic diagram of the SH3-5.3-CAR element structure. It consists of a gene encoding SH3 shRNA (SH3) capable of knocking down CS1 expression and mock-5.3-CAR shown in FIG. 1. Transcription of this shRNA is initiated by the H1 promoter.
  • SH3 shRNA SH3 shRNA
  • FIG. 4 is a schematic diagram of the structure of the SH3-CS1-CAR element. It consists of a gene encoding SH3 shRNA (SH3) capable of knocking down CS1 expression and mock-CS1-CAR shown in FIG. 2. Transcription of this shRNA is initiated by the H1 promoter.
  • SH3 shRNA SH3 shRNA
  • FIG. 5 is a schematic diagram of a BCMA-T2A-puro construct overexpressing BCMA, which includes BCMA, T2A, and a puromycin resistance gene (puro).
  • FIG. 6 is a schematic diagram of a CS1-IRES-puro construct overexpressing CS1, which includes CS1, IRES, and a puromycin resistance gene (puro).
  • Figure 7 shows the expression of BCMA on the surface of B-K562 cells as determined by flow cytometry.
  • the left peak is un-stained B-K562, and the right peak is B-K562 stained with APC anti-human CD269 (BCMA) Antibody.
  • BCMA APC anti-human CD269
  • Figure 8 shows the expression of CS1 on the surface of C-K562 cells measured by flow cytometry.
  • the left peak is unstained C-K562, and the right peak is C-K562 stained with Human CRACC / SLAMF7 APC-conjugated Antibody.
  • Figure 9 is a bar graph showing the expression of CS1 protein in CAR T cells and control T cells treated with SH3 knockdown.
  • Figure 10 shows the proliferation of CAR T cells and control T cells. Because CS1 is expressed on the surface of T cells, suicide will occur during CS1-CAR T culture, making it difficult for cells to expand. After the inventors knocked down the expression of CS1 in CAR T cells, the cells could expand normally as well as control T cells and BCMA-CAR T cells (mock-5.3 and SH3-5.3). The expansion curves of mock-5.3 and SH3-5.3 cells also showed no significant effect on cell proliferation after CS1 knockdown.
  • FIG. 11 shows the composition of various CAR T cell subpopulations. Knockdown of CS1 expression does not affect the proportion of cell subsets in CAR T cells.
  • CM and EM refer to T cells, T CM cells and T EM cells;
  • Figure 12 shows the in vitro killing ability of various CAR T cells to different target cells.
  • Mock-5.3 cells and SH3-5.3 cells can specifically kill BCMA-positive target cells (MM1S and B-K562), but compared with mock-5.3 cells, the killing effect of SH3-5.3 cells on MM1S is slightly reduced. It is possible that knocking down CS1 has an effect on the killing function of T cells, but further experiments need to be confirmed.
  • Mock-CS1 cells and SH3-CS1 cells can specifically kill CS1-positive target cells (MM1S and C-K562), and no weakening of the tumor-killing effect of SH3-CS1 cells is observed, which may be through knocking down CS1-CAR of CS1.
  • Reduced T-cell suicides offset the negative effects of knockdown of CS1 on T-cell killing function.
  • Figure 13 shows the results of CD107a detection on various CAR T cells and control T cells after target cell stimulation.
  • Mock-CS1 cells and SH3-CS1 cells were specifically degranulated when co-cultured with CS1-positive target cells (MM1S and C-K562), and no decrease in degranulation level of SH3-CS1 cells was observed.
  • Low CS1 leads to reduced suicide of CS1-CAR T cells, which offsets its adverse effects.
  • Figure 14 shows the expression of immune checkpoint molecules in various CAR T cells.
  • PD-1 and CTLA-4 (top) and TIM-3 and LAG3 (bottom) immune checkpoints are signs of T cell depletion.
  • the expression of the above four markers in the SH3-CS1 group is lower, suggesting that knockdown of CS1 CAR T cells are less depleted due to reduced suicide, suggesting that CS1-CAR T that knocks down CS1 may have better therapeutic potential.
  • an antibody refers to immunoglobulins secreted by plasma cells (effector B cells) and used by the body's immune system to identify and neutralize foreign substances (polypeptides, viruses, bacteria, etc.). This foreign substance is accordingly called an antigen.
  • the basic structure of an antibody molecule is a 4-mer consisting of two identical heavy chains and two identical light chains. According to the conservative differences in amino acid sequences, the heavy and light chains are divided into a variable region at the amino terminus and a constant region at the carboxy terminus. The variable regions of a heavy chain and a light chain interact to form an antigen-binding site. Thus, a complete antibody molecule includes two antigen-binding sites.
  • single chain antibody refers to a single peptide chain formed by linking the variable region of an antibody's heavy chain and the variable region of a light chain through a short peptide.
  • the variable region of the heavy chain and the variable region of the light chain form an antigen-binding site through non-covalent bond interactions, which can better retain the affinity activity of the source antibody for the antigen.
  • chimeric antigen receptor refers to an engineered protein receptor molecule that can impart a desired specificity to an immune effector cell (such as a T cell), such as the ability to bind to a specific tumor antigen. These receptors are called “chimeric” because they are fusion proteins and are composed of components from different sources. Chimeric antigen receptors typically include an extracellular domain (or extracellular binding domain), a transmembrane domain (or transmembrane region), and an intracellular domain (or intracellular signaling domain). The extracellular domain typically includes a scFv sequence that is responsible for recognizing and binding to specific antigens (target antigens) on target cells.
  • Intracellular domains usually include immunoreceptor tyrosine activation motifs (ITAM), such as signaling domains derived from the CD3 ⁇ molecule, which are responsible for activating immune effector cells, which can increase the cytotoxicity, proliferative capacity, and prolong T cells. Survival time.
  • ITAM immunoreceptor tyrosine activation motifs
  • the chimeric antigen receptor can also include a signal peptide at the amino terminus responsible for localization of the nascent protein on the cell, and a hinge region between the scFv sequence and the transmembrane domain.
  • downstreamregulating refers to a reduction in the ability of a gene or coding sequence to express its target product (such as a protein or RNA) in a cell compared to normal levels. This can be achieved in a variety of ways, for example, by inhibiting the initiation of transcription, interfering with mRNA translation, promoting mRNA degradation, or promoting degradation of expressed proteins.
  • siRNA small interfering RNA
  • siRNA refers to a double-stranded RNA molecule that is approximately 21 nt in length. They can be complementary to the homologous sequence in the target mRNA, causing the mRNA to lose function or be degraded, so it cannot be translated. protein.
  • One way to introduce siRNA into a cell is to express the corresponding short hairpin RNA (shRNA) molecule in the cell. The "loop" of the short hairpin RNA molecule is replaced by a nuclease (such as Dicer) in the cell. Enzymes) degrade to form siRNA, which plays a role in down-regulating the expression of target genes.
  • transformation refers to the introduction of an expression vector (e.g., a plasmid expression vector, a viral expression vector) containing a gene of interest into a host cell (e.g., a T cell) and allowing the gene of interest to be expressed in the host cell.
  • an expression vector e.g., a plasmid expression vector, a viral expression vector
  • a host cell e.g., a T cell
  • Processes include virus-mediated transduction and transfection using liposomes, calcium phosphate, microinjection, electroporation, and the like.
  • suicide in this context refers to a situation where the CAR T cells themselves also express the target antigen, which results in the mutual recognition and killing of cells within the CAR T cell population, as a whole It appears that this CAR T cell population is in a state of suicide.
  • a short hairpin RNA targeting CS1 mRNA is introduced into a lentivirus expression vector encoding a chimeric antigen receptor (CS1-CAR) targeting CS1.
  • CS1-CAR chimeric antigen receptor
  • the shRNA coding genes are transcribed in the T cells and processed to form siRNA.
  • the siRNA degrades CS1 mRNA through an RNA interference pathway, thereby reducing the expression level of CS1 protein in CS1-CAR T cells, and protecting the CS1-CAR T cells from mutual recognition and killing.
  • the inventors also studied the cell proliferation and cell subpopulation composition of knockdown CS1-expressing CAR T cells, and compared their in vitro killing function with CAR T cells that did not knock-down CS1.
  • CAR is a chimeric protein that fuses an antigen-binding domain that specifically recognizes a target antigen (such as CS1) with an intracellular signal transduction structure capable of activating or stimulating immune cells.
  • CAR includes extracellular domains, transmembrane domains, and intracellular domains.
  • the extracellular domain includes a single-chain antibody (scFv) capable of specifically binding to CS1, a CD8 ⁇ signal peptide, and a CD8 hinge region.
  • the hinge region is usually derived from a CD8 or IgG4 molecule, which is used to connect intracellular and extracellular proteins.
  • the inventors used a CD8 Hinger derived from CD8.
  • the transmembrane domain is a structure that connects the extracellular and intracellular domains of the CAR.
  • the CD28 transmembrane domain is used.
  • the CAR intracellular domain used in this study is an intracellular signaling domain that is capable of transducing the information of CS1CAR binding to human CS1 into the interior of immune effector cells to trigger effector cell functions such as activation, cytokine production, proliferation And cytotoxic activity).
  • the "first-generation" CAR intracellular signaling domain contains only CD3 ⁇
  • the "second-generation" CAR intracellular signaling domain contains a costimulatory molecule (eg, CD28 or 4-1BB4-1BB) and CD3 ⁇ .
  • a “third generation” CAR contains multiple costimulatory molecules (eg, CD28 and 4-1BB) and CD3 ⁇ .
  • CD28 and 4-1BB costimulatory molecules
  • CD3 ⁇ costimulatory molecules
  • Different researchers have carried out research with different targets and co-stimulation signals, and the comparison results between the second-generation CAR and the third-generation CAR have some differences.
  • Some studies have reported that recombinant T cells expressing "third-generation" CARs have significantly improved antitumor activity, survival cycle, and cytokine release12,13 . The results of Wilkie et al.
  • the pLVX-EF1 ⁇ -IRES-Puro vector (purchased from Miaoling Plasmid Platform) contains relevant elements required for lentivirus production, as well as elements capable of increasing virus titer and increasing transgene expression.
  • WPRE can promote RNA processing events and enhance the nuclear export of viral RNA, leading to increased viral titers produced by packaging cells;
  • Rev response elements (RRE) enhance the transport of unspliced viral RNA from the nucleus, further increasing viral titers;
  • the central polypurine / central termination sequence element (cPPT / CTS) generates a central DNA flap, thereby increasing nuclear import of the viral genome during target cell infection, resulting in improved vector integration and more efficient transduction.
  • pLVX-EF1 ⁇ -IRES-Puro contains restriction endonuclease sites and multiple cloning sites (MCS), so that researchers can subclon the DNA sequence of interest into this vector as needed.
  • MCS is a short DNA sequence containing multiple (up to 20) restriction sites, and is the standard configuration sequence of vector plasmids commonly used in genetic engineering. In MCS, each restriction site is usually unique, that is, they appear only once in a particular vector plasmid, and the restriction sites of different enzymes may overlap.
  • CD8 ⁇ signal peptide (nucleic acid sequence is SEQ ID NO: 1; amino acid sequence is SEQ ID NO: 2), BCMA scFv (nucleic acid sequence is SEQ ID NO: 11; amino acid The sequence is SEQ ID NO: 12), CD8 Hinger (nucleic acid sequence is SEQ ID NO: 3; amino acid sequence is SEQ ID NO: 4), CD28 transmembrane region and CD28 intracellular stimulation domain (nucleic acid sequence is SEQ ID NO: 5; amino acid sequence is SEQ ID NO: 6), 4-1BB intracellular co-stimulation domain (nucleic acid sequence is SEQ ID NO: 7; amino acid sequence is SEQ ID NO: 8), and CD3 ⁇ intracellular signal domain (nucleic acid sequence is SEQ ID NO: 9; the amino acid sequence is SEQ ID NO: 10), and EcoRI and MluI restriction enzyme restriction sites are designed at both ends.
  • the fusion gene fragment was gene synthesized by Nanjing Kingsray Biotechnology Company.
  • the above BCMA scFv consists of a heavy chain (the nucleic acid sequence is SEQ ID NO: 13; the amino acid sequence is SEQ ID NO: 14) and the light chain (the nucleic acid sequence is SEQ ID NO: 15; the amino acid sequence is SEQ ID NO: 16) through a linker peptide (The nucleic acid sequence is SEQ ID NO: 17; the amino acid sequence is SEQ ID NO: 18).
  • the synthetic gene fragment was subcloned into the EcoRI and MluI restriction enzyme restriction sites of the pLVX-EF1 ⁇ -IRES-Puro vector, and the product was transformed into E.
  • coli DH5 ⁇ competent cells purchased from Takara. (Ampicillin, Amp) screened for positive colonies. Pick the positive colonies and send them to Wuhan Tianyi Huiyuan Biotechnology Co., Ltd. to sequence the entire sequence of the plasmid. Inoculate the correct colonies into 250 mL of LB liquid medium containing Amp and shake the bacteria for 16-24 h at 37 ° C and 220 rpm. EndoFree Plasmid Giga Kit (purchased from Qiagen) was used to extract the plasmid from the bacterial solution, and the plasmid was named "pLVX-mock-5.3-CAR" (Fig. 1).
  • SEQ ID NO: 8 Amino acid sequence of artificially synthesized 4-1BB intracellular costimulatory domain KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL
  • SEQ ID NO: 17 DNA sequence of a synthetic linker peptide linking light and heavy chains in BCMA scFV GGCAGCACCAGCGGCTCCGGCAAGCCTGGCTCTGGCGAGGGCAGCACAAAGGGA
  • SEQ ID: NO: 18 Amino acid sequence of a synthetic linker peptide linking light and heavy chains in BCMA scFV GSTSGSGKPGSGEGSTKG
  • CD8 ⁇ signal peptide CD8 ⁇ signal peptide
  • CS1scFv nucleic acid sequence is SEQ ID NO: 19; amino acid sequence is SEQ ID NO: 20
  • CD8 Hinger CD28 transmembrane region and CD28
  • the -1BB intrastimulatory domain and the CD3 ⁇ intracellular signaling domain are designed with restriction sites for BamHI and MluI restriction enzymes at each end.
  • the fusion gene fragment was gene synthesized by Nanjing Kingsray Biotechnology Company.
  • the above CS1scFv consists of a heavy chain (the nucleic acid sequence is SEQ ID NO: 21; the amino acid sequence is SEQ ID NO: 22) and the light chain (the nucleic acid sequence is SEQ ID NO: 23; the amino acid sequence is SEQ ID NO: 24) through a connecting peptide ( The nucleic acid sequence is SEQ ID NO: 25; the amino acid sequence is SEQ ID NO: 26).
  • the gene fragment synthesized by the above gene was subcloned into the BamHI and MluI restriction enzyme restriction sites of the pLVX-EF1 ⁇ -IRES-Puro vector, and the plasmid pLVX-mock-CS1 was obtained by referring to the method for screening colonies and extracting plasmids above.
  • -CAR Figure 2.
  • SEQ ID: NO: 25 DNA sequence of a synthetic linker peptide linking heavy and light chains in CS1scFv
  • SEQ ID: NO: 26 Amino acid sequence of a synthetic linker peptide linking heavy and light chains in CS1scFv GGGGSGGGGSGGGGS
  • SEQ ID NO: 29 Target DNA sequence of synthetic SH3 shRNA GTCGGGAAACTCCTAACATAT
  • Lentivirus is a gene therapy vector developed based on human immunodeficiency virus (HIV). It has the ability to infect dividing cells and non-dividing cells and can be expressed in cells for a long period of time.
  • the lentivirus used in this study is a "suicide" virus, that is, the virus will no longer infect other cells after infecting the target cell, nor will it use the host cell to generate new virus particles.
  • Some genes in the lentivirus have been deleted and replaced by genes for exogenous purposes, which are pseudotyped viruses.
  • the pCMV-VSV-G vector contains the VSV-G gene and provides the envelope protein required for viral packaging.
  • the psPAX2 vector contains the gag gene of the HIV virus, which encodes the main structural protein of the virus; the pol gene, which encodes a virus-specific enzyme; the rev gene, which encodes a regulator that regulates the expression of the gag and pol genes.
  • 293T cells are human embryonic kidney epithelial cell lines derived from 293 cells and expressing the SV40 large T antigen. They are widely used for transient transfection to overexpress various target proteins, or for packaging viruses.
  • transfection reagents such as polyethyleneimine (PEI)
  • PEI polyethyleneimine
  • the lentiviral backbone carried on the pLVX vector was transcribed into viral RNA, and combined with psPAX2 and pCMV- Proteins translated from lentivirus-related genes carried on VSV-G are assembled into lentiviruses.
  • the supernatant was filtered through a 0.45 ⁇ m filter, and the filtrate was centrifuged at 30000 g at 4 ° C. for 2.5 h. Discard the supernatant and resuspend the pellet in pre-chilled PBS to obtain the corresponding LV-mock-5.3, LV-SH3-5.3, LV-mock-CS1, LV-SH3-CS1 lentiviral concentrates, and store at -80 ° C. spare.
  • FITC-conjugated AffiniPure Anti-Mouse IgG (H + L) (purchased from Jackson ImmunoResearch) is labeled with fluorescein, and it can bind to single-chain antibodies in CAR.
  • the fluorescence signal detected by flow cytometry can indirectly reflect the expression of CAR encoded by lentiviral vector in 293T cells, identify positive cells successfully infected by lentivirus, and calculate lentivirus based on the proportion of positive cells Active titer data.
  • a 6-well plate Into a 6-well plate, 5.0 ⁇ 10 5 cells / well 293T cells were inserted, and 0.1 ⁇ L, 0.5 ⁇ L, and 1 ⁇ L of lentivirus concentrated solution were added to each well, and a negative control was added without lentivirus concentrated solution.
  • the 6-well plate was cultured in an incubator containing 5% CO 2 at 37 ° C. Three days later, 293T cells were collected with Versene solution (purchased from Gibco), stained with FITC-conjugated AffiniPure Goat Anti-Mouse IgG (H + L), and detected by flow cytometry (instrument model: Beckman Cytoflex). Version 7.6.3) Analyze the proportion of CAR-positive 293T cells.
  • lymphocyte separation solution purchased from Tianjin Yeyang Biological Products Technology Co., Ltd.
  • density gradient centrifugation obtained from Miltenyi Biotech.
  • T cell culture medium To 1 L of CTS TM OpTmizer TM T Cell Expansion Basal Medium (purchased from Gibco) was added 26 ml of CTS TM OpTmizer TM T-cell Expansion supplement (purchased from Gibco), and L-glutamine was added to a final concentration of 2 mM (Purchased from Gibco), and IL-2 (purchased from Shuanglu Pharmaceutical) was added at a final concentration of 500 IU / ml.
  • Cells and magnetic beads Dynabeads TM HumanT-Activator CD3 / CD28 purchased from Gibco were added to the T cell culture medium at a ratio of 1: 1.
  • Magnetic beads Dynabeads TM HumanT-Activator CD3 / CD28 is used to stimulate T cells with an activation density of about 1 ⁇ 10 6 / ml. T cells were cultured at 37 ° C in a 5% CO 2 incubator overnight to obtain activated T cells.
  • puro is a puromycin resistance gene.
  • Puromycin is an aminoglycoside antibiotic produced by the fermentation and metabolism of Streptomyces alboniger, which kills Gram-positive bacteria, various animal and insect cells by inhibiting protein synthesis. Puromycin is commonly used to screen and maintain stable transfection of mammals containing puro resistance genes.
  • the "puro" in the vector name means that the vector contains a puro resistance gene.
  • the characteristics of puro resistance gene are used to select K562 that stably expresses BCMA or CS1.
  • T2A 2A peptide is a type of amino acid sequence.
  • T2A is a 2A element from the Thalassima virus.
  • the inventors used T2A to link BCMA and puromycin resistance genes, co-express BCMA and puromycin resistance genes in K562, and then screened through puromycin to obtain K562, which stably expresses BCMA protein, below Called B-K562.
  • IRES internal ribosome entry site, internal ribosome entry site
  • IRES internal ribosome entry site
  • IRES has been widely used in the construction of binary expression vectors.
  • IRES nucleotide sequence: SEQ ID NO: 36
  • CS1 and puromycin resistance genes were used to co-express CS1 and puromycin resistance genes, and then screened through puromycin to obtain K562, which is hereinafter referred to as C-K562.
  • CD8 ⁇ signal peptide nucleic acid sequence is SEQ ID NO: 1; amino acid sequence is SEQ ID NO: 2)
  • BCMA nucleic acid sequence is SEQ ID NO: 30; amino acid sequence is SEQ ID NO: 31
  • T2A nucleic acid sequence is SEQ ID NO: 34; amino acid sequence is SEQ ID NO: 35
  • puroR nucleic acid sequence is SEQ ID NO: 37; amino acid sequence is SEQ ID NO: 38
  • the fusion gene fragment was synthesized by Nanjing Kingsley Biotechnology Company, and then subcloned into the XbaI and MluI restriction enzyme sites of the pLVX-EF1 ⁇ -IRES-Puro vector. The colonies were screened and the plasmid was extracted in reference to Example 1. Method to obtain plasmid pLVX-BCMA-T2A-puro ( Figure 5).
  • SEQ ID NO: 34 DNA sequence of artificially synthesized T2A GAGGGCAGAGGAAGTCTTCTAACATGCGGTGACGTGGAGGAGAATCCCGGCCCT
  • Two expression vectors (pLVX-BCMA-T2A-puro and pLVX-CS1-IRES-puro) were mixed with pCMV-VSV-G helper plasmid and psPAX2 helper plasmid at a ratio of 6: 2: 3, respectively, and co-transfected 293T cells. 72 hours after transfection, the cell culture supernatant containing the virus was collected. Centrifuge at 3000 g for 5 min at 4 ° C. After the supernatant was filtered through a 0.45 ⁇ m filter, the virus solution was centrifuged at 30,000 g at 4 ° C. for 2.5 h.
  • Human CRACC / SLAMF7 APC-conjugated Antibody and APC anti-human CD269 (BCMA) Antibody are labeled with fluorescein, which can bind to CS1 protein and BCMA protein, respectively.
  • the fluorescence signal detected by flow cytometry can indirectly reflect the expression of BCMA or CS1 in 293T cells, identify positive cells successfully infected by lentivirus, and calculate the active titer of lentivirus based on the proportion of positive cells .
  • lentivirus concentrate LV-BCMA or LV-CS1
  • LV-BCMA or LV-CS1 lentivirus concentrate
  • Negative control The cells were cultured in a 37 ° C incubator containing 5% CO 2 . Three days later, 293T cells were collected with Versene solution, and the proportion of CS1 or BCMA-positive 293T cells was detected using Human CRACC / SLAMF7 APC-conjugated Antibody (purchased from R & D Systems) and APC anti-human CD269 (BCMA) Antibody (purchased from Biolegend).
  • the active titer of the LV-BCMA or LV-CS1 lentivirus concentrated solution was calculated by referring to the formula in Example 1.
  • RPMI 1640 medium purchased from GIBCO
  • B-K562 and C-K562 cells were stained with Human CRACC / SLAMF7 APC-conjugated Antibody and APC anti-human CD269 (BCMA) Antibody, respectively, detected by flow cytometry, and analyzed by FlowJo software.
  • shRNAs short hairpin RNAs
  • short hairpin RNAs are non-coding small RNA molecules designed to form hairpin structures.
  • the inventors introduced a short hairpin RNA targeting CS1 mRNA into a lentiviral expression vector encoding CS1CAR. After the lentivirus infects T cells and integrates the CS1-CAR and shRNA coding genes into the T cell genome, the shRNA coding genes are transcribed and processed to form siRNA. It degrades CS1 mRNA through the RNA interference pathway, thereby inhibiting or down-regulating the expression of CS1 gene in T cells.
  • Example 1 Take the lentiviral transduced T cells (mock-CS1, SH3-CS1, mock-5.3, and SH3-5.3) obtained in Example 1 and the control T cells (Table 1) that were not transduced by lentivirus, using FITC- conjugated AffiniPure Goat Anti-Mouse IgG (H + L) and Human CRACC / SLAMF7 APC-conjugated Antibody staining, and the expression of CAR and CS1 on the cell surface were detected by flow cytometry.
  • CS1 on the surface of control T cells not transduced with lentivirus and mock-5.3, SH3-5.3, mock-CS1, and SH3-CS1 transduced by lentivirus were 56%, 55.6%, 27.6%, 12.7, respectively. % And 4.21%.
  • the expression rate of CS1 on SH3-5.3 cell surface was lower than mock-5.3, indicating that SH3 shRNA can effectively knock down the expression of CS1 on T cell surface.
  • the expression rate of CS1 on the surface of mock-CS1 cells was lower than that of control T cells not transduced by lentivirus and mock-5.3 and SH3-5.3 transduced by lentivirus, probably because CS1-CAR T cells killed CS1 positive in the population The number of CS1 positive cells is reduced.
  • the expression rate of CS1 on the surface of SH3-CS1 cells decreased, which also showed that SH3 shRNA can effectively knock down the expression of CS1 on the surface of T cells (Figure 9).
  • T cells can be expanded in vitro by stimulation with CD3 / CD28 magnetic beads. Through continuous cell counting, the rate of T cell expansion in vitro can be observed, reflecting the T cell expansion in vitro.
  • SH3-CS1, SH3-5.3, mock-CS1, mock-5.3, and control T cells (T) without lentiviral transduction were cultured according to the conditions in Example 1, and the number of cells was recorded every 2 to 3 days. The time point of lentiviral transduction is recorded as 0 days, and the number of cells from 0 to 10 days is recorded.
  • the initial number of cells in the mock-CS1 group was 7 ⁇ 10 6 , and the remaining groups were 5 ⁇ 10 6. According to the culture conditions in Example 1, the cells were cultured for 10 days in vitro, except for the mock-CS1 group, which was expanded less than 10 times. The group normally expanded 25 to 35 times.
  • T cells are an inhomogeneous cell population and can be classified in many ways. According to the cell surface differentiation antigen (CD), it can be divided into two major subgroups of CD4 + and CD8 +; according to the different response to the antigen, it is divided into initial T cells ( T cells), activated T cells, and memory T cells.
  • CD4 + T cells can promote the proliferation and differentiation of B cells, T cells and other immune cells, and coordinate the interaction between immune cells.
  • CD8 + T cells are mainly responsible for the elimination of target cells through direct killing.
  • Memory T cells are derived from effector T cells and can also be directly transformed from the original T cells. Memory T cells can be divided into T CM (central memory T cells) and T EM (effect memory T cells).
  • the central memory T cells can quickly produce effects and up-regulate the expression of CD40L, and can secrete a large amount of IL-2 and proliferate multiple times, further differentiate into effector T cells, and maintain immune memory for a long time.
  • Effector memory T cells have an immune effect immediately after being stimulated by the antigen, exert cytotoxic effects and secrete effector molecules, but the ability to secrete IL-2 and proliferate is low. Therefore, T EM maintains immune memory for a short time, and mainly plays a role in the front line of immune defense.
  • T cells can be distinguished according to different surface antigens.
  • the initial T cells are characterized by CD45RA + CCR7 + CD62L high
  • T CM is characterized by CD45RA-CCR7 + CD62L high
  • T EM is characterized by CD45RA- CCR7 + CD62L low .
  • researchers can use the corresponding flow cytometry antibodies to analyze the proportion of each subgroup in T cells.
  • T cells can be divided into the following four subgroups based on the expression of CD45RA and CCR7: initial T cells, T CM cells, T EM cells, and T EMRA cells.
  • the initial T cells were CD45RA + CCR7 +
  • T CM cells were CD45RA-CCR7 +
  • T EM cells were CD45RA-CCR7-
  • T EMRA cells were CD45RA + CCR7-.
  • T cells Take lentivirus-transduced T cells (mock-CS1, SH3-CS1, mock-5.3, and SH3-5.3) and control T cells (T) that were not transduced by lentivirus in Example 1, and use FITC-conjugated AffiniPure Goat Anti-Mouse IgG (H + L), Pacific Blue TM anti-human CD8 Antibody (purchased from Biolegend), FITC Mouse Anti-Human CD45RA (purchased from BD Biosciences), PE anti-human CD197 (CCR7) Antibody (purchased from RND) ) Stain the cell surface and analyze the results with FlowJo software after detection by flow cytometry.
  • Multicolor flow cytometry was used to analyze the CD8 + ratio of T cells in each group, as well as the ratio of initial T cells, T CM cells, and T EM cells.
  • the proportion of CD8 + cells without control lentivirus transduction was 59.3%, among which the initial T cell proportion was 20.4%, the T CM cell proportion was 24.0%, and the T EM cell proportion was 55.6%.
  • the proportion of CD8 + cells in the Mock-5.3 group was 65.2%, of which the initial T cell ratio was 10.7%, the T CM cell ratio was 17.9%, and the T EM cell ratio was 71.4%.
  • the proportion of CD8 + cells in the SH3-5.3 group was 66.9%, of which the initial T cell ratio was 18.2%, the T CM cell ratio was 18.1%, and the T EM cell ratio was 63.6%.
  • the proportion of CD8 + cells in the Mock-CS1 group was 64.7%, of which the initial T cell ratio was 2.7%, the T CM cell ratio was 15.0%, and the T EM cell ratio was 82.3%.
  • the proportion of CD8 + cells in the SH3-CS1 group was 65.5%, of which the initial T cell ratio was 8.7%, the T CM cell ratio was 14.5%, and the T EM cell ratio was 81.5% (Table 2).
  • a calcein-AM ester (calcein-AM) release method was used to measure the tumor killing effect of CAR T cells.
  • Calcein-AM is a cell staining reagent that can fluorescently label cells. Methyl acetate is very lipophilic and can penetrate cell membranes. In living cells, Calcein-AM is cleaved by intracellular esterases to form calcein, which remains in the cells. Calcein can emit green fluorescence. When target cells are lysed, calcein is released into the supernatant. The tumor killing effect of CAR T cells was measured by detecting the fluorescence intensity of calcein in the supernatant 16 .
  • the MM1S cell line expresses CS1 and BCMA, and is a positive target cell of CS1-CAR T cells and BCMA-CAR T cells.
  • B-K562 is a K562 cell line that overexpresses BCMA.
  • C-K562 is a K562 cell line that overexpresses CS1.
  • K562 cell line expresses neither CS1 nor BCMA.
  • the target cells MM1S purchased from the Chinese Academy of Sciences Type Culture Collection Committee Cell Bank
  • the target cells B-K56, C-K562, and K562 prepared in Example 2 were taken. After counting, it was washed twice with 5% FBS (available from GIBCO) in PBS (available from GIBCO). The cell density was adjusted to 1 ⁇ 10 6 / ml. Add 10 ⁇ l Calcein-AM (purchased from Aladdin) solution to each 1ml of cell suspension, mix well, and incubate at 37 ° C in the dark for 30min.
  • the effector cells (mock-5.3, SH3-5.3, mock-CS1, and SH3-CS1) and control T cells prepared in Example 1 were counted, and then an appropriate amount of cells were taken and washed twice with 5% FBS-containing PBS. Adjust CAR + T cell density to 2.5 ⁇ 10 6 / ml according to the measured transfection efficiency.
  • the effector cells and the target cells were co-cultured in a 96-well plate at a ratio of the number of effector cells to the number of target cells of 50: 1, 10: 1, and 2: 1, respectively.
  • the killing efficiency of mock-5.3 cells on MM1S cells was 97.17%, 92.43%, and 66.89%, and the killing efficiency of K562 cells was 21.16%.
  • the tumor-killing efficiency of B-K562 cells was 100.00%, 87.40%, 36.16%
  • the tumor-killing efficiency of C-K562 cells were 12.26%, 7.80%, and 4.57% (Table 3).
  • the tumor killing efficiency of SH3-5.3 cells on MM1S cells was 79.89%, 60.30%, and 40.5%, and the killing efficiency of K562 cells was 4.55%, 7.14%, and 3.67%, respectively.
  • the tumor-killing efficiency was 92.52%, 68.68%, and 24.64%, and the tumor-killing efficiency on C-K562 cells was 1.18%, 4.74%, and 0.62%, respectively.
  • the killing efficiency of mock-CS1 on MM1S cells was 85.42%, 66.84%, and 52.77%, and the killing efficiency of K562 cells was 28.84%, 30.23%, and 15.74%, respectively.
  • the tumor efficiencies were 7.31%, 2.70%, 1.78%, and the tumor-killing efficiencies of C-K562 cells were 99.07%, 79.47%, and 36.38%, respectively.
  • the killing efficiency of SH3-CS1 on MM1S cells was 90.58%, 75.65%, and 42.52%, and the killing efficiency of K562 cells was 21.39%, 17.35%, and 5.84%, respectively.
  • the tumor efficiencies were 7.36%, 8.11%, and 0.71%, respectively.
  • the tumor-killing efficiencies of C-K562 cells were 100.00%, 76.00%, and 24.69%, respectively.
  • Untransduced lentivirus control T cells had no significant killing effect on all four cell lines.
  • the tumor killing efficiency of untransduced lentiviral control T cells against MM1S cells was 29.98%, 13.90%, and 2.87%, and the killing efficiency of K562 cells was 12.94%, 8.21%, and 4.49%.
  • the tumor-killing efficiency of B-K562 cells was 10.20%, 7.37%, and 4.99%, and the tumor-killing efficiency of C-K562 cells was 9.65%, 5.07%, and 1.23%, respectively.
  • Mock-5.3 cells and SH3-5.3 cells can specifically kill BCMA-positive target cells (MM1S and B-K562), but compared with mock-5.3 cells, the killing effect of SH3-5.3 cells on MM1S is slightly reduced. It may be that knocking down CS1 has a certain effect on the killing function of T cells, but further experiments need to be confirmed. Mock-CS1 cells and SH3-CS1 cells can specifically kill CS1-positive target cells (MM1S and C-K562), and no weakening of the tumor-killing effect of SH3-CS1 is observed, which may be through knocking down CS1-CAR of T1 Reduced cell suicide, offsetting the negative effect of knockdown of CS1 on T cell killing function ().
  • CD8 + T cell cytoplasm contains a high concentration of cytotoxic particles in the form of vesicles, which contain perforin and granzymes.
  • Lysosomal-associated membrane protein-1 (LAMP-1 or CD107a) is a highly glycosylated protein that is distributed on the surface of these vesicles.
  • CD107a With the occurrence of degranulation, CD107a is transported to the surface of cell membranes, so the expression of CD107a on the surface of T cell membranes can reflect the level of degranulation of T cells, and the detection of CD107a on the surface of T cells can reflect the effect of T cell lysis target cells.
  • CAR T cells (mock-5.3, SH3-5.3, mock-CS1, and SH3-CS1) prepared in Example 1, control T cells not transduced with lentivirus, and various target cells were taken. The cells were counted and all cell densities were adjusted to 2 ⁇ 10 6 / ml. 250 ⁇ l of each effector cell and 250 ⁇ l of different target cells were co-cultured in a 24-well plate (the number of effector cells and target cells in each well was 5 ⁇ 10 5 ). Set up effector cells without target cells. For co-cultivation, CTS TM OpTmizer TM medium without IL-2 was used.
  • Anti-CD107a flow antibody (purchased from BD Biosciences) was added at 20 ⁇ l / ml and incubated in a 37 ° C incubator. After 1 h, add monesin (purchased from BD Biosciences), 3 ⁇ l / well, and continue incubation for 3 h with PE / Cy7anti-human CD3 Antibody (purchased from Biolegend), Pacific Blue TM anti-human CD8 Antibody (purchased from Biolegend), PE Mouse Anti -Human CD107a (purchased from BD Biosciences) stained the cells and analyzed the level of CD107a on the surface of CD3 + CD8 + cell membranes.
  • CD107a expression was 12.3%, 49.6%, 57.8% and 15.4%, respectively.
  • the expression of CD107a was 10.7%, 24.1%, 32.4% and 10.5%, respectively.
  • mock-CS1 cells were co-cultured with K562, MM1S, B-K562, and C-K562 cells, CD107a expression was 12.1%, 32.0%, 11.7%, and 34.2%, respectively.
  • CD107a expression was 13.2%, 35.8%, 11.3%, and 46.4%, respectively.
  • CD107a expression was 12.3%, 49.6%, 57.8%, and 15.4%, respectively.
  • Mock-5.3 cells and SH3-5.3 cells were specifically degranulated when co-cultured with BCMA-positive target cells (MM1S and B-K562), but compared with mock-5.3 cells, degranulation levels of SH3-5.3 cells were weakened.
  • Mock-CS1 and SH3-CS1 were specifically degranulated when co-cultured with CS1-positive target cells (MM1S and C-K562), and no decrease in SH3-CS1 degranulation was observed. It is still possible to knock down CS1 CS1-CAR T cells reduced suicide, offsetting their adverse effects (Figure 13).
  • Immune checkpoints can be simply defined as signal molecules that inhibit the activation of T cells and prevent T cells from participating in immune responses.
  • T cells up-regulate the expression of inhibitory receptors such as PD-1 and CTLA-4 when activated or up-regulate ligands on APC through pro-inflammatory cytokines (such as PD-1 for PDL1 and PD-L2) to participate in the inhibition of TCR signals Conduction.
  • pro-inflammatory cytokines such as PD-1 for PDL1 and PD-L2
  • CAR T cells Take CAR T cells (mock-5.3, SH3-5.3, mock-CS1, and SH3-CS1) prepared in Example 1 and control T cells that have not been transduced with virus, and use PE anti-human CD279 (PD-1) Antibody (Purchased from Biolegend), PE anti-human CD152 (CTLA-4) Antibody (purchased from Biolegend), APC anti-human CD366 (Tim-3) Antibody (purchased from Biolegend), APC anti-human CD223 (LAG-3) Antibody (purchased from Biolegend), Pacific Blue TM anti-human CD8 Antibody, and FITC-conjugated AffiniPure Goat Anti-Mouse IgG (H + L) staining, measurement by flow cytometry, and data analysis.
  • PD-1 Antibody Purchased from Biolegend
  • CTLA-4 PE anti-human CD152
  • APC anti-human CD366 Tim-3) Antibody (purchased from Biolegend)
  • the PD-1 and CTLA-4 expression of CAR + cells in Mock-5.3 cells were 51.3% and 64.5%, respectively, and the average fluorescence intensity (MFI) of CAR + CD8 + cells was 80429 and 25508, respectively.
  • the expression of PD-1 and CTLA-4 of CAR + cells in SH3-5.3 cells was 58.0% and 55.9%, and the mean fluorescence intensity (MFI) of CAR + CD8 + cells was 87907 and 26866, respectively.
  • Mock-CS1 cells the expression of PD-1 and CTLA-4 of CAR + cells were 42.3% and 66.4%, and the average fluorescence intensity (MFI) of CAR + CD8 + cells' TIM-3 was 111750 and 59086, respectively.
  • the expression of PD-1 and CTLA-4 of CAR + cells in SH3-CS1 cells was 31.8% and 39.2%, and the mean fluorescence intensity (MFI) of CAR + CD8 + cells was 99556 and 37902, respectively.
  • PD-1 expression and CTLA-4 expression of untransduced lentiviral control T cells were 25.8% and 50.4%, respectively, and the average fluorescence intensity (MFI) of TIM-3 of CD8 + cells was 53383 and 17100, respectively.
  • Immunity checkpoints such as PD-1, CTLA-4, TIM-3, and LAG3 are signs of T cell depletion, and the expression of the above four markers in the SH3-CS1 group is lower, suggesting that knockdown of the CAR of CS1 due to reduced suicide in T cells Resulting in a lower degree of depletion, showing that CS1-CAR T cells that knock down CS1 may have better therapeutic potential (Figure 14).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

La présente invention concerne un lymphocyte T exprimant un récepteur antigénique chimérique. Le récepteur antigénique chimérique comprend un domaine extracellulaire, et le domaine extracellulaire reconnaît un antigène cible sur la surface d'une cellule cible, ce qui permet de favoriser la destruction de la cellule cible par le lymphocyte T. Le lymphocyte T exprime également lui-même l'antigène cible, et pour empêcher que les lymphocytes T ne se détruisent les uns les autres, l'expression de l'antigène cible dans le lymphocyte T est régulée négativement. L'invention concerne en outre un vecteur d'expression comprenant une séquence codant pour le récepteur antigénique chimérique. Le lymphocyte T selon l'invention peut être utilisé pour traiter des maladies provoquées par une prolifération anormale des cellules positives CS1 par ciblage de l'antigène CS1, ainsi, les lymphocytes T sont empêchés de se détruire les uns les autres en raison de la régulation négative de l'expression de CS1 du lymphocyte T, facilitant l'expansion et la survie du lymphocyte T in vitro et in vivo.
PCT/CN2019/099000 2018-08-03 2019-08-02 Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée WO2020025039A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201980049205.XA CN112469734A (zh) 2018-08-03 2019-08-02 表达嵌合抗原受体的t细胞、该嵌合抗原相关表达载体以及它们的应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810880137.9 2018-08-03
CN201810880137 2018-08-03

Publications (1)

Publication Number Publication Date
WO2020025039A1 true WO2020025039A1 (fr) 2020-02-06

Family

ID=69231408

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/099000 WO2020025039A1 (fr) 2018-08-03 2019-08-02 Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée

Country Status (2)

Country Link
CN (1) CN112469734A (fr)
WO (1) WO2020025039A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023116782A1 (fr) * 2021-12-21 2023-06-29 上海驯鹿生物技术有限公司 Anticorps entièrement humain ciblant gprc5d et récepteur antigénique chimérique (car) et son utilisation
CN116410331B (zh) * 2021-12-31 2024-01-30 合源生物科技(天津)有限公司 靶向cs1的嵌合抗原受体、靶向bcma/cs1的双特异性嵌合抗原受体及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105377897A (zh) * 2013-05-03 2016-03-02 俄亥俄州创新基金会 Cs1-特异性嵌合抗原受体工程化的免疫效应细胞
CN107429253A (zh) * 2014-12-05 2017-12-01 希望之城公司 Cs1靶向性嵌合抗原受体修饰的t细胞

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104829730A (zh) * 2015-04-14 2015-08-12 苏静 一种能联合免疫细胞增强肿瘤杀伤能力的双特异性抗体及其制备方法和应用
CN105906720A (zh) * 2016-05-16 2016-08-31 武汉汉密顿生物科技股份有限公司 靶向性嵌合抗原受体修饰的免疫细胞及其制备方法和应用

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105377897A (zh) * 2013-05-03 2016-03-02 俄亥俄州创新基金会 Cs1-特异性嵌合抗原受体工程化的免疫效应细胞
CN107429253A (zh) * 2014-12-05 2017-12-01 希望之城公司 Cs1靶向性嵌合抗原受体修饰的t细胞

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
GALETTO, R. ET AL.: "Bypassing the Constraint for Chimeric Antigen Receptor (CAR) Development in T-Cells Expressing the Targeted Antigen: Improvement of Anti-CSl CAR Activity in Allogenic TCRa/ CS 1 Double Knockout T-Cells for the Treatment of Multiple Myeloma (MM", BLOOD, vol. 126, no. 23, 3 December 2015 (2015-12-03), XP055684551 *
GOGISHVILI, T. ET AL.: "SLAMF7-CAR T cells eliminate myeloma and confer selective fratricide of SLAMF71 normal lymphocytes", BLOOD, vol. 130, no. 26, 31 October 2017 (2017-10-31), pages 2838 - 2847, XP055621481, DOI: 10.1182/blood-2017-04- *
NI, YAPING ET AL.: "Progress in Clinical Studies of Chimeric Antigen Receptor Engineered T Cells for Treatment of Childhood Cancer", CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS, no. 11, 30 November 2017 (2017-11-30), pages 1219 - 1223 *
ROTOLO, A. ET AL.: "The prospects and promise of chimeric antigen receptor immunotherapy in multiple myeloma", BRITISH JOURNAL OF HAEMATOLOGY, vol. 173, no. 3, 8 March 2016 (2016-03-08), pages 350 - 364, XP055620675 *
TAI, Y. T. ET AL.: "Anti-CSl humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces antibody-dependent cellular cytotoxicity in the bone marrow milieu", BLOOD, vol. 112, no. 4, 15 August 2008 (2008-08-15), pages 1329 - 1337, XP002571731 *
ZHANG, MIAOMIAO ET AL.: "Risk and Solutions to Chimeric Antigen Receptor-engineered T Cell -based Cancer Immunotherapy", ACTA PHARMACEUTICA SINICA, no. 7, 31 December 2016 (2016-12-31), pages 1032 - 1037 *

Also Published As

Publication number Publication date
CN112469734A (zh) 2021-03-09

Similar Documents

Publication Publication Date Title
US20230340113A1 (en) CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
JP6961497B2 (ja) キメラ抗体受容体(CARs)の構成およびその使用方法
CN108018299B (zh) 靶向bcma的嵌合抗原受体及其用途
JP2022064952A (ja) キメラ抗原受容体およびその使用方法
AU2014259675B2 (en) CS1-specific chimeric antigen receptor engineered immune effector cells
Moyes et al. Genetically engineered macrophages: a potential platform for cancer immunotherapy
BR112019018288A2 (pt) Métodos e composições para transduzir e expandir linfócitos e regular a atividade dos mesmos
JP7288503B2 (ja) 改変された抗cd19 car-t細胞
JP2018502583A (ja) 修飾された肝炎転写後調節エレメント
US20240041926A1 (en) Dendritic cell activating chimeric antigen receptors and uses thereof
JP7233720B2 (ja) ヒトメソセリンを特異的に認識する細胞表面分子、il-7、及びccl19を発現する免疫担当細胞
CA3154281A1 (fr) Vecteur retroviral pour therapie du recepteur universel
US20220241328A1 (en) Use of cd8-targeted viral vectors
WO2022199125A1 (fr) Cellule immunitaire pour la sécrétion autocrine de protéine de fusion il-15 et anti-pd1
CN111743923A (zh) 包含分离的重组溶瘤腺病毒和免疫细胞的治疗剂及其应用
JP2018500337A (ja) 炭酸脱水酵素ix特異的キメラ抗原受容体およびその使用方法
WO2020025039A1 (fr) Lymphocyte t exprimant un récepteur antigénique chimérique, vecteur d'expression associé à un antigène chimère et utilisation associée
WO2020019983A1 (fr) Cellule génétiquement modifiée utilisée pour traiter une tumeur
WO2023015217A1 (fr) Utilisation de vecteurs viraux ciblant cd4
WO2023024084A1 (fr) Récepteur antigénique chimérique et son utilisation
WO2024123895A2 (fr) Commutateur moléculaire servant à accorder l'affinité fonctionnelle de cellules immunitaires à leur cible
WO2023081878A2 (fr) Cellules immunitaires modifiées et leurs procédés d'utilisation
EP4381081A1 (fr) Utilisation de vecteurs viraux ciblant cd4
CN117098849A (zh) 靶向cd8的病毒载体的用途

Legal Events

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

Ref document number: 19845412

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: 19845412

Country of ref document: EP

Kind code of ref document: A1