WO2023005219A1 - Chimeric antigen receptor modified by ubiquitin coupling and immune cell - Google Patents

Chimeric antigen receptor modified by ubiquitin coupling and immune cell Download PDF

Info

Publication number
WO2023005219A1
WO2023005219A1 PCT/CN2022/080608 CN2022080608W WO2023005219A1 WO 2023005219 A1 WO2023005219 A1 WO 2023005219A1 CN 2022080608 W CN2022080608 W CN 2022080608W WO 2023005219 A1 WO2023005219 A1 WO 2023005219A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
ubiquitin
car
chimeric antigen
cancer
Prior art date
Application number
PCT/CN2022/080608
Other languages
French (fr)
Chinese (zh)
Inventor
王皞鹏
邱士真
李文涛
王琨
陈健
宋献民
Original Assignee
上海科技大学
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 上海科技大学 filed Critical 上海科技大学
Publication of WO2023005219A1 publication Critical patent/WO2023005219A1/en

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
    • A61K39/463Cellular 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
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma

Definitions

  • the invention relates to the field of biomedicine, in particular to a ubiquitin-coupled modified chimeric antigen receptor and immune cells.
  • Chimeric antigen receptor is an artificially engineered tumor-targeting antigen receptor.
  • CAR-T therapy is an adoptive immunotherapy that expresses CARs that specifically recognize antigens and transmit T cell signals on the patient's own T cells, enabling them to recognize and kill tumor cells in the patient's body.
  • the composition of CAR mainly includes: a single chain antibody (single chain antibody fragment, scFv) that can recognize and bind to a specific tumor antigen, and its specificity can realize precise recognition and "targeted attack” on tumor cells; T cell receptor (T cell The intracellular domain of the receptor (TCR) ⁇ chain can effectively activate T cells after the scFv structure recognizes tumor antigens, and the activated T cells can secrete a large amount of cytokines, and effectively kill tumor cells while rapidly proliferating themselves.
  • a single chain antibody single chain antibody fragment, scFv
  • T cell receptor T cell receptor
  • TCR T cell receptor
  • TCR The intracellular domain of the receptor (TCR) ⁇ chain can effectively activate T cells after the scFv structure recognizes tumor antigens, and the activated T cells can secrete a large amount of cytokines, and effectively kill tumor cells while rapidly proliferating themselves.
  • adding the signaling domain of co-stimulatory molecules such as CD28 or 41BB to the intracellular region of CAR can enhance the ability of CAR to activate T cells and improve the ability of CAR-T cells to activate T cells. ability to survive in vivo.
  • CAR-T therapy has achieved remarkable success in the clinical treatment of various types of tumors, especially for blood tumors.
  • CAR-T therapy still has many limitations in the treatment of solid tumors.
  • the existing optimal design strategies for CAR-T mainly include: reducing the signal of CAR by mutating and inactivating different combinations of ITAM regions in CD3 ⁇ ; mutating the co-stimulatory domain of CD28 to reduce the co-stimulatory signal of CD28; trying different structural combinations , explore the most suitable CAR structure for different targets and the lowest level of background self-activation; co-transduce transcription factors involved in the regulation of T cell proliferation and differentiation into T cells with CAR to reduce the self-activation of CAR-T cells Signaling, suppression of exhaustion of T cells, etc.
  • this type of design often needs to try CARs targeting different targets, which consumes a lot of manpower and material resources, and has the disadvantages of large errors and low universality. Therefore, a more practical and universal transformation aimed at the CAR structure itself is a more optimal solution.
  • the purpose of the present invention is to provide a ubiquitin-modified chimeric antigen receptor and immune cells to solve the problems in the prior art.
  • the present invention firstly provides a chimeric antigen receptor modification method, which includes coupling ubiquitin to the C-terminus of the chimeric antigen receptor.
  • the present invention provides a ubiquitin-modified chimeric antigen receptor, the ubiquitin-modified chimeric antigen receptor comprises a transmembrane domain, an intracellular domain and an extracellular domain, and the C-terminus of the intracellular domain is coupled with ubiquitin .
  • the present invention provides an isolated polynucleotide comprising nucleotides encoding the ubiquitin-modified chimeric antigen receptor.
  • the present invention provides a nucleic acid construct comprising the isolated polynucleotide.
  • the present invention provides an immune cell, which includes the nucleic acid construct or the exogenous polynucleotide integrated in the genome, or can express the ubiquitin-modified chimeric antigen receptor.
  • the immune cells are selected from CAR-T cells, CAR-NK cells, CAR-macrophages or CAR-TIL cells.
  • the present invention also provides a preparation method of the immune cells, the preparation method comprising introducing the nucleic acid construct or the polynucleotide into immune effector cells or stem cells producing immune effector cells.
  • the immune effector cells are selected from T cells, NK cells, macrophages or TIL cells.
  • the present invention also provides the use of the immune cells in the preparation of medicines for treating cancer or immune diseases.
  • the present invention also provides a treatment method for tumors or immune diseases, the treatment method comprising administering a therapeutically effective amount of the immune cells to a subject.
  • the ubiquitin-conjugated modified chimeric antigen receptor and immune cells of the present invention have the following beneficial effects: provide a method for the optimization and transformation of CAR-T, especially for CAR-T that is not capable of proliferating in solid tumors Provides solutions to the best problems.
  • the optimization and transformation scheme of CAR with high self-activation level which is simple and applicable to different targets, has the advantages of simple application, high repeatability, good universality, high degree of optimization, and good anti-tumor activity. Suitable for all kinds of current CAR-T cell production technologies, especially suitable for CAR-T therapeutic products developed for solid tumor targets. , Excessive exhaustion and other problems provide a new solution.
  • Figure 1 shows the detection of the self-activation level of CAR-T cell lines targeting different targets in the human T lymphoma cell line Jurkat, using the expression of indicator proteins such as CD69 and ICOS to measure the activation level of T cells In this case, the background self-activation level of these cell lines was evaluated.
  • the left panel shows the detected expression level of CD69, and the right panel shows the detected ICOS expression level.
  • CARs targeting different targets used in a large number of clinical and preclinical trials lead to high levels of background self-activation of T cells.
  • FIG. 2-1 to Figure 2-3 show that CD19-28WT-CAR (nucleotide sequence such as SEQ ID NO.12, amino acid sequence such as SEQ ID NO.28) and a CD19-targeting CAR C-terminal coupling Wild-type ubiquitin (wtUb) (nucleotide sequence such as SEQ ID NO.13, amino acid sequence such as SEQ ID NO.29) or monoubiquitin (monoUb) (nucleotide sequence such as SEQ ID NO.14, amino acid sequence such as The CD19-28wtUb-CAR and CD19-28MonoUb-CAR obtained from SEQ ID NO.30) were expressed in Jurkat T cell line (Fig. 2-1) or primary human T cells (Fig.
  • Figure 3-1 to Figure 3-4 show that after expressing a variety of monoubiquitin-coupled CD28 CARs targeting different targets in the Jurkat cell line, the effect of the modification on the background self-activation level of CAR-T was tested to verify this Universality of the program.
  • GD2 nucleotide sequence such as SEQ ID NO.15, amino acid sequence such as SEQ ID NO.31, the results as shown in Figure 3-2
  • GPC3 Nucleotide sequence such as SEQ ID NO.16, amino acid sequence such as SEQ ID NO.32, the results are shown in Figure 3-3
  • CD19 and CD22 as the dual target (nucleotide sequence such as SEQ ID NO.33, Amino acid sequence such as SEQ ID NO.34, results shown in Figure 3-4) the expression level of monoubiquitin-coupled CAR on the cell membrane, and the background self-activation signal of the corresponding CAR-T cells (with CD19 and CD22 as the
  • SEQ ID NO.35 The nucleotide sequence of the dual-target WT-CAR is shown in SEQ ID NO.35, and the amino acid sequence is shown in SEQ ID NO.36).
  • Figure 4-1 to Figure 4-4 show the detection of wild-type targets (ie CD19-28WT-CAR T cells, GD2-28WT-CAR T cells, GPC3-28WT-CAR T cells, GPC3-28WT-CAR T cells, CD19 ⁇ 22WT-CAR T cells) and CAR-T cells coupled with monoubiquitin (i.e. CD19-28MonoUb-CAR T cells, GD2-28MonoUb-CAR T cells, GPC3-28MonoUb-CAR T cells, CD19 ⁇ 22MonoUb-CAR T cells, CD19 ⁇ 22MonoUb-CAR T cells differentiation of T cells).
  • wild-type targets ie CD19-28WT-CAR T cells, GD2-28WT-CAR T cells, GPC3-28WT-CAR T cells, GPC3-28WT-CAR T cells, CD19 ⁇ 22MonoUb-CAR T cells, CD19 ⁇ 22MonoUb-CAR T cells differentiation
  • the differentiation status of T cells can be divided into CD45RA+CD62L+ (Stem cell memory T cell, T SCM ), CD45RA-CD62L+ (Central memory T cell, T CM ), CD45RA- CD62L- (Effector memory T cell, T EM ), and CD45RA+CD62L- (Effector T cell, T EFF ), the differentiation level increases successively, and the proliferation and differentiation ability decrease successively.
  • Figure 4-1 is CAR-T cells targeting CD19
  • Figure 4-2 is CAR-T cells targeting GD2
  • Figure 4-3 CAR-T cells targeting GPC3
  • Figure 4-4 shows CAR-T cells targeting both CD19 and CD22, the left figure in each figure is the detected CD4, and the right figure is the detected CD8), coupled with single
  • the modification of ubiquitin can effectively slow down the differentiation of T cells, so that more CAR-T cells remain in the TSCM state, which also implies that CAR-T cells coupled with monoubiquitin should have better persistence. proliferation and tumor killing ability.
  • Figure 5 shows the in vitro detection of the proliferation of GD2-targeting wild-type and monoubiquitin-coupled CAR-T cells under continuous stimulation of target cells.
  • the same amount of GD2-28WT-CAR and GD2-28MonoUb-CAR T cells were given the same amount of target cells (irradiated) to stimulate.
  • live cell counts were performed every 2 days to record cell proliferation.
  • the results of this experiment showed that in the presence and continuous stimulation of target cells, monoubiquitin-coupled CAR-T cells had lower background self-activation levels and cell differentiation degrees, and had better sustained proliferation capabilities.
  • Figure 6-1 to Figure 6-2 show the in vivo tumor killing experiments in tumor-bearing mice, verifying the functional differences between wild-type and monoubiquitin-coupled CAR-T cells targeting CD19.
  • the tumor-bearing mice were randomly divided into 3 groups, and the same amount of normal T cells not transfected with CAR, CD19 WT CAR-T cells and CD19 MonoUb CAR-T cells were given to the mice by tail vein injection respectively.
  • Imaging equipment in vivo imaging system, IVIS
  • IVIS in vivo imaging system
  • wild-type CAR-T cells have limited ability to control tumors, while CAR-T cells coupled with monoubiquitin can still effectively control tumor growth to a certain extent.
  • tumor clearance can be achieved ( Figure 6-1), which effectively improves the survival rate of tumor-bearing mice ( Figure 6-2), and its efficacy is significantly better than that of wild-type CAR-T cells.
  • Figure 7-1 to Figure 7-2 show the detection of CAR-T cell proliferation, differentiation and exhaustion in tumor-bearing mice.
  • the results showed that under the stimulation of tumor cells in vivo, CAR-T cells coupled with monoubiquitin still had a higher proportion of stem cell memory T cells (T SCM ) with higher sustained proliferation ability (Figure 7-1, The left picture shows CD4 CAR-T cells in the bone marrow, and the middle picture shows CD8 CAR-T cells in the bone marrow); and correspondingly, the number of CAR-T cells coupled with monoubiquitin is also significantly more than that of wild-type CAR-T cells ( Figure 7-1, the right figure is the count of CAR-T cells in the spleen), and the indicative proteins of its cell exhaustion (PD1, LAG3, TIM3, that is, Figure 7-2, the left figure is the CD4 CAR-T cells in the bone marrow, and the right figure The expression level of CD8 CAR-T cells in bone marrow is also significantly lower, which may
  • the present invention provides an optimized transformation scheme of CAR with high self-activation level that is simple and applicable to different targets.
  • a ubiquitin protein mutant (monoubiquitin, MonoUb for short) is coupled to the C-terminus of CARs targeting different targets to obtain MonoUb-CAR. Therefore, the present invention firstly provides a chimeric antigen receptor modification method, which includes coupling ubiquitin to the C-terminus of the chimeric antigen receptor.
  • Ubiquitin is a small protein present in all eukaryotes (most eukaryotic cells). Ubiquitin consists of 76 amino acids with a molecular weight of about 8.451kDa. Its main function is to mark proteins that need to be broken down so that they can be degraded by the 26S proteasome. For membrane proteins, especially some special ubiquitin modification types may be involved in the down-regulation, transport and vesicle sorting of membrane proteins, as well as the regulation of their signal transmission. Ubiquitin as referred to in this application refers to any wild-type ubiquitin or mutant ubiquitin from any eukaryotic cellular source, including mammals such as primates (e.g.
  • Ubiquitin is a very conserved protein. Almost all eukaryotic cells express ubiquitin since yeast, and its amino acid sequence is basically identical (yeast and human ubiquitin have only one amino acid difference).
  • the type of ubiquitin is wild-type ubiquitin or mutant ubiquitin.
  • Wild-type ubiquitin can be modified by intracellular endogenous ubiquitin but cannot be used as a substrate to modify other proteins, so it can form ubiquitin chains.
  • the ubiquitin is mutant ubiquitin. Because the mutant ubiquitin cannot be modified by endogenous ubiquitin, and the mutant ubiquitin cannot be used as a substrate to ubiquitinate other proteins, the mutant ubiquitin is monoubiquitin, so the mutant ubiquitin can also be Known as mutant monoubiquitin or monoubiquitin.
  • the ubiquitin is coupled to the C-terminus of the chimeric antigen receptor through a linker peptide.
  • the connecting peptides described in the present application may be commonly used connecting peptides in the art.
  • the connecting peptide is, for example, GSGGSG, GSGGSGG GSGGSGGG or GGGGSGGG or GGSGGGSGGGSAAA.
  • the present invention also provides a ubiquitin-modified chimeric antigen receptor, which includes a transmembrane domain, an intracellular domain, and an extracellular domain, and the C-terminus of the intracellular domain is coupled with ubiquitin white.
  • the ubiquitin-modified chimeric antigen receptor is obtained by the engineering method.
  • the transmembrane domain may include transmembrane domains of protein molecules such as CD8 ⁇ , CD28, and DAP10.
  • the amino acid sequence of CD8 ⁇ may include the following sequence: IYIWAPLAGTCGVLLLSLVITLYC.
  • NM_001145873 for the sequence of CD8 ⁇
  • NM_006139 for the sequence of CD28.
  • the intracellular domain may comprise a co-stimulatory domain and/or a signaling domain.
  • the intracellular domain may include one or a combination of several of the following: signal transduction domains of protein molecules such as 4-1BB, CD28, OX40, ICOS, CD3 ⁇ , and DAP10.
  • amino acid sequence of 4-1BB includes as follows:
  • the amino acid sequence of the CD3 ⁇ includes as follows:
  • the intracellular domain sequentially includes CD28 and CD3 ⁇ from the N-terminus to the C-terminus.
  • the chimeric antigen receptor includes an extracellular domain, a transmembrane domain, and an intracellular domain sequentially from the N-terminus to the C-terminus.
  • the chimeric antigen receptor includes a single-chain antibody, a transmembrane domain, and an intracellular domain in sequence from the N-terminus to the C-terminus.
  • the chimeric antigen receptor includes a single-chain antibody, a CD8 ⁇ transmembrane region, a 4-1BB co-stimulatory domain, and a CD3 ⁇ signaling domain sequentially from the N-terminus to the C-terminus.
  • the chimeric antigen receptor comprises a single-chain antibody, a CD28 transmembrane region, a CD28 co-stimulatory domain, and a CD3 ⁇ signaling domain sequentially from the N-terminus to the C-terminus.
  • the chimeric antigen receptor includes a single-chain antibody, a CD8 ⁇ transmembrane region, an OX40 co-stimulatory domain, and a CD3 ⁇ signaling domain sequentially from the N-terminal to the C-terminal.
  • the chimeric antigen receptor comprises a single-chain antibody, a CD8 ⁇ transmembrane region, an ICOS co-stimulatory domain, and a CD3 ⁇ signaling domain sequentially from the N-terminal to the C-terminal.
  • the chimeric antigen receptor comprises a single-chain antibody, a CD8 ⁇ transmembrane region, a 4-1BB co-stimulatory domain, and a CD3 ⁇ signaling domain sequentially from the N-terminus to the C-terminus.
  • the chimeric antigen receptor comprises a single-chain antibody, a CD28 transmembrane region, a CD28 co-stimulatory domain, an OX40 co-stimulatory domain, and a CD3 ⁇ signaling domain from the N-terminus to the C-terminus.
  • the chimeric antigen receptor includes CD8 ⁇ signal peptide, myc tag, scFv, extracellular domain composed of CD8 ⁇ hinge, transmembrane domain of CD8 ⁇ , CD28 and CD3 ⁇ in sequence from N-terminal to C-terminal Intracellular domains organized in tandem.
  • the ubiquitin-modified chimeric antigen receptor that is, the CD3 ⁇ of any one of the above chimeric antigen receptors is connected with ubiquitin through a linker peptide (or linker).
  • the single-chain antibody of the present invention is not specifically limited, and can be selected from any single-chain antibody.
  • Examples of single-chain antibodies used in the present invention include any one or more of CD19 scFv, GD2 scFv, GPC3 scFv, Her2 scFv, CSPG4 scFv, EGFR scFv, Meso scFv, TRBC1 scFv, CD133 scFv, and BCMA scFv.
  • the chimeric antigen receptor is a multi-target chimeric antigen receptor.
  • the ubiquitin referred to in this application is selected from wild-type ubiquitin or mutant ubiquitin.
  • amino acid sequence of wild-type ubiquitin is shown in SEQ ID NO.17.
  • the ubiquitin is selected from mutant ubiquitin.
  • the amino acid sequence of the mutant ubiquitin selected in the present invention is shown in SEQ ID NO.18.
  • the present invention provides an isolated polynucleotide comprising nucleotides encoding the ubiquitin-modified chimeric antigen receptor.
  • polynucleotides encoding the transmembrane domain, intracellular domain and extracellular domain can all be selected from polynucleotides in the prior art.
  • the polynucleotide encoding the ubiquitin-modified chimeric antigen receptor sequentially includes nucleotides encoding the CD8 ⁇ signal peptide, myc tag, scFv, and the ectodomain composed of the CD8 ⁇ hinge, Nucleotides encoding the transmembrane domain of CD8 ⁇ , nucleotides encoding the intracellular domain composed of CD28, CD3 ⁇ , and ubiquitin in tandem.
  • the nucleotide sequence of wild-type ubiquitin is shown in SEQ ID NO.1.
  • the ubiquitin is selected from mutant ubiquitin.
  • the polynucleotide sequence encoding mutant ubiquitin selected in the present invention is shown in SEQ ID NO.2.
  • the present invention provides a nucleic acid construct comprising the isolated polynucleotide.
  • nucleic acid construct refers to an artificially constructed nucleic acid segment that can be introduced into cells or tissues, and the nucleic acid construct is a non-viral vector or a viral vector.
  • the viral vectors are lentiviral vectors, adenoviral vectors, adeno-associated viral vectors, and baculoviral vectors.
  • the nucleic acid construct is a lentiviral vector, and the lentiviral vector includes a vector backbone, that is, an empty vector and an expression framework.
  • the empty vector includes various expression-controlling elements, including a promoter sequence, a transcription initiation sequence, an enhancer sequence, a selection element and a reporter gene.
  • the vector may also contain an origin of replication.
  • the empty vector is, for example, pHR-hEF1 ⁇ -IRES-EGFP empty vector.
  • the expression framework is the isolated polynucleotide.
  • vector refers to a nucleic acid fragment or polynucleotide fragment used to introduce or transfer one or more nucleic acids or one or more polynucleotides into a target cell or tissue.
  • vectors are used to introduce foreign DNA into another cell or tissue.
  • the vector may contain a bacterial resistance gene for growth in bacteria and a promoter for expressing a protein of interest in an organism.
  • DNA can be produced in vitro by PCR or any other suitable technique or techniques known to those skilled in the art.
  • the present invention provides an immune cell, which includes the nucleic acid construct or the exogenous polynucleotide integrated in the genome, or is capable of expressing the ubiquitin-modified chimeric antigen receptor.
  • the immune cells are selected from CAR-T cells, CAR-NK cells, CAR-macrophages or CAR-TIL cells.
  • the present invention also provides a preparation method of the immune cells, the preparation method comprising introducing the nucleic acid construct or the polynucleotide into immune effector cells or stem cells producing immune effector cells.
  • the immune effector cells are selected from T cells, NK cells, macrophages or TIL cells.
  • the present invention also provides the use of the immune cells in the preparation of medicines for treating cancer or immune diseases.
  • Cancer in this application refers to any medical condition that is mediated by the growth, proliferation or metastasis of tumor or malignant cells and causes solid tumors and non-solid tumors such as leukemia.
  • Tumor in the present invention refers to the solid substance of tumor and/or malignant cells.
  • the cancer is, for example, non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, esophageal cancer, mesothelioma, melanoma, head and neck cancer , thyroid cancer, sarcoma, prostate cancer, glioblastoma, cervical cancer, thymus cancer; leukemia, lymphoma, myeloma, mycoses fungoids (mycoses fungoids), Merkel cell carcinoma and other hematological malignancies, Such as classical Hodgkin lymphoma (CHL), primary mediastinal large B-cell lymphoma, T-cell/histiocytic-rich large B-cell lymphoma, EBV-positive and negative PTLD, and EBV-associated diffuse large B-cell lymphoma lymphoma (DLBCL), plasmablastic lymphoma, extranodal NK/T cell lympho
  • the immune diseases are, for example, systemic lupus erythematosus (SLE), autoimmune diabetes, psoriasis, vitiligo, scleroderma, and rheumatoid arthritis.
  • SLE systemic lupus erythematosus
  • autoimmune diabetes for example, systemic lupus erythematosus (SLE)
  • psoriasis vitiligo
  • scleroderma rheumatoid arthritis
  • Treatment or “therapy” for a condition includes preventing or alleviating a condition, reducing the rate at which a condition occurs or develops, reducing the risk of developing a condition, preventing or delaying the development of symptoms associated with a condition , to reduce or terminate symptoms associated with a condition, to produce a complete or partial reversal of a condition, to cure a condition, or a combination of the above.
  • treating or “therapy” can refer to inhibiting or slowing the growth, reproduction, or metastasis of tumor or malignant cells, or some combination thereof.
  • treatment or “therapy” includes eradicating all or part of the tumor, inhibiting or slowing tumor growth and metastasis, preventing or delaying tumor progression, or some combination of the above.
  • the present invention also provides a treatment method for tumors or immune diseases, the treatment method comprising administering a therapeutically effective amount of the immune cells to a subject.
  • the cancer is, for example, non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, esophageal cancer, mesothelioma, melanoma, head and neck cancer , thyroid cancer, sarcoma, prostate cancer, glioblastoma, cervical cancer, thymus cancer; leukemia, lymphoma, myeloma, mycoses fungoids (mycoses fungoids), Merkel cell carcinoma and other hematological malignancies, Such as classical Hodgkin lymphoma (CHL), primary mediastinal large B-cell lymphoma, T-cell/histiocytic B-rich lymphoma, EBV-positive and negative PTLD, and EBV-associated diffuse large B-cell lymphoma ( DLBCL), plasmablastic lymphoma, extranodal NK/T cell lymphoma, nas
  • the immune diseases are, for example, systemic lupus erythematosus (SLE), autoimmune diabetes, psoriasis, vitiligo, scleroderma, rheumatoid arthritis.
  • SLE systemic lupus erythematosus
  • autoimmune diabetes for example, systemic lupus erythematosus (SLE)
  • psoriasis vitiligo
  • scleroderma rheumatoid arthritis
  • the "therapeutically effective amount” or “effective dose” in the present invention refers to the dose or concentration of a drug that can effectively treat the disease or state associated with the antigen of the chimeric antigen receptor.
  • the therapeutically effective amount is at the dosage or concentration, the antibody or antigen-binding compound can eliminate all or part of the tumor, inhibit or slow down tumor growth, inhibit Mediating the growth or proliferation of cells in a cancerous state, inhibiting tumor cell metastasis, alleviating any symptoms or markers associated with a tumor or cancerous state, preventing or delaying the development of a tumor or cancerous state, or some combination of the above.
  • monoUb-CAR can effectively promote the endocytosis and degradation of CAR, greatly reduce the expression of CAR on the cell surface, realize a substantial decrease in the background self-activation level of immune cells such as CAR-T cells, and significantly reduce the over-differentiation of immune cells such as T cells. Speed, degree of functional exhaustion, etc.
  • monoUb-CAR significantly enhances the sustained proliferation ability of CD28 CAR-T cells in vitro and in vivo under target cell stimulation conditions, and effectively improves the tumorigenicity of CAR-T cells in tumor-bearing mice. killing ability, thereby improving the survival rate of mice.
  • the present invention compares the anti-tumor effects of CD28 monoUb CAR-T and CD28 WT CAR-T in vivo on a tumor mouse model.
  • CD28 monoUb CAR-T has a stronger proliferative response and longer-lasting proliferative ability; in terms of cell differentiation phenotype, whether in the spleen, blood, or tumor, the engineered CAR-T has accumulated more Stem cell memory T cells (Stem cell memory T cells, T SCM ) and reduced differentiation to terminal effector T cells. Therefore, the modified CAR-T can more effectively infiltrate and kill tumor tissues, and at the same injection dose of T cells, the modified CAR-T can more effectively control the development of tumors.
  • Stem cell memory T cells Stem cell memory T cells
  • Embodiment 1 Vector construction of CAR
  • the antigen-specific single-chain antibody (scFv) sequences of CD19, GD2, and GPC3 CAR used in the present invention are respectively from the clinically used FMC63, 14g2A, and GPC3 sequences; the dual-target CAR is the integration of specific antibodies targeting CD19 and CD22
  • the integration method refer to the report in the article "CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy".
  • the extracellular segment structure of CAR is composed of CD8 ⁇ signal peptide sequence, myc tag sequence, scFv sequence, and CD8 ⁇ hinge sequence in series; the transmembrane sequence is the transmembrane region sequence of CD8 ⁇ ; the intracellular segment structure is composed of human CD28 intracellular segment sequence in series Sequence composition of human CD3 ⁇ intracellular segment.
  • the ubiquitin-modified chimeric antigen receptor connects ubiquitin to CD3 ⁇ through a linker peptide in the intracellular domain.
  • the above scFv amino acid sequences were converted into base sequences after codon optimization, and synthesized by a third-party company (genscript).
  • the base sequences of all CARs in the present invention were finally cloned into the pHR-hEF1 ⁇ -IRES-EGFP vector (derived from addgene) by Gibson connection.
  • Example 2 Human primary T cell culture and lentivirus infection
  • Human primary T cells were obtained from healthy informed volunteers. Primary T cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin sulfate, 1 mM sodium pyruvate, non-essential amino acids, and 55 ⁇ M 2-mercaptoethanol (The above reagents were purchased from Gibco). In order to maintain the proliferation of T cells, 100 U/ml of hIL-2 (Sigma-Aldrich) was added to the culture medium.
  • hIL-2 Sigma-Aldrich
  • Preparation of lentivirus Resuspend Lenti-X 293T cells (TaKaRa#632180) in DMEM medium (Gibco#11995-065) containing 10% fetal bovine serum and without antibiotics, and make 6.5 ⁇ 10 5 cells/well The density was inoculated in 6-well cell culture plate (Corning#CLS3516) and cultured for 24 hours.
  • liposome transfection system (Mirus#2300), mix 500ng lentiviral packaging plasmid pCMVdR8.92 (Addgene#8455) and 50ng pMD2.G (Addgene#12259) with 500ng lentiviral plasmid to be packaged, according to liposome After mixing the operation steps of the transfection instructions, add Lenti-X 293T cells. Discard the liposome-containing medium after 16-18 hours, and add an appropriate amount of fresh medium; collect the cell supernatant after 48 hours, concentrate directly or by ultracentrifugation, and store in a -80°C refrigerator for later use.
  • Lentiviral infection of primary T cells Use magnetic beads (Life Technologies #11132D) coated with anti-human CD3 and anti-human CD28 antibodies to activate T cells, mix T cells with magnetic beads 1:3, add to the preparation after 24 hours of culture Infect with a good lentivirus; after 18 hours, the medium containing the virus solution was discarded and replaced with fresh T cell complete medium. After T cells were stimulated by magnetic beads for 4-5 days, the magnetic beads were removed, and the cell density was adjusted to 0.8-1*10 ⁇ 6/ml with T cell complete medium, and fresh T cell complete medium was supplemented every 2 days.
  • FACS buffer phosphate buffered saline PBS + 2% fetal bovine serum
  • FACS buffer phosphate buffered saline PBS + 2% fetal bovine serum
  • Flow cytometry data were acquired by a BD LSR Fortessa machine (BD bioscience) and analyzed using FlowJo software (Tree Star). Antibodies used in flow cytometry are listed below.
  • Example 4 In vitro killing function detection of CAR-T based on flow cytometry
  • K562 target cells expressing CD19 and mCherry fluorescence After mixing double-positive K562 target cells expressing CD19 and mCherry fluorescence with K562 non-target cells not expressing CD19 and mCherry fluorescence at a ratio of 1:1, they were mixed with CAR-T cells at a certain ratio of effector cells: target cells and co-incubated 24 hours. Cells were cultured in complete T cell medium without IL-2.
  • CD19-28WT-CAR and CD19-28Mono-CAR T cells were counted, they were mixed with irradiated target cells Nalm6 3:1, and the cells were resuspended in IL-2-free T cell complete medium to a cell density of 1 *10 ⁇ 6/ml; Count the viable cells every 2 days to calculate the amount of cell proliferation, and adjust the cell density to 1*10 ⁇ 6/ml with the complete medium of T cells without IL-2; when the cells obtained by the viable cell count When the density is less than or equal to 1*10 ⁇ 6/ml, it indicates that the cells are no longer proliferating and the experiment is terminated.
  • Example 6 Mouse tumor model and CAR-T cell function detection
  • NSG mice were first inoculated with 1 ⁇ 106 B lymphoma cells Nalm6 expressing the firefly luciferase gene in the tail vein; Treat NSG mice with 2 ⁇ 10 6 CAR-T cells by injection; detect the intensity of firefly luciferase carried by mouse tumor cells every week through a small animal in vivo imaging system to reflect the load of tumor cells, To track the development of tumors in the body.
  • the specific implementation of the small animal in vivo imaging system includes: giving the firefly luciferase substrate (D-luciferin sodium salt) to tumor-bearing mice by intraperitoneal injection, and the substrate dosage is 0.15 mg/g mouse body weight; 10 minutes later After the substrate was fully circulated to the whole body of the mouse, the mouse was anesthetized with 2.5%-3.5% isoflurane gas and then imaged. Bioluminescence imaging was performed with an IVIS Spectral Imaging System (Perkin Elmer), and fluorescence quantitative data were acquired with an in vivo imaging software (Perkin Elmer).
  • Example 7 Detection of CAR-T cell proliferation, differentiation and exhaustion in tumor-bearing mice
  • NSG mice 5- to 8-week-old combined immunodeficiency mice.
  • NSG mice were first inoculated with 2 ⁇ 106 B lymphoma cells Nalm6 in the tail vein; after the target cells grew in vivo for 4 days, they were injected through the tail vein.
  • 2 ⁇ 10 6 CAR-T cells were administered to NSG mice; 3 mice inoculated with CD19-28 WT and CD19-28 MonoUb CAR-T cells were sacrificed 7 days, 14 days, and 21 days later, and their spleens and hind limbs were collected.

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

Abstract

Provided are a chimeric antigen receptor modified by ubiquitin coupling and an immune cell. The ubiquitin is coupled to a C-terminus of the chimeric antigen receptor, and is suitable for a variety of current CAR-T cell production technologies, and particularly suitable for CAR-T therapeutic products developed against solid tumor targets.

Description

一种泛素偶联修饰的嵌合抗原受体及免疫细胞A ubiquitin-conjugated modified chimeric antigen receptor and immune cells 技术领域technical field
本发明涉及生物医药领域,特别是涉及一种泛素偶联修饰的嵌合抗原受体及免疫细胞。The invention relates to the field of biomedicine, in particular to a ubiquitin-coupled modified chimeric antigen receptor and immune cells.
背景技术Background technique
嵌合抗原受体(chimeric antigen receptor,CAR)是一种人工改造的靶向肿瘤的抗原受体。CAR-T疗法是通过在患者自身T细胞上表达特异性识别抗原和传递T细胞信号的CAR,使其能在患者体内识别并杀伤肿瘤细胞的一种过继性免疫疗法。CAR组成主要包括:能够识别并结合特异性肿瘤抗原的单链抗体(single chain antibody fragment,scFv),其特异性可以实现对肿瘤细胞的精确识别与“定点打击”;T细胞受体(T cell receptor,TCR)ζ链的胞内结构域,可以在scFv结构识别肿瘤抗原后,有效激活T细胞,而激活后的T细胞能够大量分泌细胞因子,在自身快速增殖的同时,有效杀伤肿瘤细胞,并通过分泌的炎症因子招募其他免疫细胞;在此基础上,在CAR胞内区域加入CD28或者41BB等共刺激分子的信号结构域,则可以增强CAR激活T细胞的能力,提升CAR-T细胞在体内存活的能力。Chimeric antigen receptor (CAR) is an artificially engineered tumor-targeting antigen receptor. CAR-T therapy is an adoptive immunotherapy that expresses CARs that specifically recognize antigens and transmit T cell signals on the patient's own T cells, enabling them to recognize and kill tumor cells in the patient's body. The composition of CAR mainly includes: a single chain antibody (single chain antibody fragment, scFv) that can recognize and bind to a specific tumor antigen, and its specificity can realize precise recognition and "targeted attack" on tumor cells; T cell receptor (T cell The intracellular domain of the receptor (TCR) ζ chain can effectively activate T cells after the scFv structure recognizes tumor antigens, and the activated T cells can secrete a large amount of cytokines, and effectively kill tumor cells while rapidly proliferating themselves. And recruit other immune cells through secreted inflammatory factors; on this basis, adding the signaling domain of co-stimulatory molecules such as CD28 or 41BB to the intracellular region of CAR can enhance the ability of CAR to activate T cells and improve the ability of CAR-T cells to activate T cells. ability to survive in vivo.
目前,CAR-T疗法已经在多种类型的肿瘤,尤其是针对血液类肿瘤的临床治疗中,取得了显著的成功。2017年,美国FDA先后批准上市了两款靶向CD19抗原的商业化CAR-T治疗产品,用于治疗复发难治性恶性B细胞白血病及淋巴瘤,并取得了显著的疗效。然而,CAR-T疗法在应对实体瘤的治疗中仍然存在诸多局限性因素。2019年Robbie等人总结了当前CAR-T疗法在临床应用过程中遇到的问题,并指出其中限制CAR-T细胞在体内发挥功能的原因之一是由于肿瘤微环境、肿瘤靶标负担过大、T细胞本底自激活水平过高等因素导致的T细胞功能耗竭,从而无法有效杀伤肿瘤。大量研究表明,降低CAR对于T细胞自激活的影响,能够有效地缓解CAR-T细胞在培养增殖过程中由于过度分化、提前耗竭等原因导致的CAR-T细胞功能低下的问题,进而能够有效地提升CAR-T治疗的效果,这为优化CAR-T的设计提供了方向。At present, CAR-T therapy has achieved remarkable success in the clinical treatment of various types of tumors, especially for blood tumors. In 2017, the U.S. FDA successively approved the marketing of two commercial CAR-T therapeutic products targeting CD19 antigen for the treatment of relapsed and refractory malignant B-cell leukemia and lymphoma, and achieved remarkable curative effect. However, CAR-T therapy still has many limitations in the treatment of solid tumors. In 2019, Robbie et al. summarized the problems encountered in the clinical application of current CAR-T therapy, and pointed out that one of the reasons for limiting the function of CAR-T cells in vivo is due to the tumor microenvironment, excessive burden of tumor targets, The exhaustion of T cell function caused by factors such as excessive self-activation level of T cell background can not effectively kill tumors. A large number of studies have shown that reducing the influence of CAR on the self-activation of T cells can effectively alleviate the problem of low function of CAR-T cells caused by excessive differentiation and premature exhaustion in the process of culture and proliferation of CAR-T cells, and then can effectively Improve the effect of CAR-T therapy, which provides a direction for optimizing the design of CAR-T.
目前,已有的关于CAR-T的优化设计策略主要包括:通过突变失活CD3ζ中不同ITAM区组合,降低CAR的信号;突变CD28共刺激结构域,降低CD28的共刺激信号;尝试不同结构组合,探索针对不同靶点的最适合、本底自激活水平最低的CAR结构形式;将参与调控T细胞增殖和分化的转录因子与CAR共同转导入T细胞中,以降低CAR-T细胞的自激活信号、遏制T细胞的耗竭等。然而,这类设计往往需要对针对不同靶点的CAR进行尝试,耗费大量人力物力,且有误差大、普适性低等缺点。因此,针对CAR结构本身的、更具实用性 与普适性的改造是更优化的方案。At present, the existing optimal design strategies for CAR-T mainly include: reducing the signal of CAR by mutating and inactivating different combinations of ITAM regions in CD3ζ; mutating the co-stimulatory domain of CD28 to reduce the co-stimulatory signal of CD28; trying different structural combinations , explore the most suitable CAR structure for different targets and the lowest level of background self-activation; co-transduce transcription factors involved in the regulation of T cell proliferation and differentiation into T cells with CAR to reduce the self-activation of CAR-T cells Signaling, suppression of exhaustion of T cells, etc. However, this type of design often needs to try CARs targeting different targets, which consumes a lot of manpower and material resources, and has the disadvantages of large errors and low universality. Therefore, a more practical and universal transformation aimed at the CAR structure itself is a more optimal solution.
发明内容Contents of the invention
鉴于以上所述现有技术的缺点,本发明的目的在于提供一种泛素偶联修饰的嵌合抗原受体及免疫细胞,用于解决现有技术中的问题。In view of the above-mentioned shortcomings of the prior art, the purpose of the present invention is to provide a ubiquitin-modified chimeric antigen receptor and immune cells to solve the problems in the prior art.
为实现上述目的及其他相关目的,本发明首先提供一种嵌合抗原受体的改造方法,所述改造方法包括将泛素偶联至嵌合抗原受体的C端。To achieve the above purpose and other related purposes, the present invention firstly provides a chimeric antigen receptor modification method, which includes coupling ubiquitin to the C-terminus of the chimeric antigen receptor.
本发明提供一种泛素修饰的嵌合抗原受体,所述泛素修饰的嵌合抗原受体包括跨膜域、胞内域和胞外域,所述胞内域C端偶联有泛素。The present invention provides a ubiquitin-modified chimeric antigen receptor, the ubiquitin-modified chimeric antigen receptor comprises a transmembrane domain, an intracellular domain and an extracellular domain, and the C-terminus of the intracellular domain is coupled with ubiquitin .
本发明提供一种分离的多核苷酸,所述分离的多核苷酸包括编码所述泛素修饰的嵌合抗原受体的核苷酸。The present invention provides an isolated polynucleotide comprising nucleotides encoding the ubiquitin-modified chimeric antigen receptor.
本发明提供一种核酸构建体,所述核酸构建体中包括所述分离的多核苷酸。The present invention provides a nucleic acid construct comprising the isolated polynucleotide.
本发明提供一种免疫细胞,所述免疫细胞中包括所述核酸构建体或基因组中整合有外源的所述的多核苷酸,或能够表达所述泛素修饰的嵌合抗原受体。The present invention provides an immune cell, which includes the nucleic acid construct or the exogenous polynucleotide integrated in the genome, or can express the ubiquitin-modified chimeric antigen receptor.
所述免疫细胞选自CAR-T细胞、CAR-NK细胞、CAR-巨噬细胞或CAR-TIL细胞。The immune cells are selected from CAR-T cells, CAR-NK cells, CAR-macrophages or CAR-TIL cells.
本发明还提供所述免疫细胞的制备方法,所述制备方法包括将所述核酸构建体或所述多核苷酸导入免疫效应细胞或产生免疫效应细胞的干细胞中。The present invention also provides a preparation method of the immune cells, the preparation method comprising introducing the nucleic acid construct or the polynucleotide into immune effector cells or stem cells producing immune effector cells.
所述免疫效应细胞选自T细胞、NK细胞、巨噬细胞或TIL细胞。The immune effector cells are selected from T cells, NK cells, macrophages or TIL cells.
本发明还提供所述免疫细胞在制备治疗癌症或免疫性疾病药物中的用途。The present invention also provides the use of the immune cells in the preparation of medicines for treating cancer or immune diseases.
本发明还提供一种肿瘤或免疫性疾病的治疗方法,所述治疗方法包括向受试者施用治疗有效量的所述免疫细胞。The present invention also provides a treatment method for tumors or immune diseases, the treatment method comprising administering a therapeutically effective amount of the immune cells to a subject.
如上所述,本发明的泛素偶联修饰的嵌合抗原受体及免疫细胞,具有以下有益效果:为CAR-T的优化改造提供方法,尤其是针对CAR-T在实体瘤中增殖能力不佳的问题提供解决思路。本发明提供的简单易行且适用于不同靶点的高自激活水平CAR的优化改造方案,具有应用简单、可重复性高、普适性好和优化程度高、抗肿瘤活性好等优势,适用于各种现行的CAR-T细胞生产技术,尤其适合于针对实体瘤靶点开发的CAR-T治疗产品,为CAR-T细胞本底自激活水平过高、增殖能力不佳、分化程度过高、耗竭过快等问题提供了一种新的解决思路。As mentioned above, the ubiquitin-conjugated modified chimeric antigen receptor and immune cells of the present invention have the following beneficial effects: provide a method for the optimization and transformation of CAR-T, especially for CAR-T that is not capable of proliferating in solid tumors Provides solutions to the best problems. The optimization and transformation scheme of CAR with high self-activation level, which is simple and applicable to different targets, has the advantages of simple application, high repeatability, good universality, high degree of optimization, and good anti-tumor activity. Suitable for all kinds of current CAR-T cell production technologies, especially suitable for CAR-T therapeutic products developed for solid tumor targets. , Excessive exhaustion and other problems provide a new solution.
附图说明Description of drawings
图1显示为在人类T淋巴瘤细胞系Jurkat中,检测构建的针对不同靶点的CAR-T细胞系的自激活水平,利用测量T细胞激活水平的指征蛋白,如CD69、ICOS等的表达情况,评估这些细胞系的本底自激活水平,左图为检测的CD69表达水平,右图为检测的ICOS表达水平。如图所示,大量临床和临床前试验中采用的、针对不同靶标的CAR都会导致T细胞较高的本底自激活水平。尤其是针对实体肿瘤的GD2、Her2、CSPG4、EGFR、Meso、GPC3、多个靶点的CAR(核苷酸序列依次如SEQ ID NO.3-11,氨基酸依次如SEQ ID NO.19-27),其本底自激活水平比较高。前人的研究证明CAR-T细胞自激活水平过高会导致其过早分化、增殖能力下降、细胞耗竭水平提高、抗肿瘤杀伤能力大幅下降等结果,与其临床治疗效果不佳呈正相关。Figure 1 shows the detection of the self-activation level of CAR-T cell lines targeting different targets in the human T lymphoma cell line Jurkat, using the expression of indicator proteins such as CD69 and ICOS to measure the activation level of T cells In this case, the background self-activation level of these cell lines was evaluated. The left panel shows the detected expression level of CD69, and the right panel shows the detected ICOS expression level. As shown in the figure, CARs targeting different targets used in a large number of clinical and preclinical trials lead to high levels of background self-activation of T cells. Especially CARs targeting GD2, Her2, CSPG4, EGFR, Meso, GPC3, and multiple targets of solid tumors (the nucleotide sequence is as shown in SEQ ID NO.3-11, and the amino acid sequence is as in SEQ ID NO.19-27) , and its background self-activation level is relatively high. Previous studies have proved that excessive self-activation of CAR-T cells will lead to premature differentiation, decreased proliferation ability, increased cell exhaustion level, and greatly reduced anti-tumor killing ability, etc., which is positively correlated with poor clinical treatment effect.
图2-1至图2-3显示为将CD19-28WT-CAR(核苷酸序列如SEQ ID NO.12,氨基酸序列如SEQ ID NO.28)以及在靶向CD19的CAR C端偶联一个野生型泛素(wtUb)(核苷酸序列如SEQ ID NO.13,氨基酸序列如SEQ ID NO.29)或者单泛素(monoUb)(核苷酸序列如SEQ ID NO.14,氨基酸序列如SEQ ID NO.30)得到的CD19-28wtUb-CAR与CD19-28MonoUb-CAR分别表达于Jurkat T细胞系(图2-1)或原代人T细胞(图2-2)中,检测它们和野生型CAR在细胞膜上表达水平的变化,以及该修饰对CAR-T细胞肿瘤杀伤能力的影响。检测结果显示,wtUb以及monoUb的偶联都能够有效的下调细胞膜表面CAR的表达水平,且wtUb CAR的下调幅度更为显著。体外杀伤试验是将CAR-T细胞与CD19+靶细胞按照一定比例共孵育,18小时后通过检测靶细胞的量来反应CAR-T细胞的肿瘤杀伤能力(图2-3)。检测结果显示,monoUb修饰的CAR仍然能够有效的介导CAR-T细胞对肿瘤靶细胞的杀伤作用,而wtUb修饰的CAR或因其表达水平过低,导致其肿瘤杀伤能力显著下降。因而,后续的研究和优化均集中于偶联单泛素的CAR。Figure 2-1 to Figure 2-3 show that CD19-28WT-CAR (nucleotide sequence such as SEQ ID NO.12, amino acid sequence such as SEQ ID NO.28) and a CD19-targeting CAR C-terminal coupling Wild-type ubiquitin (wtUb) (nucleotide sequence such as SEQ ID NO.13, amino acid sequence such as SEQ ID NO.29) or monoubiquitin (monoUb) (nucleotide sequence such as SEQ ID NO.14, amino acid sequence such as The CD19-28wtUb-CAR and CD19-28MonoUb-CAR obtained from SEQ ID NO.30) were expressed in Jurkat T cell line (Fig. 2-1) or primary human T cells (Fig. 2-2), respectively. Changes in the expression level of CAR-type on the cell membrane, and the impact of this modification on the tumor-killing ability of CAR-T cells. The test results showed that the coupling of wtUb and monoUb can effectively down-regulate the expression level of CAR on the cell membrane surface, and the down-regulation of wtUb CAR is more significant. In the in vitro killing test, CAR-T cells were co-incubated with CD19+ target cells at a certain ratio, and the tumor killing ability of CAR-T cells was reflected by detecting the amount of target cells after 18 hours (Figure 2-3). The test results showed that the monoUb-modified CAR can still effectively mediate the killing effect of CAR-T cells on tumor target cells, while the wtUb-modified CAR may have a significantly reduced tumor-killing ability due to its low expression level. Therefore, subsequent research and optimization focused on CARs coupled with monoubiquitin.
图3-1至图3-4显示为将多种针对不同靶标的偶联单泛素的CD28 CAR表达于Jurkat细胞系后,检测该修饰对于CAR-T本底自激活水平的影响,验证本方案的普适性。检测了针对以CD19为靶标(图3-1),以GD2(核苷酸序列如SEQ ID NO.15,氨基酸序列如SEQ ID NO.31,结果如图3-2)为靶标,以GPC3(核苷酸序列如SEQ ID NO.16,氨基酸序列如SEQ ID NO.32,结果如图3-3)为靶标,以及以CD19和CD22为双靶标(核苷酸序列如SEQ ID NO.33,氨基酸序列如SEQ ID NO.34,结果如图3-4)的偶联单泛素的CAR在细胞膜上的表达水平,以及所对应CAR-T细胞的本底自激活信号(以CD19和CD22为双靶标的WT-CAR的核苷酸序列如SEQ ID NO.35,氨基酸序列如SEQ ID NO.36)。结果如图所示,4种CAR的表达水平均明显下降;以T细胞激活指征性蛋白CD69,ICOS表达水平反应CAR-T细胞的本底自激 活信号,则4种CAR-T细胞的本底自激活水平均有显著的改善。表明本方案具有普适性,对于不同靶标的、高自激活水平的CAR,均能有效的下调它们的自激活信号。Figure 3-1 to Figure 3-4 show that after expressing a variety of monoubiquitin-coupled CD28 CARs targeting different targets in the Jurkat cell line, the effect of the modification on the background self-activation level of CAR-T was tested to verify this Universality of the program. Detection of CD19 as the target (Figure 3-1), GD2 (nucleotide sequence such as SEQ ID NO.15, amino acid sequence such as SEQ ID NO.31, the results as shown in Figure 3-2) as the target, GPC3 ( Nucleotide sequence such as SEQ ID NO.16, amino acid sequence such as SEQ ID NO.32, the results are shown in Figure 3-3) as the target, and CD19 and CD22 as the dual target (nucleotide sequence such as SEQ ID NO.33, Amino acid sequence such as SEQ ID NO.34, results shown in Figure 3-4) the expression level of monoubiquitin-coupled CAR on the cell membrane, and the background self-activation signal of the corresponding CAR-T cells (with CD19 and CD22 as the The nucleotide sequence of the dual-target WT-CAR is shown in SEQ ID NO.35, and the amino acid sequence is shown in SEQ ID NO.36). The results are shown in the figure, the expression levels of the four CAR-T cells were significantly decreased; the expression levels of CD69 and ICOS, the indicator proteins for T cell activation, reflected the background self-activation signal of CAR-T cells, and the expression levels of the four CAR-T cells Significant improvement was seen in the level of self-activation at the bottom. It shows that this scheme is universal, and it can effectively down-regulate the self-activation signals of CARs with different targets and high levels of self-activation.
图4-1至图4-4显示为在体外培养条件下,检测多种靶标的野生型(即CD19-28WT-CAR T细胞、GD2-28WT-CAR T细胞、GPC3-28WT-CAR T细胞、CD19×22WT-CAR T细胞)以及偶联单泛素的CAR-T细胞(即CD19-28MonoUb-CAR T细胞、GD2-28MonoUb-CAR T细胞、GPC3-28MonoUb-CAR T细胞、CD19×22MonoUb-CAR T细胞)的分化情况。通过流式检测细胞表达CD45RA与CD62L的水平,可以将T细胞的分化状况分为CD45RA+CD62L+(Stem cell memory T cell,T SCM),CD45RA-CD62L+(Central memory T cell,T CM),CD45RA-CD62L-(Effector memory T cell,T EM),以及CD45RA+CD62L-(Effector T cell,T EFF),其分化水平依次递增,增殖和分化能力依次递减。如图所示,在针对4种不同靶标的CAR-T细胞中(图4-1为靶向CD19的CAR-T细胞,图4-2为靶向GD2的CAR-T细胞,图4-3为靶向GPC3的CAR-T细胞,图4-4为靶向CD19和CD22双靶点的CAR-T细胞,各图中左图为检测的CD4,右图为检测的CD8),偶联单泛素的修饰均可以有效的减缓T细胞的分化,使得更多的CAR-T细胞保持在的T SCM状态,这也暗示了偶联单泛素的CAR-T细胞应当会具有更好的持续增殖能力和肿瘤杀伤能力。 Figure 4-1 to Figure 4-4 show the detection of wild-type targets (ie CD19-28WT-CAR T cells, GD2-28WT-CAR T cells, GPC3-28WT-CAR T cells, GPC3-28WT-CAR T cells, CD19×22WT-CAR T cells) and CAR-T cells coupled with monoubiquitin (i.e. CD19-28MonoUb-CAR T cells, GD2-28MonoUb-CAR T cells, GPC3-28MonoUb-CAR T cells, CD19×22MonoUb-CAR T cells, CD19×22MonoUb-CAR T cells differentiation of T cells). By flow cytometric detection of the levels of CD45RA and CD62L expressed by cells, the differentiation status of T cells can be divided into CD45RA+CD62L+ (Stem cell memory T cell, T SCM ), CD45RA-CD62L+ (Central memory T cell, T CM ), CD45RA- CD62L- (Effector memory T cell, T EM ), and CD45RA+CD62L- (Effector T cell, T EFF ), the differentiation level increases successively, and the proliferation and differentiation ability decrease successively. As shown in the figure, among CAR-T cells targeting 4 different targets (Figure 4-1 is CAR-T cells targeting CD19, Figure 4-2 is CAR-T cells targeting GD2, Figure 4-3 CAR-T cells targeting GPC3, Figure 4-4 shows CAR-T cells targeting both CD19 and CD22, the left figure in each figure is the detected CD4, and the right figure is the detected CD8), coupled with single The modification of ubiquitin can effectively slow down the differentiation of T cells, so that more CAR-T cells remain in the TSCM state, which also implies that CAR-T cells coupled with monoubiquitin should have better persistence. proliferation and tumor killing ability.
图5显示为体外检测靶向GD2的野生型以及偶联单泛素的CAR-T细胞在靶细胞持续刺激下的增殖情况。首先,给予等量GD2-28WT-CAR以及GD2-28MonoUb-CAR T细胞等量的靶细胞(经辐照处理)刺激。之后,每2天进行活细胞计数,记录细胞增殖情况。同时,补充适量培养基、调整细胞密度至1*10^6/ml继续培养。该实验结果表明,在靶细胞存在且持续刺激的情况下,偶联单泛素的CAR-T细胞具有更低的本底自激活水平和细胞分化程度,且具有更好的持续增殖能力。Figure 5 shows the in vitro detection of the proliferation of GD2-targeting wild-type and monoubiquitin-coupled CAR-T cells under continuous stimulation of target cells. First, the same amount of GD2-28WT-CAR and GD2-28MonoUb-CAR T cells were given the same amount of target cells (irradiated) to stimulate. Afterwards, live cell counts were performed every 2 days to record cell proliferation. At the same time, add an appropriate amount of medium and adjust the cell density to 1*10^6/ml to continue the culture. The results of this experiment showed that in the presence and continuous stimulation of target cells, monoubiquitin-coupled CAR-T cells had lower background self-activation levels and cell differentiation degrees, and had better sustained proliferation capabilities.
图6-1至图6-2显示为荷瘤小鼠体内肿瘤杀伤实验,验证了靶向CD19的野生型以及偶联单泛素的CAR-T细胞之间的功能性差异。将荷瘤小鼠随机分为3组,分别通过尾静脉注射的方式给予小鼠等量未转染CAR的正常T细胞、CD19 WT CAR-T细胞以及CD19 MonoUb CAR-T细胞,通过小动物活体成像设备(in vivo imaging system,IVIS)对小鼠体内的肿瘤细胞生长情况进行跟踪监测。在回输较少CAR-T细胞的情况下,野生型CAR-T细胞对肿瘤的控制能力有限,而偶联单泛素的CAR-T细胞仍然能够在一定程度上有效控制肿瘤的生长,在个别小鼠中能够实现肿瘤的清除(图6-1),有效提升了荷瘤小鼠的生存率(图6-2),其功效性显著优于野生型CAR-T细胞。Figure 6-1 to Figure 6-2 show the in vivo tumor killing experiments in tumor-bearing mice, verifying the functional differences between wild-type and monoubiquitin-coupled CAR-T cells targeting CD19. The tumor-bearing mice were randomly divided into 3 groups, and the same amount of normal T cells not transfected with CAR, CD19 WT CAR-T cells and CD19 MonoUb CAR-T cells were given to the mice by tail vein injection respectively. Imaging equipment (in vivo imaging system, IVIS) tracked and monitored the growth of tumor cells in mice. In the case of reinfusion of fewer CAR-T cells, wild-type CAR-T cells have limited ability to control tumors, while CAR-T cells coupled with monoubiquitin can still effectively control tumor growth to a certain extent. In individual mice, tumor clearance can be achieved (Figure 6-1), which effectively improves the survival rate of tumor-bearing mice (Figure 6-2), and its efficacy is significantly better than that of wild-type CAR-T cells.
图7-1至图7-2显示为对于荷瘤小鼠体内CAR-T细胞增殖、分化以及耗竭情况的检测。 结果显示,在体内肿瘤细胞等的刺激下,偶联单泛素的CAR-T细胞仍然存在更高比例的、具有较高持续增殖能力的干细胞记忆T细胞(T SCM)(图7-1,左图为骨髓内CD4CAR-T细胞,中图为骨髓内CD8 CAR-T细胞);且与之相应的,偶联单泛素的CAR-T细胞数量也显著多于野生型CAR-T细胞(图7-1,右图为脾内CAR-T细胞计数),其细胞耗竭的指征性蛋白(PD1,LAG3,TIM3,即图7-2,左图为骨髓内CD4CAR-T细胞,右图为骨髓内CD8 CAR-T细胞)表达水平也明显较低,这可能与偶联单泛素的CAR在靶细胞刺激条件下的激活信号较弱有关,而较弱的激活信号足以使T细胞发挥正常的肿瘤杀伤功能;这也暗示了野生型CAR过强的激活信号可能导致T细胞快速的分化和功能的提前耗竭,进而限制CAR-T细胞的抗肿瘤功效。 Figure 7-1 to Figure 7-2 show the detection of CAR-T cell proliferation, differentiation and exhaustion in tumor-bearing mice. The results showed that under the stimulation of tumor cells in vivo, CAR-T cells coupled with monoubiquitin still had a higher proportion of stem cell memory T cells (T SCM ) with higher sustained proliferation ability (Figure 7-1, The left picture shows CD4 CAR-T cells in the bone marrow, and the middle picture shows CD8 CAR-T cells in the bone marrow); and correspondingly, the number of CAR-T cells coupled with monoubiquitin is also significantly more than that of wild-type CAR-T cells ( Figure 7-1, the right figure is the count of CAR-T cells in the spleen), and the indicative proteins of its cell exhaustion (PD1, LAG3, TIM3, that is, Figure 7-2, the left figure is the CD4 CAR-T cells in the bone marrow, and the right figure The expression level of CD8 CAR-T cells in bone marrow is also significantly lower, which may be related to the weaker activation signal of CAR coupled with monoubiquitin under the condition of target cell stimulation, and the weaker activation signal is enough to make T cells exert Normal tumor killing function; this also implies that the excessive activation signal of wild-type CAR may lead to rapid differentiation and premature exhaustion of T cells, thereby limiting the anti-tumor efficacy of CAR-T cells.
具体实施方式Detailed ways
本发明提供一种简单易行且适用于不同靶点的高自激活水平CAR的优化改造方案。具体的为将一个泛素蛋白突变体(简称单泛素,MonoUb)偶联在针对不同靶点的CAR的C端得到MonoUb-CAR。所以本发明首先提供一种嵌合抗原受体的改造方法,所述改造方法包括将泛素偶联至嵌合抗原受体的C端。The present invention provides an optimized transformation scheme of CAR with high self-activation level that is simple and applicable to different targets. Specifically, a ubiquitin protein mutant (monoubiquitin, MonoUb for short) is coupled to the C-terminus of CARs targeting different targets to obtain MonoUb-CAR. Therefore, the present invention firstly provides a chimeric antigen receptor modification method, which includes coupling ubiquitin to the C-terminus of the chimeric antigen receptor.
泛素(ubiquitin)是一种存在于所有真核生物(大部分真核细胞)中的小蛋白。泛素由76个氨基酸组成,分子量大约8.451kDa。它的主要功能是标记需要分解掉的蛋白质,使其被26S蛋白酶体降解。对于膜蛋白而言,特别是一些特殊的泛素修饰类型,可能参与了膜蛋白的下调、转运和膜泡分选,参与了其信号传递的调节。本申请中所指的泛素指来自任何真核细胞来源,包括哺乳动物诸如灵长类(例如人)和啮齿类动物(例如小鼠和大鼠)的任何野生型泛素或突变型泛素。泛素是一个非常保守的蛋白,从酵母开始几乎所有真核细胞均表达泛素蛋白,且其氨基酸序列基本完全相同(酵母和人的泛素只有一个氨基酸不同)。Ubiquitin is a small protein present in all eukaryotes (most eukaryotic cells). Ubiquitin consists of 76 amino acids with a molecular weight of about 8.451kDa. Its main function is to mark proteins that need to be broken down so that they can be degraded by the 26S proteasome. For membrane proteins, especially some special ubiquitin modification types may be involved in the down-regulation, transport and vesicle sorting of membrane proteins, as well as the regulation of their signal transmission. Ubiquitin as referred to in this application refers to any wild-type ubiquitin or mutant ubiquitin from any eukaryotic cellular source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats) . Ubiquitin is a very conserved protein. Almost all eukaryotic cells express ubiquitin since yeast, and its amino acid sequence is basically identical (yeast and human ubiquitin have only one amino acid difference).
所述泛素的类型为野生型泛素或突变型泛素。野生型泛素可以被胞内内源性泛素修饰而不可作为底物修饰于其他蛋白,故可形成泛素链。在一较佳的实施方式中,所述泛素为突变型泛素。因突变型泛素不能被内源性泛素修饰,且该突变型泛素不可作为底物对其他蛋白进行泛素化修饰,故突变型泛素为单泛素,所以突变型泛素也可称为突变型单泛素或单泛素。The type of ubiquitin is wild-type ubiquitin or mutant ubiquitin. Wild-type ubiquitin can be modified by intracellular endogenous ubiquitin but cannot be used as a substrate to modify other proteins, so it can form ubiquitin chains. In a preferred embodiment, the ubiquitin is mutant ubiquitin. Because the mutant ubiquitin cannot be modified by endogenous ubiquitin, and the mutant ubiquitin cannot be used as a substrate to ubiquitinate other proteins, the mutant ubiquitin is monoubiquitin, so the mutant ubiquitin can also be Known as mutant monoubiquitin or monoubiquitin.
所述泛素通过连接肽偶联至嵌合抗原受体的C端。本申请中所述的连接肽可以为本领域常用的连接肽。所述连接肽例如为GSGGSG、GSGGSGG GSGGSGGG或GGGGSGGG或GGSGGGSGGGSAAA。The ubiquitin is coupled to the C-terminus of the chimeric antigen receptor through a linker peptide. The connecting peptides described in the present application may be commonly used connecting peptides in the art. The connecting peptide is, for example, GSGGSG, GSGGSGG GSGGSGGG or GGGGSGGG or GGSGGGSGGGSAAA.
本发明还提供一种泛素修饰的嵌合抗原受体,所述泛素修饰的嵌合抗原受体包括跨膜域、胞内域和胞外域,所述胞内域C端偶联有泛素。The present invention also provides a ubiquitin-modified chimeric antigen receptor, which includes a transmembrane domain, an intracellular domain, and an extracellular domain, and the C-terminus of the intracellular domain is coupled with ubiquitin white.
所述泛素修饰的嵌合抗原受体通过所述改造方法获得。The ubiquitin-modified chimeric antigen receptor is obtained by the engineering method.
在本发明某些实施方式中,所述跨膜域可以包括CD8α、CD28、DAP 10等蛋白分子的跨膜结构域。例如,所述CD8α的氨基酸序列可以包括如下所示序列:IYIWAPLAGTCGVLLLSLVITLYC。再例如,CD8α的序列可参照NM_001145873,CD28的序列可参照NM_006139。In some embodiments of the present invention, the transmembrane domain may include transmembrane domains of protein molecules such as CD8α, CD28, and DAP10. For example, the amino acid sequence of CD8α may include the following sequence: IYIWAPLAGTCGVLLLSLVITLYC. For another example, refer to NM_001145873 for the sequence of CD8α, and refer to NM_006139 for the sequence of CD28.
在本发明某些实施方式中,所述胞内域可以包括共刺激结构域和/或信号结构域。例如,所述胞内域可以包括以下中的一种或几种的组合:4-1BB、CD28、OX40、ICOS、CD3ζ、DAP 10等蛋白分子的信号转导结构域。In certain embodiments of the invention, the intracellular domain may comprise a co-stimulatory domain and/or a signaling domain. For example, the intracellular domain may include one or a combination of several of the following: signal transduction domains of protein molecules such as 4-1BB, CD28, OX40, ICOS, CD3ζ, and DAP10.
再例如,所述4-1BB的氨基酸序列包括如下所示:For another example, the amino acid sequence of 4-1BB includes as follows:
Figure PCTCN2022080608-appb-000001
Figure PCTCN2022080608-appb-000001
所述CD3ζ的氨基酸序列包括如下所示:The amino acid sequence of the CD3ζ includes as follows:
Figure PCTCN2022080608-appb-000002
Figure PCTCN2022080608-appb-000002
再例如,4-1BB的序列可参照NM_001561,CD28的序列可参照NM_006139,OX40的序列可参照NM_003327,ICOS的序列可参照NM_012092,CD3ζ的序列可参照NM_198053、DAP 10的序列可参照NM_014266。在本发明一具体实施方式中,所述胞内域自N端至C端依次包括CD28和CD3ζ。在本发明某些实施方式中,所述嵌合抗原受体自N端至C端依次包括胞外域、跨膜域、胞内域。For another example, refer to NM_001561 for the sequence of 4-1BB, refer to NM_006139 for the sequence of CD28, refer to NM_003327 for the sequence of OX40, refer to NM_012092 for the sequence of ICOS, refer to NM_198053 for the sequence of CD3ζ, and refer to NM_014266 for the sequence of DAP 10. In a specific embodiment of the present invention, the intracellular domain sequentially includes CD28 and CD3ζ from the N-terminus to the C-terminus. In some embodiments of the present invention, the chimeric antigen receptor includes an extracellular domain, a transmembrane domain, and an intracellular domain sequentially from the N-terminus to the C-terminus.
在本发明某些实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、跨膜域、胞内域。在本发明一些具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD8α跨膜区、4-1BB共刺激结构域、CD3ζ信号结构域。在本发明一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD28跨膜区、CD28共刺激结构域、CD3ζ信号结构域。在本发明另一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD8α跨膜区、OX40共刺激结构域、CD3ζ信号结构域。在本发明另一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD8α跨膜区、ICOS共刺激结构域、CD3ζ信号结构域。在本发明另一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD8α跨膜区、4-1BB共刺激结构域、CD3ζ信号结构域。在本发明另一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括单链抗体、CD28跨膜区、CD28共刺激结构域、OX40共刺激结构域、CD3ζ信号结构域。在本发明另一具体实施方式中,所述嵌合抗原受体自N端至C端依次包括CD8α信号肽、myc标签、scFv、CD8α铰链组成的胞外域,CD8α的跨膜域,CD28和CD3ζ串联组成的胞内域。所述泛素修饰的嵌合抗原受体即上述任一种嵌合抗原受体的CD3ζ后通过连接肽(或称linker)连接有泛素。In some embodiments of the present invention, the chimeric antigen receptor includes a single-chain antibody, a transmembrane domain, and an intracellular domain in sequence from the N-terminus to the C-terminus. In some specific embodiments of the present invention, the chimeric antigen receptor includes a single-chain antibody, a CD8α transmembrane region, a 4-1BB co-stimulatory domain, and a CD3ζ signaling domain sequentially from the N-terminus to the C-terminus. In a specific embodiment of the present invention, the chimeric antigen receptor comprises a single-chain antibody, a CD28 transmembrane region, a CD28 co-stimulatory domain, and a CD3ζ signaling domain sequentially from the N-terminus to the C-terminus. In another specific embodiment of the present invention, the chimeric antigen receptor includes a single-chain antibody, a CD8α transmembrane region, an OX40 co-stimulatory domain, and a CD3ζ signaling domain sequentially from the N-terminal to the C-terminal. In another specific embodiment of the present invention, the chimeric antigen receptor comprises a single-chain antibody, a CD8α transmembrane region, an ICOS co-stimulatory domain, and a CD3ζ signaling domain sequentially from the N-terminal to the C-terminal. In another specific embodiment of the present invention, the chimeric antigen receptor comprises a single-chain antibody, a CD8α transmembrane region, a 4-1BB co-stimulatory domain, and a CD3ζ signaling domain sequentially from the N-terminus to the C-terminus. In another specific embodiment of the present invention, the chimeric antigen receptor comprises a single-chain antibody, a CD28 transmembrane region, a CD28 co-stimulatory domain, an OX40 co-stimulatory domain, and a CD3ζ signaling domain from the N-terminus to the C-terminus. . In another specific embodiment of the present invention, the chimeric antigen receptor includes CD8α signal peptide, myc tag, scFv, extracellular domain composed of CD8α hinge, transmembrane domain of CD8α, CD28 and CD3ζ in sequence from N-terminal to C-terminal Intracellular domains organized in tandem. The ubiquitin-modified chimeric antigen receptor, that is, the CD3ζ of any one of the above chimeric antigen receptors is connected with ubiquitin through a linker peptide (or linker).
本发明所述单链抗体不做具体限制,可以选自任何单链抗体。本发明中作为举例使用的单链抗体例如有CD19 scFv、GD2 scFv、GPC3 scFv、Her2 scFv、CSPG4 scFv、EGFR scFv、Meso scFv、TRBC1 scFv、CD133 scFv、BCMA scFv中的任一个或多个。当使用多个例如两个单链抗体时,所述嵌合抗原受体为多靶点嵌合抗原受体。The single-chain antibody of the present invention is not specifically limited, and can be selected from any single-chain antibody. Examples of single-chain antibodies used in the present invention include any one or more of CD19 scFv, GD2 scFv, GPC3 scFv, Her2 scFv, CSPG4 scFv, EGFR scFv, Meso scFv, TRBC1 scFv, CD133 scFv, and BCMA scFv. When multiple, eg, two, single chain antibodies are used, the chimeric antigen receptor is a multi-target chimeric antigen receptor.
本申请中所指的泛素选自野生型泛素或突变型泛素。The ubiquitin referred to in this application is selected from wild-type ubiquitin or mutant ubiquitin.
野生型泛素的氨基酸序列如SEQ ID NO.17所示。The amino acid sequence of wild-type ubiquitin is shown in SEQ ID NO.17.
在一较佳实施方式中,所述泛素选自突变型泛素。本发明中选用的突变型泛素的氨基酸序列如SEQ ID NO.18所示。In a preferred embodiment, the ubiquitin is selected from mutant ubiquitin. The amino acid sequence of the mutant ubiquitin selected in the present invention is shown in SEQ ID NO.18.
本发明提供一种分离的多核苷酸,所述分离的多核苷酸包括编码所述泛素修饰的嵌合抗原受体的核苷酸。The present invention provides an isolated polynucleotide comprising nucleotides encoding the ubiquitin-modified chimeric antigen receptor.
编码所述跨膜域、胞内域和胞外域的多核苷酸均可选自现有技术中的多核苷酸。The polynucleotides encoding the transmembrane domain, intracellular domain and extracellular domain can all be selected from polynucleotides in the prior art.
在本发明的一具体实施方式中,编码所述泛素修饰的嵌合抗原受体的多核苷酸中依次包括编码CD8α信号肽、myc标签、scFv、CD8α铰链组成的胞外域的核苷酸,编码CD8α跨膜域的核苷酸,编码CD28、CD3ζ、泛素串联组成的胞内域的核苷酸。In a specific embodiment of the present invention, the polynucleotide encoding the ubiquitin-modified chimeric antigen receptor sequentially includes nucleotides encoding the CD8α signal peptide, myc tag, scFv, and the ectodomain composed of the CD8α hinge, Nucleotides encoding the transmembrane domain of CD8α, nucleotides encoding the intracellular domain composed of CD28, CD3ζ, and ubiquitin in tandem.
野生型泛素的核苷酸序列如SEQ ID NO.1所示。在一较佳实施方式中,所述泛素选自突变型泛素。本发明中选用的编码突变型泛素的多核苷酸序列如SEQ ID NO.2所示。The nucleotide sequence of wild-type ubiquitin is shown in SEQ ID NO.1. In a preferred embodiment, the ubiquitin is selected from mutant ubiquitin. The polynucleotide sequence encoding mutant ubiquitin selected in the present invention is shown in SEQ ID NO.2.
本发明提供一种核酸构建体,所述核酸构建体中包括所述分离的多核苷酸。The present invention provides a nucleic acid construct comprising the isolated polynucleotide.
术语“核酸构建体”是指可以被引入细胞或组织中的人工构建的核酸区段,所述核酸构建体为非病毒载体或病毒载体。所述病毒载体为慢病毒载体、腺病毒载体、腺相关病毒载体、杆状病毒载体。在本发明某些实施方式中,所述核酸构建体为慢病毒载体,慢病毒载体包括载体骨架即空载体与表达框架。所述空载体中包括多种控制表达的元件,包括启动子序列、转录起始序列、增强子序列、选择元件及报告基因。另外,载体还可含有复制起始位点。所述空载体例如为pHR-hEF1α-IRES-EGFP空载体。所述表达框架即为所述分离的多核苷酸。The term "nucleic acid construct" refers to an artificially constructed nucleic acid segment that can be introduced into cells or tissues, and the nucleic acid construct is a non-viral vector or a viral vector. The viral vectors are lentiviral vectors, adenoviral vectors, adeno-associated viral vectors, and baculoviral vectors. In some embodiments of the present invention, the nucleic acid construct is a lentiviral vector, and the lentiviral vector includes a vector backbone, that is, an empty vector and an expression framework. The empty vector includes various expression-controlling elements, including a promoter sequence, a transcription initiation sequence, an enhancer sequence, a selection element and a reporter gene. In addition, the vector may also contain an origin of replication. The empty vector is, for example, pHR-hEF1α-IRES-EGFP empty vector. The expression framework is the isolated polynucleotide.
术语“载体”是指用于将一个或多个核酸或一个或多个多核苷酸引入或转移到靶细胞或组织中的核酸片段或多核苷酸片段。典型地,载体用于将外源DNA引入另一个细胞或组织中。载体可以包含用于在细菌中生长的细菌抗性基因和用于在生物体中表达目的蛋白质的启动子。DNA可以通过PCR或任何其他本领域技术人员已知的一种或多种合适的技术在体外产生。The term "vector" refers to a nucleic acid fragment or polynucleotide fragment used to introduce or transfer one or more nucleic acids or one or more polynucleotides into a target cell or tissue. Typically, vectors are used to introduce foreign DNA into another cell or tissue. The vector may contain a bacterial resistance gene for growth in bacteria and a promoter for expressing a protein of interest in an organism. DNA can be produced in vitro by PCR or any other suitable technique or techniques known to those skilled in the art.
本发明提供一种免疫细胞,所述免疫细胞中包括所述核酸构建体或基因组中整合有外源的所述的多核苷酸,或能够表达所述的泛素修饰的嵌合抗原受体。The present invention provides an immune cell, which includes the nucleic acid construct or the exogenous polynucleotide integrated in the genome, or is capable of expressing the ubiquitin-modified chimeric antigen receptor.
所述免疫细胞选自CAR-T细胞、CAR-NK细胞、CAR-巨噬细胞或CAR-TIL细胞。The immune cells are selected from CAR-T cells, CAR-NK cells, CAR-macrophages or CAR-TIL cells.
本发明还提供所述免疫细胞的制备方法,所述制备方法包括将所述核酸构建体或所述多核苷酸导入免疫效应细胞或产生免疫效应细胞的干细胞中。The present invention also provides a preparation method of the immune cells, the preparation method comprising introducing the nucleic acid construct or the polynucleotide into immune effector cells or stem cells producing immune effector cells.
所述免疫效应细胞选自T细胞、NK细胞、巨噬细胞或TIL细胞。The immune effector cells are selected from T cells, NK cells, macrophages or TIL cells.
本发明还提供所述免疫细胞在制备治疗癌症或免疫性疾病药物中的用途。The present invention also provides the use of the immune cells in the preparation of medicines for treating cancer or immune diseases.
本申请中的“癌症”是指任何由肿瘤或恶性细胞生长、增殖或转移所介导,并引发实体瘤和非实体瘤如白血病的医学状态。本发明中的“肿瘤”是指肿瘤和/或恶性细胞的实体物质。"Cancer" in this application refers to any medical condition that is mediated by the growth, proliferation or metastasis of tumor or malignant cells and causes solid tumors and non-solid tumors such as leukemia. "Tumor" in the present invention refers to the solid substance of tumor and/or malignant cells.
所述癌症例如为非小细胞肺癌、小细胞肺癌、肾细胞癌、结肠直肠癌、卵巢癌、乳癌、胰脏癌、胃癌、膀胱癌、食管癌、间皮瘤、黑色素瘤、头颈部癌、甲状腺癌、肉瘤、前列腺癌、成胶质细胞瘤、子宫颈癌、胸腺癌;白血病、淋巴瘤、骨髓瘤、蕈样肉芽肿(mycoses fungoids)、默克尔细胞癌和其它恶性血液病、如经典型霍奇金淋巴瘤(CHL)、原发性纵隔大B细胞淋巴瘤、富于T细胞/组织细胞的大B细胞淋巴瘤、EBV阳性和阴性PTLD和EBV相关弥漫性大B细胞淋巴瘤(DLBCL)、浆母细胞性淋巴瘤、结外NK/T细胞淋巴瘤、鼻咽癌和HHV8相关原发性渗出性淋巴瘤、霍奇金淋巴瘤。The cancer is, for example, non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, esophageal cancer, mesothelioma, melanoma, head and neck cancer , thyroid cancer, sarcoma, prostate cancer, glioblastoma, cervical cancer, thymus cancer; leukemia, lymphoma, myeloma, mycoses fungoids (mycoses fungoids), Merkel cell carcinoma and other hematological malignancies, Such as classical Hodgkin lymphoma (CHL), primary mediastinal large B-cell lymphoma, T-cell/histiocytic-rich large B-cell lymphoma, EBV-positive and negative PTLD, and EBV-associated diffuse large B-cell lymphoma lymphoma (DLBCL), plasmablastic lymphoma, extranodal NK/T cell lymphoma, nasopharyngeal carcinoma and HHV8-related primary effusion lymphoma, Hodgkin lymphoma.
所述免疫性疾病例如为系统性红斑狼疮(SLE),自身免疫性糖尿病、银屑病、白癜风、硬皮症、类风湿性关节炎。The immune diseases are, for example, systemic lupus erythematosus (SLE), autoimmune diabetes, psoriasis, vitiligo, scleroderma, and rheumatoid arthritis.
对某种状态的“治疗”或“疗法”包括预防或减轻某种状态,降低某种状态发生或发展的速度,减少发展出某种状态的风险,预防或延迟与某种状态相关的症状发展,减少或终止与某种状态相关的症状,产生某种状态的完全或部分的逆转,治愈某种状态,或以上的组合。对于癌症来说,“治疗”或“疗法”可以指抑制或减缓肿瘤或恶性细胞生长,繁殖,或转移,或以上的某些组合。对于肿瘤来说,“治疗”或“疗法”包括清除全部或部分的肿瘤,抑制或减缓肿瘤生长和转移,预防或延缓肿瘤的发展,或以上的某些组合。"Treatment" or "therapy" for a condition includes preventing or alleviating a condition, reducing the rate at which a condition occurs or develops, reducing the risk of developing a condition, preventing or delaying the development of symptoms associated with a condition , to reduce or terminate symptoms associated with a condition, to produce a complete or partial reversal of a condition, to cure a condition, or a combination of the above. With respect to cancer, "treating" or "therapy" can refer to inhibiting or slowing the growth, reproduction, or metastasis of tumor or malignant cells, or some combination thereof. With respect to tumors, "treatment" or "therapy" includes eradicating all or part of the tumor, inhibiting or slowing tumor growth and metastasis, preventing or delaying tumor progression, or some combination of the above.
本发明还提供一种肿瘤或免疫性疾病的治疗方法,所述治疗方法包括向受试者施用治疗有效量的所述免疫细胞。The present invention also provides a treatment method for tumors or immune diseases, the treatment method comprising administering a therapeutically effective amount of the immune cells to a subject.
所述癌症例如为非小细胞肺癌、小细胞肺癌、肾细胞癌、结肠直肠癌、卵巢癌、乳癌、胰脏癌、胃癌、膀胱癌、食管癌、间皮瘤、黑色素瘤、头颈部癌、甲状腺癌、肉瘤、前列腺癌、成胶质细胞瘤、子宫颈癌、胸腺癌;白血病、淋巴瘤、骨髓瘤、蕈样肉芽肿(mycoses fungoids)、默克尔细胞癌和其它恶性血液病、如经典型霍奇金淋巴瘤(CHL)、原发性纵隔大B细胞淋巴瘤、T细胞/组织细胞的富B细胞淋巴瘤、EBV阳性和阴性PTLD和EBV相关弥漫性大B细胞淋巴瘤(DLBCL)、浆母细胞性淋巴瘤、结外NK/T细胞淋巴瘤、鼻咽癌和HHV8相 关原发性渗出性淋巴瘤、霍奇金淋巴瘤。The cancer is, for example, non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, esophageal cancer, mesothelioma, melanoma, head and neck cancer , thyroid cancer, sarcoma, prostate cancer, glioblastoma, cervical cancer, thymus cancer; leukemia, lymphoma, myeloma, mycoses fungoids (mycoses fungoids), Merkel cell carcinoma and other hematological malignancies, Such as classical Hodgkin lymphoma (CHL), primary mediastinal large B-cell lymphoma, T-cell/histiocytic B-rich lymphoma, EBV-positive and negative PTLD, and EBV-associated diffuse large B-cell lymphoma ( DLBCL), plasmablastic lymphoma, extranodal NK/T cell lymphoma, nasopharyngeal carcinoma and HHV8-related primary effusion lymphoma, Hodgkin lymphoma.
所述免疫性疾病例如为系统性红斑狼疮(SLE)、自身免疫性糖尿病、银屑病、白癜风、硬皮症、类风湿性关节炎。The immune diseases are, for example, systemic lupus erythematosus (SLE), autoimmune diabetes, psoriasis, vitiligo, scleroderma, rheumatoid arthritis.
本发明中的“治疗有效量”或“有效剂量”是指,某种药物有效治疗与嵌合抗原受体的抗原相关疾病或状态的剂量或浓度。例如,对于本发明中公开的抗体或其抗原结合片段的用途来说,治疗有效量是在该剂量或浓度下,该抗体或抗原结合物可以清除全部或部分肿瘤、抑制或减缓肿瘤生长、抑制介导癌状态的细胞的生长或繁殖、抑制肿瘤细胞转移、减轻任何与肿瘤或癌状态相关的症状或标记,预防或延缓肿瘤或癌状态的发展,或以上的某些组合。The "therapeutically effective amount" or "effective dose" in the present invention refers to the dose or concentration of a drug that can effectively treat the disease or state associated with the antigen of the chimeric antigen receptor. For example, for the use of the antibody or antigen-binding fragment thereof disclosed in the present invention, the therapeutically effective amount is at the dosage or concentration, the antibody or antigen-binding compound can eliminate all or part of the tumor, inhibit or slow down tumor growth, inhibit Mediating the growth or proliferation of cells in a cancerous state, inhibiting tumor cell metastasis, alleviating any symptoms or markers associated with a tumor or cancerous state, preventing or delaying the development of a tumor or cancerous state, or some combination of the above.
monoUb-CAR可以有效地促进CAR的内吞和降解,使细胞表面的CAR表达量大幅下降,实现免疫细胞例如CAR-T细胞本底自激活水平的大幅下降,明显降低免疫细胞例如T细胞分化过快、功能耗竭程度等情况。在本发明的某些实施方式中,monoUb-CAR显著增强了CD28 CAR-T细胞在体外、体内靶细胞刺激条件下的持续增殖能力,有效地提升了CAR-T细胞在荷瘤小鼠体内肿瘤的杀伤能力,进而提高了小鼠存活率。monoUb-CAR can effectively promote the endocytosis and degradation of CAR, greatly reduce the expression of CAR on the cell surface, realize a substantial decrease in the background self-activation level of immune cells such as CAR-T cells, and significantly reduce the over-differentiation of immune cells such as T cells. Speed, degree of functional exhaustion, etc. In some embodiments of the present invention, monoUb-CAR significantly enhances the sustained proliferation ability of CD28 CAR-T cells in vitro and in vivo under target cell stimulation conditions, and effectively improves the tumorigenicity of CAR-T cells in tumor-bearing mice. killing ability, thereby improving the survival rate of mice.
本发明在肿瘤小鼠模型上比较了CD28 monoUb CAR-T和CD28 WT CAR-T在体内的抗肿瘤效果。在增殖水平上,CD28 monoUb CAR-T有更强的增殖响应并且增殖能力更持久;在细胞分化表型上,无论在脾脏,血液,还是肿瘤中,改造的CAR-T都积累了更多的干细胞记忆T细胞(Stem cell memory T cell,T SCM)并减少了向终末效应型T细胞的分化。因而改造的CAR-T能更有效的浸润肿瘤组织进行杀伤,在相同的T细胞注射剂量下,改造的CAR-T能更有效地控制肿瘤的发展。 The present invention compares the anti-tumor effects of CD28 monoUb CAR-T and CD28 WT CAR-T in vivo on a tumor mouse model. At the level of proliferation, CD28 monoUb CAR-T has a stronger proliferative response and longer-lasting proliferative ability; in terms of cell differentiation phenotype, whether in the spleen, blood, or tumor, the engineered CAR-T has accumulated more Stem cell memory T cells (Stem cell memory T cells, T SCM ) and reduced differentiation to terminal effector T cells. Therefore, the modified CAR-T can more effectively infiltrate and kill tumor tissues, and at the same injection dose of T cells, the modified CAR-T can more effectively control the development of tumors.
以下通过特定的具体实例说明本发明的实施方式,本领域技术人员可由本说明书所揭露的内容轻易地了解本发明的其他优点与功效。本发明还可以通过另外不同的具体实施方式加以实施或应用,本说明书中的各项细节也可以基于不同观点与应用,在没有背离本发明的精神下进行各种修饰或改变。Embodiments of the present invention are described below through specific examples, and those skilled in the art can easily understand other advantages and effects of the present invention from the content disclosed in this specification. The present invention can also be implemented or applied through other different specific implementation modes, and various modifications or changes can be made to the details in this specification based on different viewpoints and applications without departing from the spirit of the present invention.
在进一步描述本发明具体实施方式之前,应理解,本发明的保护范围不局限于下述特定的具体实施方案;还应当理解,本发明实施例中使用的术语是为了描述特定的具体实施方案,而不是为了限制本发明的保护范围;在本发明说明书和权利要求书中,除非文中另外明确指出,单数形式“一个”、“一”和“这个”包括复数形式。Before further describing the specific embodiments of the present invention, it should be understood that the protection scope of the present invention is not limited to the following specific specific embodiments; it should also be understood that the terms used in the examples of the present invention are to describe specific specific embodiments, It is not intended to limit the protection scope of the present invention; in the description and claims of the present invention, unless the context clearly indicates otherwise, the singular forms "a", "an" and "the" include plural forms.
当实施例给出数值范围时,应理解,除非本发明另有说明,每个数值范围的两个端点以及两个端点之间任何一个数值均可选用。除非另外定义,本发明中使用的所有技术和科学术语与本技术领域技术人员通常理解的意义相同。除实施例中使用的具体方法、设备、材料外, 根据本技术领域的技术人员对现有技术的掌握及本发明的记载,还可以使用与本发明实施例中所述的方法、设备、材料相似或等同的现有技术的任何方法、设备和材料来实现本发明。When the examples give numerical ranges, it should be understood that, unless otherwise stated in the present invention, the two endpoints of each numerical range and any value between the two endpoints can be selected. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition to the specific methods, equipment, and materials used in the embodiments, according to those skilled in the art's grasp of the prior art and the description of the present invention, the methods, equipment, and materials described in the embodiments of the present invention can also be used Any methods, apparatus and materials of the prior art similar or equivalent to the practice of the present invention.
除非另外指明,本发明实施所采用的分子生物学、微生物学、细胞生物学、生物化学以及免疫学等常规技术操作均在技术人员的理解认知内。这些技术是广泛使用的并可以在以下文献中找到充分说明,诸如:“Molecular cloning:A Laboratory Manual,Fourth edition”(M.R.Green,et al.2014);“Oligonucleotide Synthesis”(M.J.Gait,et al.1984);“Polymerase Chain Reaction:Principles,Applications and Troubleshooting”(M.E.Babar,et al.2011);“Short Protocols in Molecular Biology,Fifth edition”(F.M.Ausubel,et al.2002);“Methods in Molecular Biology”(Humana Press);“Gene Transfer Vectors for Mammalian Cells”(J.H.Miller and M.P.Calos.1987);“Culture of Animal Cell”(R.I.Freshney,et al.2010);“Methods in Enzymology”(Academic Press,Inc);“Using Antibodies:A Laboratory Manual”(E.Harlow and D.Lane.1999);“Handbook of Experimental Immunology”(L.A.Herzenberg,et al.1997);“Current Protocols in Immunology”(J.E.Coligan,et al.2002)。Unless otherwise specified, conventional technical operations such as molecular biology, microbiology, cell biology, biochemistry and immunology used in the practice of the present invention are within the understanding of skilled artisans. These techniques are widely used and can be found fully described in literature such as: "Molecular cloning: A Laboratory Manual, Fourth edition" (M.R. Green, et al. 2014); "Oligonucleotide Synthesis" (M.J. Gait, et al. 1984); "Polymerase Chain Reaction: Principles, Applications and Troubleshooting" (M.E.Babar, et al.2011); "Short Protocols in Molecular Biology, Fifth edition" (F.M.Ausubel, et al.2002); "Methods in Molecular Biology" (Humana Press); "Gene Transfer Vectors for Mammalian Cells" (J.H.Miller and M.P.Calos.1987); "Culture of Animal Cell" (R.I.Freshney, et al.2010); "Methods in Enzymology" (Academic Press, Inc) ; "Using Antibodies: A Laboratory Manual" (E.Harlow and D.Lane.1999); "Handbook of Experimental Immunology" (L.A.Herzenberg, et al.1997); "Current Protocols in Immunology" (J.E.Coligan, et al. 2002).
实施例1:CAR的载体构建Embodiment 1: Vector construction of CAR
本发明使用的CD19、GD2、GPC3CAR的抗原特异性单链抗体(scFv)序列分别来自临床上使用的FMC63、14g2A和GPC3序列;双靶点CAR是将靶向CD19和CD22的特异性抗体进行整合,整合方式参考文章《CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy》中的报道。CAR的胞外段结构由CD8α信号肽序列、myc标签序列、scFv序列、CD8α的铰链序列串联构成;跨膜区序列为CD8α的跨膜区序列;胞内段结构由人CD28胞内段序列串联人的CD3ζ胞内段序列构成。所述泛素修饰的嵌合抗原受体即在胞内域中通过连接肽将泛素与CD3ζ连接。以上scFv氨基酸序列均经过密码子优化后转换成碱基序列,并由第三方公司合成(genscript)。本发明中所有CAR的碱基序列最终通过Gibson连接的方式克隆至pHR-hEF1α-IRES-EGFP载体(源自addgene)中。The antigen-specific single-chain antibody (scFv) sequences of CD19, GD2, and GPC3 CAR used in the present invention are respectively from the clinically used FMC63, 14g2A, and GPC3 sequences; the dual-target CAR is the integration of specific antibodies targeting CD19 and CD22 For the integration method, refer to the report in the article "CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy". The extracellular segment structure of CAR is composed of CD8α signal peptide sequence, myc tag sequence, scFv sequence, and CD8α hinge sequence in series; the transmembrane sequence is the transmembrane region sequence of CD8α; the intracellular segment structure is composed of human CD28 intracellular segment sequence in series Sequence composition of human CD3ζ intracellular segment. The ubiquitin-modified chimeric antigen receptor connects ubiquitin to CD3ζ through a linker peptide in the intracellular domain. The above scFv amino acid sequences were converted into base sequences after codon optimization, and synthesized by a third-party company (genscript). The base sequences of all CARs in the present invention were finally cloned into the pHR-hEF1α-IRES-EGFP vector (derived from addgene) by Gibson connection.
实施例2:人的原代T细胞培养及慢病毒感染Example 2: Human primary T cell culture and lentivirus infection
人的原代T细胞均取自健康的知情志愿者。原代T细胞培养在含有10%的胎牛血清、100U/ml的青霉素和100μg/ml的硫酸链霉、1mM丙酮酸钠、非必须氨基酸、55μM 2-巯基乙醇的RPMI-1640培养基中(以上试剂均购自Gibco)。为了维持T细胞的增殖,培养基中加入100U/ml的hIL-2(Sigma-Aldrich)。Human primary T cells were obtained from healthy informed volunteers. Primary T cells were cultured in RPMI-1640 medium containing 10% fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin sulfate, 1 mM sodium pyruvate, non-essential amino acids, and 55 μM 2-mercaptoethanol ( The above reagents were purchased from Gibco). In order to maintain the proliferation of T cells, 100 U/ml of hIL-2 (Sigma-Aldrich) was added to the culture medium.
制备慢病毒:将Lenti-X 293T细胞(TaKaRa#632180)重悬于含10%胎牛血清、不含抗生素的DMEM培养基(Gibco#11995-065),并以6.5×10 5个细胞/孔的密度接种于6孔细胞培养板(Corning#CLS3516)里培养24小时。利用脂质体转染系统(Mirus#2300),将500ng慢病毒包装质粒pCMVdR8.92(Addgene#8455)和50ng pMD2.G(Addgene#12259)与500ng待包装的慢病毒质粒,按照脂质体转染说明书的操作步骤混合后,加入Lenti-X 293T细胞。16-18小时后弃去含脂质体的培养基,并补充适量新鲜培养基;48小时后收集细胞上清,直接或超速离心浓缩后,冻存于-80℃冰箱中备用。 Preparation of lentivirus: Resuspend Lenti-X 293T cells (TaKaRa#632180) in DMEM medium (Gibco#11995-065) containing 10% fetal bovine serum and without antibiotics, and make 6.5×10 5 cells/well The density was inoculated in 6-well cell culture plate (Corning#CLS3516) and cultured for 24 hours. Using liposome transfection system (Mirus#2300), mix 500ng lentiviral packaging plasmid pCMVdR8.92 (Addgene#8455) and 50ng pMD2.G (Addgene#12259) with 500ng lentiviral plasmid to be packaged, according to liposome After mixing the operation steps of the transfection instructions, add Lenti-X 293T cells. Discard the liposome-containing medium after 16-18 hours, and add an appropriate amount of fresh medium; collect the cell supernatant after 48 hours, concentrate directly or by ultracentrifugation, and store in a -80°C refrigerator for later use.
原代T细胞的慢病毒感染:使用包被了抗人CD3和抗人CD28抗体的磁珠(Life Technologies#11132D)激活T细胞,T细胞与磁珠1:3混合,培养24小时后加入制备好的慢病毒进行感染;18小时后将含病毒液的培养基弃除并置换为新鲜的T细胞完全培养基。T细胞经磁珠刺激4-5天后,去除磁珠,以T细胞完全培养基调整细胞密度至0.8-1*10^6/ml,每隔2天补充新鲜的T细胞完全培养基。Lentiviral infection of primary T cells: Use magnetic beads (Life Technologies #11132D) coated with anti-human CD3 and anti-human CD28 antibodies to activate T cells, mix T cells with magnetic beads 1:3, add to the preparation after 24 hours of culture Infect with a good lentivirus; after 18 hours, the medium containing the virus solution was discarded and replaced with fresh T cell complete medium. After T cells were stimulated by magnetic beads for 4-5 days, the magnetic beads were removed, and the cell density was adjusted to 0.8-1*10^6/ml with T cell complete medium, and fresh T cell complete medium was supplemented every 2 days.
将针对不同靶标的偶联单泛素的CD28 CAR表达于Jurkat细胞系的步骤与表达于T细胞的步骤相同。The procedure for expressing monoubiquitin-conjugated CD28 CARs targeting different targets in Jurkat cell lines was the same as in T cells.
实施例3:流式细胞术分析Example 3: Flow Cytometry Analysis
对于细胞表面标志物的染色:将抗体以合适比例稀释于FACS缓冲液(磷酸盐缓冲液PBS+2%胎牛血清)中,并以适量抗体稀释液重悬细胞,在4℃黑暗环境下共孵育25分钟;以FACS缓冲液洗涤细胞3次后,以含有适当浓度DAPI的FACS缓冲液重悬细胞,上机检测。For staining of cell surface markers: dilute the antibody in FACS buffer (phosphate buffered saline PBS + 2% fetal bovine serum) at an appropriate ratio, and resuspend the cells with an appropriate amount of antibody diluent, and co-suspend in a dark environment at 4°C. Incubate for 25 minutes; wash the cells 3 times with FACS buffer, resuspend the cells in FACS buffer containing appropriate concentration of DAPI, and detect on the machine.
流式细胞术的数据通过BD LSRFortessa机器(BD bioscience)获得并使用FlowJo software软件(Tree Star)进行数据分析。流式细胞术所用抗体信息列表如下。Flow cytometry data were acquired by a BD LSR Fortessa machine (BD bioscience) and analyzed using FlowJo software (Tree Star). Antibodies used in flow cytometry are listed below.
Figure PCTCN2022080608-appb-000003
Figure PCTCN2022080608-appb-000003
Figure PCTCN2022080608-appb-000004
Figure PCTCN2022080608-appb-000004
实施例4:基于流式细胞术的CAR-T体外杀伤功能检测Example 4: In vitro killing function detection of CAR-T based on flow cytometry
将表达CD19和mCherry荧光的双阳性K562靶细胞与不表达CD19和mCherry荧光的K562非靶细胞按照1:1比例混合后,与CAR-T细胞按照一定的效应细胞:靶细胞比例混合并共孵育24小时。细胞培养在无IL-2的T细胞完全培养基中。流式细胞术分析:以不含T细胞的K562混合细胞群作为对照组,分析得到K562靶细胞所占K562细胞的比例(CK%);在含T细胞的实验组中通过CD3ε染色将T细胞区分出K562细胞群,然后分析CD19-K562靶细胞所占总K562细胞群的比例(EX%),T细胞的杀伤效率=(1-EX%/CK%)×100%After mixing double-positive K562 target cells expressing CD19 and mCherry fluorescence with K562 non-target cells not expressing CD19 and mCherry fluorescence at a ratio of 1:1, they were mixed with CAR-T cells at a certain ratio of effector cells: target cells and co-incubated 24 hours. Cells were cultured in complete T cell medium without IL-2. Flow cytometry analysis: the K562 mixed cell population without T cells was used as the control group, and the proportion of K562 target cells in K562 cells (CK%) was analyzed; in the experimental group containing T cells, the T cells were stained by CD3ε Distinguish the K562 cell population, and then analyze the proportion (EX%) of CD19-K562 target cells in the total K562 cell population, the killing efficiency of T cells=(1-EX%/CK%)×100%
实施例5:CAR-T细胞体外增殖能力检测Example 5: Detection of CAR-T cell proliferation ability in vitro
将CD19-28WT-CAR和CD19-28Mono-CAR T细胞计数后,分别与辐照后的靶细胞Nalm6 3:1混合,以无IL-2的T细胞完全培养基重悬细胞至细胞密度为1*10^6/ml;每2天进行活细胞计数以计算细胞增殖量,并以无IL-2的T细胞完全培养基调节细胞密度至1*10^6/ml;当活细胞计数所得细胞密度小于或等于1*10^6/ml时,表明细胞不再增殖并终止实验。After CD19-28WT-CAR and CD19-28Mono-CAR T cells were counted, they were mixed with irradiated target cells Nalm6 3:1, and the cells were resuspended in IL-2-free T cell complete medium to a cell density of 1 *10^6/ml; Count the viable cells every 2 days to calculate the amount of cell proliferation, and adjust the cell density to 1*10^6/ml with the complete medium of T cells without IL-2; when the cells obtained by the viable cell count When the density is less than or equal to 1*10^6/ml, it indicates that the cells are no longer proliferating and the experiment is terminated.
实施例6:小鼠肿瘤模型及CAR-T细胞功能检测Example 6: Mouse tumor model and CAR-T cell function detection
体内实验对象为5至8周大的联合免疫缺陷型(NSG)小鼠。为了比较CAR-T的体内抗肿瘤效果,首先给NSG小鼠尾静脉接种1×10 6个表达有萤火虫荧光素酶基因的B淋巴瘤细胞Nalm6;待靶细胞在体内生长4天后,通过尾静脉注射的方式给予NSG小鼠2×10 6个CAR-T细胞的治疗;每周通过小动物活体成像系统,检测小鼠肿瘤细胞所携带的萤火虫荧光素酶的强度,以反映肿瘤细胞的负荷,从而追踪肿瘤在体内的发展状况。小动物活体成像系统的具体实施操作包括:通过腹腔注射给予荷瘤小鼠萤火虫荧光素酶底物(D-荧光素钠盐),底物用量按照0.15mg/g小鼠体重施用;10分钟后待底物充分循环至小鼠全身,用2.5%-3.5%的异氟烷气体麻醉小鼠后进行成像。生物发光成像通过IVIS光谱成像系统(Perkin Elmer)执行,荧光定量数据通过活体成像软件(Perkin Elmer)获得。 In vivo experiments were performed on 5- to 8-week-old combined immunodeficiency (NSG) mice. In order to compare the anti-tumor effect of CAR-T in vivo, NSG mice were first inoculated with 1×106 B lymphoma cells Nalm6 expressing the firefly luciferase gene in the tail vein; Treat NSG mice with 2×10 6 CAR-T cells by injection; detect the intensity of firefly luciferase carried by mouse tumor cells every week through a small animal in vivo imaging system to reflect the load of tumor cells, To track the development of tumors in the body. The specific implementation of the small animal in vivo imaging system includes: giving the firefly luciferase substrate (D-luciferin sodium salt) to tumor-bearing mice by intraperitoneal injection, and the substrate dosage is 0.15 mg/g mouse body weight; 10 minutes later After the substrate was fully circulated to the whole body of the mouse, the mouse was anesthetized with 2.5%-3.5% isoflurane gas and then imaged. Bioluminescence imaging was performed with an IVIS Spectral Imaging System (Perkin Elmer), and fluorescence quantitative data were acquired with an in vivo imaging software (Perkin Elmer).
实施例7:荷瘤小鼠体内CAR-T细胞增殖、分化与耗竭水平检测Example 7: Detection of CAR-T cell proliferation, differentiation and exhaustion in tumor-bearing mice
体内实验对象为5至8周大的联合免疫缺陷型(NSG)小鼠。为了比较CAR-T的体内增殖、分化、耗竭水平的差异,首先给NSG小鼠尾静脉接种2×10 6个B淋巴瘤细胞Nalm6;待靶细胞在体内生长4天后,通过尾静脉注射的方式给予NSG小鼠2×10 6个CAR-T细胞; 此后7天、14天、21天各处死3只接种CD19-28 WT和CD19-28 MonoUb CAR-T细胞的小鼠,取其脾脏与后肢长骨;研磨脾脏取脾细胞;以带20号针头的5ml注射器吸取适量RPMI培养基,小心剪断长骨的两端并以注射器针头刺入长骨骨髓腔,冲出骨髓细胞;以适量红细胞裂解液处理脾细胞与骨髓细胞后,脾细胞计数,并将细胞等分,分别孵育不同待检测指标抗体稀释液,并进行流式检测。 In vivo experiments were performed on 5- to 8-week-old combined immunodeficiency (NSG) mice. In order to compare the differences in the proliferation, differentiation, and exhaustion levels of CAR-T in vivo, NSG mice were first inoculated with 2 ×106 B lymphoma cells Nalm6 in the tail vein; after the target cells grew in vivo for 4 days, they were injected through the tail vein. 2×10 6 CAR-T cells were administered to NSG mice; 3 mice inoculated with CD19-28 WT and CD19-28 MonoUb CAR-T cells were sacrificed 7 days, 14 days, and 21 days later, and their spleens and hind limbs were collected. Long bones; grind the spleen to get spleen cells; draw appropriate amount of RPMI medium with a 5ml syringe with a 20-gauge needle, carefully cut off the two ends of the long bone and insert the syringe needle into the marrow cavity of the long bone to flush out the bone marrow cells; treat the spleen with an appropriate amount of erythrocyte lysate After cells and bone marrow cells, the splenocytes were counted, the cells were aliquoted, and different antibody dilutions of the indicators to be detected were incubated respectively, and flow cytometric detection was performed.
以上的实施例是为了说明本发明公开的实施方案,并不能理解为对本发明的限制。此外,本文所列出的各种修改以及发明中方法的变化,在不脱离本发明的范围和精神的前提下对本领域内的技术人员来说是显而易见的。虽然已结合本发明的多种具体优选实施例对本发明进行了具体的描述,但应当理解,本发明不应仅限于这些具体实施例。事实上,各种如上所述的对本领域内的技术人员来说显而易见的修改来获取发明都应包括在本发明的范围内。The above examples are intended to illustrate the disclosed embodiments of the present invention, and should not be construed as limiting the present invention. In addition, various modifications set forth herein, as well as changes in the method of the invention, will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been specifically described in connection with various specific preferred embodiments of the invention, it should be understood that the invention should not be limited to these specific embodiments. In fact, various modifications as mentioned above which are obvious to those skilled in the art to obtain the invention should be included in the scope of the present invention.

Claims (17)

  1. 一种嵌合抗原受体的改造方法,其特征在于,所述改造方法包括将泛素偶联至嵌合抗原受体的C端。A chimeric antigen receptor modification method, characterized in that the modification method includes coupling ubiquitin to the C-terminus of the chimeric antigen receptor.
  2. 一种泛素修饰的嵌合抗原受体,其特征在于,所述泛素修饰的嵌合抗原受体包括跨膜域、胞内域和胞外域,所述胞内域C端偶联有泛素。A ubiquitin-modified chimeric antigen receptor, characterized in that the ubiquitin-modified chimeric antigen receptor comprises a transmembrane domain, an intracellular domain and an extracellular domain, and the C-terminus of the intracellular domain is coupled with a ubiquitous white.
  3. 根据权利要求2所述的泛素修饰的嵌合抗原受体,其特征在于,还包括以下特征中的任一项或多项:The ubiquitin-modified chimeric antigen receptor according to claim 2, further comprising any one or more of the following features:
    1)所述跨膜域选自CD8α、CD28或DAP 10;1) the transmembrane domain is selected from CD8α, CD28 or DAP 10;
    2)所述胞内域包括共刺激结构域和/或信号结构域。2) The intracellular domain includes costimulatory domain and/or signaling domain.
  4. 根据权利要求3所述的泛素修饰的嵌合抗原受体,其特征在于,所述胞内域选自以下中的一种或几种的组合:4-1BB、CD28、OX40、ICOS、CD3ζ或DAP 10。The ubiquitin-modified chimeric antigen receptor according to claim 3, wherein the intracellular domain is selected from one or a combination of the following: 4-1BB, CD28, OX40, ICOS, CD3ζ or DAP 10.
  5. 根据权利要求2所述的泛素修饰的嵌合抗原受体,其特征在于,所述嵌合抗原受体中依次包括CD8α信号肽、蛋白纯化标签、单链抗体、CD8α铰链组成的胞外域,CD8α跨膜域,CD28和CD3ζ串联组成的胞内域。The ubiquitin-modified chimeric antigen receptor according to claim 2, wherein the chimeric antigen receptor comprises CD8α signal peptide, protein purification tag, single-chain antibody, and CD8α hinge in sequence in order to form an ectodomain, The transmembrane domain of CD8α, the intracellular domain composed of CD28 and CD3ζ in tandem.
  6. 根据权利要求5所述的泛素修饰的嵌合抗原受体,其特征在于,单链抗体选自GD2 scFv、GPC3 scFv、Her2 scFv、CSPG4 scFv、EGFR scFv、Meso scFv、TRBC1 scFv、CD133 scFv、BCMA scFv或CD19 scFv中的任一个或多个。The ubiquitin-modified chimeric antigen receptor according to claim 5, wherein the single-chain antibody is selected from GD2 scFv, GPC3 scFv, Her2 scFv, CSPG4 scFv, EGFR scFv, Meso scFv, TRBC1 scFv, CD133 scFv, Any one or more of BCMA scFv or CD19 scFv.
  7. 根据权利要求2所述的泛素修饰的嵌合抗原受体,其特征在于,所述泛素选自野生型泛素或突变型泛素。The ubiquitin-modified chimeric antigen receptor according to claim 2, wherein the ubiquitin is selected from wild-type ubiquitin or mutant ubiquitin.
  8. 根据权利要求2所述的泛素修饰的嵌合抗原受体,其特征在于,所述泛素选自突变型泛素,所述突变型泛素的氨基酸序列如SEQ ID NO.18所示。The ubiquitin-modified chimeric antigen receptor according to claim 2, wherein the ubiquitin is selected from mutant ubiquitin, and the amino acid sequence of the mutant ubiquitin is as shown in SEQ ID NO.18.
  9. 一种分离的多核苷酸,其特征在于,所述分离的多核苷酸包括编码权利要求2-8任一所述的泛素修饰的嵌合抗原受体的核苷酸。An isolated polynucleotide, characterized in that the isolated polynucleotide comprises nucleotides encoding the ubiquitin-modified chimeric antigen receptor according to any one of claims 2-8.
  10. 一种核酸构建体,其特征在于,所述核酸构建体中包括权利要求9所述的多核苷酸。A nucleic acid construct, characterized in that the nucleic acid construct comprises the polynucleotide according to claim 9.
  11. 一种免疫细胞,其特征在于,所述免疫细胞中包括权利要求10所述的核酸构建体或基因组中整合有外源的权利要求9所述的多核苷酸,或能够表达权利要求2-8任一所述的泛素修饰的嵌合抗原受体。An immune cell, characterized in that, the immune cell comprises the nucleic acid construct according to claim 10 or the exogenous polynucleotide according to claim 9 is integrated in the genome, or is capable of expressing the polynucleotide according to claims 2-8 Any of the ubiquitin-modified chimeric antigen receptors.
  12. 根据权利要求11所述的免疫细胞,其特征在于,所述免疫细胞选自CAR-T细胞、CAR-NK细胞、CAR-巨噬细胞或CAR-TIL细胞。The immune cell according to claim 11, wherein the immune cell is selected from CAR-T cells, CAR-NK cells, CAR-macrophages or CAR-TIL cells.
  13. 权利要求11-12任一所述的免疫细胞的制备方法,其特征在于,所述制备方法包括将权利要求10所述的核酸构建体或权利要求9所述的多核苷酸导入免疫效应细胞或产生免疫 效应细胞的干细胞中。The preparation method of immune cells according to any one of claims 11-12, characterized in that the preparation method comprises introducing the nucleic acid construct according to claim 10 or the polynucleotide according to claim 9 into immune effector cells or In stem cells that produce immune effector cells.
  14. 根据权利要求13所述的制备方法,其特征在于,所述免疫效应细胞选自T细胞、NK细胞、巨噬细胞或TIL细胞。The preparation method according to claim 13, characterized in that the immune effector cells are selected from T cells, NK cells, macrophages or TIL cells.
  15. 权利要求11-12任一所述的免疫细胞在制备治疗癌症或免疫性疾病药物中的用途。Use of the immune cells according to any one of claims 11-12 in the preparation of medicines for treating cancer or immune diseases.
  16. 一种肿瘤或免疫性疾病的治疗方法,其特征在于,所述治疗方法包括向受试者施用治疗有效量的权利要求11-12任一所述免疫细胞。A treatment method for tumors or immune diseases, characterized in that the treatment method comprises administering a therapeutically effective amount of the immune cells according to any one of claims 11-12 to a subject.
  17. 根据权利要求16所述的治疗方法,其特征在于,所述癌症为非小细胞肺癌、小细胞肺癌、肾细胞癌、结肠直肠癌、卵巢癌、乳癌、胰脏癌、胃癌、膀胱癌、食管癌、间皮瘤、黑色素瘤、头颈部癌、甲状腺癌、肉瘤、前列腺癌、成胶质细胞瘤、子宫颈癌、胸腺癌;白血病、淋巴瘤、骨髓瘤、蕈样肉芽肿、默克尔细胞癌和其它恶性血液病、经典型霍奇金淋巴瘤、原发性纵隔大B细胞淋巴瘤、富于T细胞或组织细胞的大B细胞淋巴瘤、EBV阳性和阴性PTLD和EBV相关弥漫性大B细胞淋巴瘤、浆母细胞性淋巴瘤、结外NK/T细胞淋巴瘤、鼻咽癌或HHV8相关原发性渗出性淋巴瘤、霍奇金淋巴瘤;所述免疫性疾病为系统性红斑狼疮、自身免疫性糖尿病、银屑病、白癜风、硬皮症、类风湿性关节炎。The treatment method according to claim 16, wherein the cancer is non-small cell lung cancer, small cell lung cancer, renal cell carcinoma, colorectal cancer, ovarian cancer, breast cancer, pancreatic cancer, gastric cancer, bladder cancer, esophageal cancer Carcinoma, Mesothelioma, Melanoma, Head and Neck Cancer, Thyroid Cancer, Sarcoma, Prostate Cancer, Glioblastoma, Cervical Cancer, Thymic Cancer; Leukemia, Lymphoma, Myeloma, Mycosis Fungoides, Merck Myeloid cell carcinoma and other hematologic malignancies, classical Hodgkin lymphoma, primary mediastinal large B-cell lymphoma, T-cell or histiocyte-rich large B-cell lymphoma, EBV-positive and negative PTLD, and EBV-associated diffuse Sexual large B-cell lymphoma, plasmablastic lymphoma, extranodal NK/T-cell lymphoma, nasopharyngeal carcinoma or HHV8-related primary effusion lymphoma, Hodgkin's lymphoma; the immune diseases are Systemic lupus erythematosus, autoimmune diabetes, psoriasis, vitiligo, scleroderma, rheumatoid arthritis.
PCT/CN2022/080608 2021-07-29 2022-03-14 Chimeric antigen receptor modified by ubiquitin coupling and immune cell WO2023005219A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110863392.4A CN115677861A (en) 2021-07-29 2021-07-29 Ubiquitin-coupled modified chimeric antigen receptor and immune cell
CN202110863392.4 2021-07-29

Publications (1)

Publication Number Publication Date
WO2023005219A1 true WO2023005219A1 (en) 2023-02-02

Family

ID=85058999

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/080608 WO2023005219A1 (en) 2021-07-29 2022-03-14 Chimeric antigen receptor modified by ubiquitin coupling and immune cell

Country Status (2)

Country Link
CN (1) CN115677861A (en)
WO (1) WO2023005219A1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120283408A1 (en) * 2009-11-03 2012-11-08 Samsung Electronic Co., Ltd. Fusion protein binding specifically to constant region of antibody, method of preparing the fusion protein, and method of isolating antibody using the fusion protein
US20130123199A1 (en) * 2011-08-16 2013-05-16 Samsung Electronics Co., Ltd. Protein complex for intracellular delivery and uses thereof
CN109021116A (en) * 2018-08-16 2018-12-18 重庆精准生物技术有限公司 The Chimeric antigen receptor and its application of anti-BCMA antigen
US20190010220A1 (en) * 2014-08-28 2019-01-10 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
CN110055281A (en) * 2019-04-25 2019-07-26 山东大学第二医院 A kind of slow virus carrier being used to prepare CAR-T and its construction method and application
CN111686128A (en) * 2019-03-12 2020-09-22 重庆精准生物技术有限公司 Application of hypoxia-controllable promoter in CAR-T

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120283408A1 (en) * 2009-11-03 2012-11-08 Samsung Electronic Co., Ltd. Fusion protein binding specifically to constant region of antibody, method of preparing the fusion protein, and method of isolating antibody using the fusion protein
US20130123199A1 (en) * 2011-08-16 2013-05-16 Samsung Electronics Co., Ltd. Protein complex for intracellular delivery and uses thereof
US20190010220A1 (en) * 2014-08-28 2019-01-10 Bioatla, Llc Conditionally active chimeric antigen receptors for modified t-cells
CN109021116A (en) * 2018-08-16 2018-12-18 重庆精准生物技术有限公司 The Chimeric antigen receptor and its application of anti-BCMA antigen
CN111686128A (en) * 2019-03-12 2020-09-22 重庆精准生物技术有限公司 Application of hypoxia-controllable promoter in CAR-T
CN110055281A (en) * 2019-04-25 2019-07-26 山东大学第二医院 A kind of slow virus carrier being used to prepare CAR-T and its construction method and application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KUNII NAOKI, ZHAO YANGBING, JIANG SHUGUANG, LIU XIAOJUN, SCHOLLER JOHN, BALAGOPALAN LAKSHMI, SAMELSON LAWRENCE E., MILONE MICHAEL : "Enhanced Function of Redirected Human T Cells Expressing Linker for Activation of T Cells That Is Resistant to Ubiquitylation", HUMAN GENE THERAPY, MARY ANN LIEBERT, INC. PUBLISHERS, GB, vol. 24, no. 1, 1 January 2013 (2013-01-01), GB , pages 27 - 37, XP055830818, ISSN: 1043-0342, DOI: 10.1089/hum.2012.130 *

Also Published As

Publication number Publication date
CN115677861A (en) 2023-02-03

Similar Documents

Publication Publication Date Title
JP7217970B2 (en) Compositions and methods for reprogramming T-cell receptors using fusion proteins
JP2021040652A (en) Chimeric antigen receptors targeting g-protein coupled receptor and uses thereof
US20220401475A1 (en) Combinations of multiple chimeric antigen receptors for immunotherapy
KR20180021137A (en) Chimeric antigen receptor (CAR), compositions and methods for their use
US20210214415A1 (en) Immunoresponsive cells expressing dominant negative fas and uses thereof
WO2022007795A1 (en) Chimeric antigen receptor and use thereof
JP2022520285A (en) Hypoxia-responsive chimeric antigen receptor
US20230087125A1 (en) Chimeric antigen receptors targeting cd127 and use thereof
TW201930589A (en) Immunocompetent cell that expresses a cell surface molecule specifically recognizing human mesothelin, il-7 and ccl19
US20230346938A1 (en) Chimeric antigen receptors targeting cd19 and use thereof
CN113416260A (en) Claudin18.2-targeted specific chimeric antigen receptor cell and preparation method and application thereof
EP3964568A1 (en) Immune cell containing tumor antigen recognition receptor and application thereof
US20230058774A1 (en) Novel dominant negative fas polypeptides, cells comprising thereof and uses thereof
WO2023005219A1 (en) Chimeric antigen receptor modified by ubiquitin coupling and immune cell
WO2022097750A1 (en) Chimeric antigen receptor
EP4060027A1 (en) Tmem59 protein dimer or chimeric expression receptor improving t cell function
JP2023552724A (en) Chimeric receptor and its use
CN115397461A (en) Chimeric antigen receptor with CD28 mutation and application thereof
RU2812917C2 (en) Hla-non-restricted t-cell receptors and their use
CN114644716A (en) anti-BXMAS 1 chimeric antigen receptor, immune cell modified by same and application of immune cell

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: 22847839

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE