WO2022083590A1 - Récepteur chimérique contenant un dap 12 et domaine de signal de molécule de signal co-stimulateur, et son procédé d'utilisation - Google Patents

Récepteur chimérique contenant un dap 12 et domaine de signal de molécule de signal co-stimulateur, et son procédé d'utilisation Download PDF

Info

Publication number
WO2022083590A1
WO2022083590A1 PCT/CN2021/124728 CN2021124728W WO2022083590A1 WO 2022083590 A1 WO2022083590 A1 WO 2022083590A1 CN 2021124728 W CN2021124728 W CN 2021124728W WO 2022083590 A1 WO2022083590 A1 WO 2022083590A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
acid sequence
seq
dap12
identity
Prior art date
Application number
PCT/CN2021/124728
Other languages
English (en)
Chinese (zh)
Inventor
王恩秀
汪晨
孙明
Original Assignee
南京卡提医学科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CN202011122767.3A external-priority patent/CN114369168A/zh
Priority claimed from CN202110560490.0A external-priority patent/CN115368471A/zh
Priority claimed from CN202110560554.7A external-priority patent/CN115368472A/zh
Application filed by 南京卡提医学科技有限公司 filed Critical 南京卡提医学科技有限公司
Publication of WO2022083590A1 publication Critical patent/WO2022083590A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464466Adhesion molecules, e.g. NRCAM, EpCAM or cadherins
    • A61K39/464468Mesothelin [MSLN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • A61K39/464412CD19 or B4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61K39/464469Tumor associated carbohydrates
    • A61K39/464471Gangliosides, e.g. GM2, GD2 or GD3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes

Definitions

  • the present disclosure relates to the field of tumor cell therapy, and more particularly, to expressing a chimeric receptor comprising DAP12 and a signaling domain of a costimulatory signaling molecule, immune effector cells thereof, compositions comprising the same, and methods of making and using the same.
  • CD19 is the Cluster of Differentiation 19 protein (CD19), which is an epitope detectable on leukemic precursor cells. CD19 is expressed on most B-lineage cancers, such as acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin's lymphoma. CD19 is also an early marker of B cell ancestry.
  • CD19 Cluster of Differentiation 19 protein
  • MSLN Mesothelin
  • GPI domain glycophosphatidylinositol domain
  • mesothelin is overexpressed in the vast majority of primary pancreatic adenomas, whereas rare and weak expression is seen in benign pancreatic tissue.
  • Epithelial malignant pleural mesothelioma (MPM) ubiquitously expresses mesothelin whereas sarcomatoid MPM does not.
  • Most serous epithelial ovarian tumors and related primary peritonomas express mesothelin.
  • mesothelin is a target of the innate immune response and has been proposed as a target for cancer immunotherapy.
  • the presence of mesothelin-specific CTLs is associated with overall survival.
  • Soluble antibody fragments of anti-mesothelin antibodies conjugated to immunotoxins have been used to treat cancer patients with mesothelin-positive tumors. This approach has demonstrated adequate safety and some clinical activity in pancreatic cancer. In ovarian cancer, this treatment strategy produced a minimal response and stable disease in a second patient who had completely resolved their ascites according to RECIST criteria.
  • Disialoganglioside (GD2, pubchem: 6450346) is a sialic acid-containing glycosphingolipid, mainly expressed on the cell surface, which is found in tumors of neuroectodermal origin, including neuroblastoma and melanoma ), whereas expression in normal tissues is highly restricted.
  • GD2 is densely, homogeneously and almost ubiquitously expressed on neuroblastomas. In normal tissues, GD2 expression is largely restricted to skin melanocytes, and peripheral pain fibrous myelin sheaths. In the CNS, GD2 appears to be an embryonic antigen, but is found dimly expressed in scattered oligodendrocytes and in the posterior pituitary. This makes GD2 very suitable for targeted antitumor therapy.
  • CAR-T chimeric antigen receptor T cell
  • the chimeric antigen receptor is the core component of CAR-T.
  • CAR Using the ligand-binding domain properties, CAR can redirect its specificity and reactivity against selected immune cells, thus giving T cells an HLA-independent manner.
  • the ability to recognize tumor antigens allows CAR-engineered T cells to recognize a wider range of targets than the native T-cell surface receptor TCR.
  • the basic design of CAR includes a tumor-associated antigen (TAA) binding region (usually derived from the scFv segment of the antigen-binding region of a monoclonal antibody), an extracellular hinge region, a transmembrane region and an intracellular signal. Area.
  • TAA tumor-associated antigen
  • CAR-T therapy has shown promise in some hematological cancer trials. Clinical trials have shown that CAR-T cell therapy has great potential in controlling advanced acute lymphoblastic leukemia (ALL) and lymphoma.
  • ALL advanced acute lymphoblastic leukemia
  • CRS cytokine release syndrome
  • cytokine storm also known as cytokine cascade or hypercytokineemia
  • cytokine cascade hypercytokineemia
  • CN107580628A discloses targeted cytotoxic cells with chimeric receptors for adoptive immunotherapy, in which DAP12/KIRS2 is used as the intracellular signaling domain of the CAR structure.
  • DAP12/KIRS2 is used as the intracellular signaling domain of the CAR structure.
  • costimulatory signaling molecules due to the lack of costimulatory signaling molecules, it has technical problems of poor sustainability.
  • the amount of IL-2 secretion of DAP12/KIRS2 CAR-T is very low, and in clinical treatment lead to ineffective treatment.
  • the present disclosure introduces costimulatory signal molecules and anti-tumor antigen single-chain antibodies into DAP12/KIRS2 to form a CAR structure of anti-tumor antigen antibodies (anti-tumor antigen single-chain antibody/ DAP12-4-1BB), and validated the in vitro pharmacodynamics (killing, cytokine secretion, proliferation, etc.) based on this CAR structure, demonstrating that the CAR structure is effective against tumor antigens (such as CD19 antigen, mesothelin antigen and GD2 antigen). ) expression-positive solid tumor cells have effective killing effect.
  • tumor antigens such as CD19 antigen, mesothelin antigen and GD2 antigen
  • the present disclosure provides a chimeric antigen receptor comprising a first fusion peptide and a second fusion peptide, wherein:
  • the first fusion peptide comprises an antigen binding domain and a transmembrane domain
  • the second fusion peptide comprises a transmembrane domain, a cytoplasmic domain and a costimulatory domain.
  • the transmembrane domain of the second fusion peptide interacts with the transmembrane domain of the first fusion peptide through charge interactions, or the second fusion peptide interacts with phosphorylated ITAM within the cytoplasmic domain
  • the sequence interacts with the signaling molecule.
  • the transmembrane domain of the first fusion peptide is a KIR transmembrane domain; preferably, the KIR transmembrane domain is selected from the group consisting of KIR2DS2, KIR2DL3, KIR2DL1, KIR2DL2, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DL1, KIR3DS1, KIR3DL2, KIR3DL3, KIR2DP1, and KIR3DP1.
  • the KIR transmembrane domain is KIRS2 or KIR2DS2.
  • the transmembrane domain of the second fusion peptide is the DAP12 transmembrane domain.
  • the cytoplasmic domain is selected from the cytoplasmic domains of DAP12 and KIR.
  • the antigen binding domain comprises an antibody or antigen binding fragment thereof, preferably the antigen binding domain comprises the heavy chain CDR1, CDR2 and CDR3 of the antibody and the CDR1, CDR2 and CDR3 of the light chain; preferably Preferably, the antigen binding domain comprises a heavy chain variable region and a light chain variable region of an antibody; preferably, the antigen binding domain comprises Fab, Fab', F(ab') 2 , single-chain Fv (scFv ), Fv, dsFv, diabodies, Fd and Fd' fragments
  • the antigen is a tumor-associated antigen.
  • the tumor-associated antigen is selected from the group consisting of: CD19, mesothelin, GD-2, CD20, CD22, CD30, CD33, CD38, CD123, CD138, CEA, CTLA4, BCMA, CS1, c-Met, EPCAM, EGFR/EGFRvIII, gp100, GPC3, IGF1R, IGF-I receptor, MAGE A3, B7-H3, MUC1, NY-ESO-1, HER2, PD1, PSMA, ROR1, WT1, glycolipid F77 or any other tumor antigen or other modifier types and any combination thereof.
  • the tumor-associated antigen is selected from the group consisting of: CD19, mesothelin, and GD-2.
  • the costimulatory domain is selected from the group consisting of 4-1BB, CD28, CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, LFA-1, CD2, CD7, LIGHT, NKG2C, B7-H3. In some preferred embodiments, the costimulatory domain is 4-1BB.
  • the present disclosure provides nucleic acids encoding chimeric antigen receptors as previously described.
  • the present disclosure provides vectors comprising the aforementioned nucleic acids.
  • the present disclosure provides cells comprising the aforementioned vectors.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising the aforementioned chimeric antigen receptor or nucleic acid encoding the same and a pharmaceutically acceptable carrier.
  • the present disclosure provides a cell preparation method by introducing the aforementioned nucleic acid or vector into immune effector cells.
  • the present disclosure provides use of the aforementioned chimeric antigen receptor, nucleic acid, vector, cell and/or pharmaceutical composition in the manufacture of a medicament for the treatment and/or prevention of a disease or disorder.
  • the present disclosure provides a method of providing anti-tumor immunity in a mammal, comprising administering to the mammal an effective amount of a chimeric antigen receptor, nucleic acid, vector and/or cell comprising the foregoing.
  • the present disclosure provides a method of treating a mammal having a disease or disorder, comprising administering to the mammal an effective amount of the aforementioned chimeric antigen receptors, nucleic acids, vectors, and/or cells.
  • Figure 1 shows the structural design of a DPK CAR targeting CD19.
  • Figure 2 shows the expansion and volume changes of four CAR-T cells targeting CD19.
  • Figure 3 shows the positive rates of four CAR-Ts targeting CD19.
  • Figure 4 shows the detection results of four CAR-T cell differentiation subtypes targeting CD19.
  • Figure 5 shows the lysis rate of target cells by CAR-T cells targeting CD19 and non-transduced T cells (NTD).
  • Figure 6 shows the results of IL-2 and IFN- ⁇ assays for four CAR-T cells targeting CD19.
  • Figure 7 shows the results of CAR-T proliferation flow assay targeting CD19.
  • Figure 8 shows the trend of tumor changes in mice in different CAR-T administration groups targeting CD19.
  • Figure 9 shows the secretion of IL-6 and IL-10 in the blood of mice 14 days after injection of CAR-T targeting CD19.
  • Figure 10 shows CD19 CAR-T occupancy in blood T lymphocytes.
  • Figure 11 shows the content of PD-1 in CAR-T lymphocytes targeting CD19.
  • Figure 12 shows a flow chart of the clinical treatment regimen of CAR-T targeting CD19.
  • M1, M3, and M6 represent 1 month, 3 months, and 6 months; ** represents an important time node for evaluating the efficacy.
  • Figure 13 shows the structural design of mesothelin-DAP12/KIRS2 CAR targeting mesothelin.
  • Figure 14 shows expansion and volume changes of two CAR-T cells targeting mesothelin.
  • Figure 15 shows the positive rate of two CAR-T cells and NTD cells targeting mesothelin.
  • Figure 16 shows the results of two CAR-T cell and NTD cell differentiation subtype assays targeting mesothelin.
  • Figure 17 shows the lysis rate of target cells by two CAR-T cells targeting mesothelin and NTD cells.
  • Figure 18 shows the results of IL-2 and IFN- ⁇ assays for two CAR-T cells targeting mesothelin and NTD cells.
  • Figure 19 shows the results of flow cytometry of the proliferation of two CAR-T cells and NTD cells targeting mesothelin.
  • Figure 20 shows the trend of tumor changes in mice in different CAR-T administration groups targeting mesothelin.
  • Figure 21 shows a flow chart of the clinical treatment protocol of CAR-T cells targeting mesothelin.
  • Figure 22 shows solid tumor survival statistics for mesothelin-targeted SS1-CAR-T therapy.
  • Figure 23 shows solid tumor progression-free survival statistics for mesothelin-targeted SS1-CAR-T therapy.
  • Figure 24 shows the structural design of a GD2-DAP12/KIRS2 CAR targeting GD2.
  • Figure 25 shows the expansion and volume changes of two CAR-T cells targeting GD2.
  • Figure 26 shows the positive rate of two CAR-T targeting GD2.
  • Figure 27 shows the results of two CAR-T cell differentiation subtype assays targeting GD2.
  • Figure 28 shows the lysis rate of target cells by CAR-T cells targeting GD2 and NTDs.
  • Figure 29 shows the results of IL-2 and IFN- ⁇ assays for two CAR-T cells targeting GD2.
  • Figure 30 shows the results of GD2-targeted CAR-T proliferation flow assay.
  • the term “about” or “approximately” means within plus or minus 10% of the given value or range. Where a whole number is required, the term refers to within plus or minus 10% of the given value or range, rounded up or down to the nearest whole number.
  • CAR Chimeric Antigen Receptor
  • the CAR refers to a chimeric polypeptide that shares structural and functional properties with cellular immune function receptors or adaptor molecules from, eg, T cells or NK cells.
  • the CAR comprises an antigen binding domain that binds to a cognate antigen (eg, a tumor antigen described herein). After binding to the cognate antigen, CAR can activate or inactivate the cytotoxic cells in which it is located, or modulate the antitumor activity of cells or modulate the immune response of cells.
  • antibody refers to a protein or polypeptide sequence derived from an immunoglobulin molecule that specifically binds to a target antigen.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources, and can be immunoreactive portions of intact immunoglobulins.
  • Antibodies can be polyclonal or monoclonal, multi-chain or single-chain or intact immunoglobulins, and can be derived from natural sources or from recombinant sources.
  • Antibodies are usually tetramers of immunoglobulin molecules.
  • Antibody molecules described herein can exist in a variety of forms in which the antigen-binding portion of the antibody is expressed as part of a contiguous polypeptide chain, including, for example, single-domain antibody fragments (sdAbs), single-chain antibodies (scFvs), and humanized or human antibodies , for example, as described herein.
  • sdAbs single-domain antibody fragments
  • scFvs single-chain antibodies
  • humanized or human antibodies for example, as described herein.
  • the phrase "substantially identical" can be understood as exhibiting at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, Antibody chains of 97%, 98%, 99% or more sequence identity.
  • nucleic acid sequences the term is to be understood as exhibiting at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98 A nucleotide sequence of %, 99% or greater sequence identity.
  • sequence identity has an art-recognized meaning, and the percent sequence identity between two nucleic acid or polypeptide molecules or regions can be calculated using published techniques. Sequence identity can be measured along the full length of a polynucleotide or polypeptide or along regions of the molecule. While many methods exist for measuring the identity between two polynucleotides or polypeptides, the term “identity” is well known to the skilled artisan (Carrillo, H. & Lipman, D., SIAM J Applied Math 48:1073 (1988) ).
  • substitutional variant is one in which at least one amino acid residue in the native sequence has been removed and a different amino acid inserted in its same position.
  • the substitutions can be single, wherein only one amino acid is substituted in the molecule, or multiple, wherein the same molecule has two or more amino acids substituted. Multiple substitutions can be made at consecutive sites.
  • one amino acid may be substituted by multiple residues, wherein such variants include both substitutions and insertions.
  • An “insertional” variant is one in which one or more amino acids are inserted into an amino acid immediately adjacent to a specific position in a native sequence. Immediately adjacent to an amino acid means attachment to the alpha-carboxyl or alpha-amino functional group of the amino acid.
  • a “deletion” variant is one in which one or more amino acids in the native amino acid sequence have been removed. Typically, deletion variants have one or two amino acids deleted in a specific region of their molecule.
  • variable domains of antibodies refers to certain portions of related molecules that differ widely in sequence between antibodies and are used for the specific recognition and binding of a particular antibody against its specific target. However, the variability is not evenly distributed throughout the variable domains of antibodies. Variability is concentrated in three segments called complementarity determining regions (CDRs; ie CDR1, CDR2 and CDR3) or hypervariable regions, all located within the variable domains of light and heavy chains. The more conserved portions of the variable domains are referred to as framework (FR) regions or framework sequences.
  • CDRs complementarity determining regions
  • FR framework regions
  • Each variable domain of native heavy and light chains includes four FR regions, predominantly in a beta-sheet configuration, linked by three CDRs that form loops that connect the beta-sheet structure and Partial ⁇ -sheet structures are formed in some cases.
  • the CDRs of each chain are usually linked in proximity by FR regions and, with the aid of CDRs from other chains, contribute to the formation of antibody target binding sites (epitopes or determinants).
  • the numbering of immunoglobulin amino acid residues is according to the immunoglobulin amino acid residue numbering system of Kabat et al., unless otherwise indicated.
  • a CDR can have the ability to specifically bind to the cognate epitope.
  • antibody fragment or "antigen-binding fragment” of an antibody refers to any portion of a full-length antibody that is less than full-length, but which comprises at least a portion of the variable region (e.g., one or more CDRs and/or the variable region of said antibody that binds an antigen) or one or more antibody binding sites), and thus retain the binding specificity and at least part of the specific binding capacity of the full-length antibody.
  • an antigen-binding fragment refers to an antibody fragment comprising an antigen-binding portion that binds to the same antigen as the antibody from which the antibody fragment is derived.
  • Antibody fragments include antibody derivatives produced by enzymatic treatment of full-length antibodies, as well as synthetically produced derivatives, eg, recombinantly produced derivatives.
  • Antibodies include antibody fragments. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab') 2 , single-chain Fv (scFv), Fv, dsFv, diabodies, Fd and Fd' fragments, and other fragments, including modified fragments (see, For example, Methods in Molecular Biology, Vol 207: Recombinant Antibodies for Cancer Therapy Methods and Protocols (2003); Chapter 1; p 3-25, Kipriyanov).
  • the fragments may comprise multiple chains linked together, eg, by disulfide bonds and/or by peptide linkers.
  • Antibody fragments generally comprise at least or about 50 amino acids, and typically at least or about 200 amino acids.
  • Antigen-binding fragments include any antibody fragment that, when inserted into the antibody framework (eg, by substituting the corresponding region), results in an antibody that immunospecifically binds (ie, exhibits a Ka of at least or at least about 107-108 M- 1 ) to an antigen .
  • a "functional fragment” is a fragment or analog that prevents or substantially reduces the ability of the receptor to bind a ligand or initiate signal transduction.
  • functional fragments generally have the same meaning as "antibody fragments” and, in the case of antibodies, may refer to fragments that prevent or substantially reduce the ability of the receptor to bind a ligand or initiate signal transduction, eg, Fv, Fab , F(ab') 2 , and so on.
  • "Fv" fragments consist of a dimer ( VH - VL dimer) formed by non-covalent association of the variable domains of a heavy chain and the variable domains of a light chain. In this configuration, the three CDRs of each variable domain interact to define the target binding site on the surface of the VH - VL dimer, as is the case with intact antibodies. The six CDRs collectively confer the target-binding specificity of the intact antibody. However, even a single variable domain (or half of an Fv that includes only 3 target-specific CDRs) can still have the ability to recognize and bind targets.
  • monoclonal antibody refers to a population of identical antibodies, meaning that each individual antibody molecule in the monoclonal antibody population is identical to other antibody molecules. This property is in contrast to that of polyclonal populations of antibodies, which comprise antibodies with a variety of different sequences.
  • Monoclonal antibodies can be prepared by a number of well-known methods (Smith et al. (2004) J. Clin. Pathol. 57, 912-917; and Nelson et al., J Clin Pathol (2000), 53, 111-117).
  • monoclonal antibodies can be prepared by immortalizing B cells, eg, by fusion with myeloma cells to generate hybridoma cell lines or by infecting B cells with a virus such as EBV.
  • Recombinant techniques can also be used to prepare antibodies from clonal populations of host cells in vitro by transforming the host cells with a plasmid carrying an artificial sequence of nucleotides encoding the antibody.
  • full-length antibody is an antibody having two full-length heavy chains (eg VH-CH1-CH2-CH3 or VH-CH1-CH2-CH3-CH4) and two full-length light chains (VL-CL) and a hinge region,
  • VH-CH1-CH2-CH3 or VH-CH1-CH2-CH3-CH4 full-length heavy chains
  • VL-CL full-length light chains
  • chimeric antibody refers to an antibody in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody of antibodies.
  • Humanized antibodies refer to non-human (eg, mouse) forms of antibodies that are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (eg, Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) containing minimal sequence derived from non-human immunoglobulins.
  • the humanized antibody is a human immunoglobulin (recipient antibody) in which the complementarity determining region (CDR) residues of the recipient antibody are derived from a non-human species with the desired specificity, affinity and capacity ( donor antibody) such as mouse, rat or rabbit CDR residue substitutions.
  • CDR complementarity determining region
  • telomeres can be mutated amino acid residues within the CDR1, CDR2 and/or CDR3 regions of VH and/or VL, thereby improving one or more binding properties (eg, affinity) of the antibody .
  • PCR-mediated mutagenesis can be performed to introduce mutations whose effect on antibody binding or other functional properties can be assessed using the in vitro or in vivo assays described herein. Typically, conservative mutations are introduced. Such mutations can be amino acid substitutions, additions or deletions.
  • CDR refers to complementarity-determining regions, and each of the heavy and light chains of antibody molecules is known to have 3 CDRs.
  • the CDRs are also referred to as hypervariable regions, and are present in the variable regions of each heavy and light chain of antibodies, with sites of very high variability in the primary structure of the CDRs.
  • the CDRs of the heavy chain are represented by CDR1, CDR2, and CDR3 derived from the amino terminal of the amino terminal sequence of the heavy chain
  • CDRs of the light chain are represented by CDR1, CDR2, and CDR3 derived from the amino terminal of the amino terminal sequence of the light chain.
  • epitopic determinants refers to any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants typically comprise chemically active surface profiles of molecules, such as amino acids or sugar side chains, and typically have specific three-dimensional structural characteristics as well as specific charge characteristics.
  • the terms “specifically binds” or “immunospecifically binds” with respect to an antibody or antigen-binding fragment thereof are used interchangeably herein and refer to an antibody or antigen-binding fragment through non-co-coding between the antibody-binding sites of the antibody and the antigen. The ability to form one or more non-covalent bonds with alloantigens.
  • the antigen may be an isolated antigen or present in tumor cells.
  • an antibody that immunospecifically binds (or specifically binds) an antigen has an affinity constant Ka of about or 1x107 M -1 or 1x108 M -1 or greater (or 1x10-7 M or 1x A dissociation constant (Kd) of 10 ⁇ 8 M or lower binds the antigen.
  • Affinity constants can be determined by standard kinetic methods of antibody responses, eg, immunoassays, surface plasmon resonance (SPR) (Rich and Myszka (2000) Curr. Opin. Biotechnol 11:54; Englebienne (1998) Analyst. 123: 1599), isothermal titration calorimetry (ITC), or other kinetic interaction assays known in the art (see, e.g., Paul, ed., Fundamental Immunology, 2nd ed., Raven Press, New York, pages 332-336 (1989); see also US Pat. No. 7,229,619 describing exemplary SPR and ITC methods for calculating the binding affinity of antibodies).
  • SPR surface plasmon resonance
  • ITC isothermal titration calorimetry
  • nucleic acid molecules refer to oligomers or polymers comprising at least two linked nucleotides or nucleotide derivatives, including deoxyribonucleic acid ( DNA) and ribonucleic acid (RNA).
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • nucleic acid molecule is intended to include DNA molecules and RNA molecules. Nucleic acid molecules can be single-stranded or double-stranded, and can be cDNA.
  • an isolated nucleic acid molecule is one that is separated from other nucleic acid molecules present in the natural source of the nucleic acid molecule.
  • An "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material or culture medium when prepared by recombinant techniques, or substantially free of chemical precursors or other chemical components when chemically synthesized.
  • Exemplary isolated nucleic acid molecules provided herein include isolated nucleic acid molecules encoding the provided antibodies or antigen-binding fragments.
  • operably linked in reference to nucleic acid sequences, regions, elements or domains means that the nucleic acid regions are functionally related to each other.
  • a promoter can be operably linked to a nucleic acid encoding a polypeptide such that the promoter regulates or mediates transcription of the nucleic acid.
  • conservative sequence modifications of the sequences described in the Sequence Listing described herein, ie, nucleotide and amino acid sequence modifications that do not eliminate binding of the antibody to the antigen encoded by the nucleotide sequence or containing the amino acid sequence.
  • conservative sequence modifications include conservative nucleotide and amino acid substitutions and nucleotide and amino acid additions and deletions.
  • modifications can be introduced into the Sequence Listing described herein by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • Conservative sequence modifications include conservative amino acid substitutions in which amino acid residues are replaced with amino acid residues having similar side chains. Families of amino acid residues with similar side chains are defined in the art.
  • amino acids with basic side chains eg, lysine, arginine, histidine
  • amino acids with acidic side chains eg, aspartic acid, glutamic acid
  • amino acids with uncharged polar side chains amino acids e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • amino acids with non-polar side chains e.g. alanine, valine
  • leucine, isoleucine, proline, phenylalanine, methionine amino acids with beta branched side chains
  • expression refers to the process by which a polypeptide is produced by transcription and translation of a polynucleotide. Expression levels of a polypeptide can be assessed using any method known in the art, including, for example, methods that determine the amount of polypeptide produced from a host cell. Such methods may include, but are not limited to, quantification of polypeptides in cell lysates by ELISA, Coomassie blue staining followed by gel electrophoresis, Lowry protein assay, and Bradford protein assay.
  • host cell is a cell used to receive, maintain, replicate and amplify a vector. Host cells can also be used to express the polypeptide encoded by the vector. When the host cell divides, the nucleic acid contained in the vector replicates, thereby amplifying the nucleic acid.
  • Host cells can be eukaryotic cells or prokaryotic cells. Suitable host cells include, but are not limited to, CHO cells, various COS cells, HeLa cells, HEK cells such as HEK 293 cells.
  • vector is a replicable nucleic acid from which one or more heterologous proteins can be expressed when transformed into an appropriate host cell.
  • References to vectors include those into which nucleic acids encoding polypeptides or fragments thereof can be introduced, typically by restriction digestion and ligation. References to vectors also include those that contain nucleic acid encoding a polypeptide. Vectors are used to introduce nucleic acid encoding a polypeptide into a host cell, to amplify the nucleic acid, or to express/display the polypeptide encoded by the nucleic acid. Vectors generally remain episomal, but can be designed to integrate the gene or portion thereof into the chromosome of the genome. Also contemplated are artificial chromosome vectors, such as yeast artificial vectors and mammalian artificial chromosomes. The selection and use of such vehicles is well known to those skilled in the art.
  • a vector also includes a "viral vector” or “viral vector.”
  • a viral vector is an engineered virus that is operably linked to a foreign gene to transfer (either as a vehicle or shuttle) the foreign gene into a cell.
  • expression vector includes vectors capable of expressing DNA operably linked to regulatory sequences, such as promoter regions, capable of affecting the expression of such DNA fragments. Such additional fragments may include promoter and terminator sequences, and optionally, one or more origins of replication, one or more selectable markers, enhancers, polyadenylation signals, and the like. Expression vectors are typically derived from plasmid or viral DNA, or may contain elements of both. Thus, an expression vector refers to a recombinant DNA or RNA construct, such as a plasmid, phage, recombinant virus, or other vector, which, when introduced into an appropriate host cell, results in the expression of cloned DNA. Appropriate expression vectors are well known to those skilled in the art and include those that are replicable in eukaryotic and/or prokaryotic cells as well as those that remain episomal or that integrate into the host cell genome.
  • stimulation refers to mediating a primary response to a signaling event by stimulating the binding of a molecule (eg, the TCR/CD3 complex) to its cognate ligand, such as, but not limited to, via the TCR/CD3 complex signal transduction.
  • a molecule eg, the TCR/CD3 complex
  • Stimulation can mediate altered expression of certain molecules, such as downregulation of TGF-beta, and/or reorganization of cytoskeletal structures, among others.
  • the term "stimulatory molecule” refers to a molecule expressed by a T cell that provides a primary cytoplasmic signaling sequence that modulates, in a stimulatory manner, the primary level of the TCR complex for at least some aspects of the T cell signaling pathway activation.
  • the primary signal is initiated by, eg, binding of the TCR/CD3 complex to the peptide-loaded MHC molecule, and it results in the mediation of T cell responses including, but not limited to, proliferation, activation, differentiation, and the like.
  • Primary cytoplasmic signaling sequences also referred to as "primary signaling domains” that act in a stimulatory manner may contain signaling motifs known as immunoreceptor tyrosine-based activation motifs or ITAMs.
  • ITAM-containing primary cytoplasmic signaling sequences particularly useful in the present disclosure include, but are not limited to, those derived from TCR ⁇ , FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, CD278 (also known as "ICOS"), Fc ⁇ RI, CD66d, DAP10 and DAP12.
  • the intracellular signaling domain in any one or more of the CARs of the present disclosure includes an intracellular signaling sequence.
  • APC antigen presenting cell
  • MHC major histocompatibility complex
  • T-cells can recognize these complexes using their T-cell receptors (TCRs).
  • TCRs T-cell receptors
  • Intracellular signaling domain refers to the intracellular portion of a molecule. Intracellular signaling domains can generate signals that promote the immune effector function of CAR-containing cells (eg, CAR-T cells or CAR-expressing NK cells). Examples of immune effector functions (eg, in CAR-T cells or CAR-expressing NK cells) include cytolytic activity and helper activity, including secretion of cytokines. In embodiments, intracellular signaling domains transduce effector function signals and direct cells to perform specialized functions. While the entire intracellular signaling domain can be used, in many cases it is not necessary to use the entire chain.
  • intracellular signaling domain In the case of using truncated portions of intracellular signaling domains, such truncated portions can be used in place of the full chain, so long as it transduces effector function signals.
  • the term intracellular signaling domain is thus intended to include any truncated portion of the intracellular signaling domain sufficient to signal effector function.
  • the intracellular signaling domain may comprise a primary intracellular signaling domain.
  • Exemplary primary intracellular signaling domains include those derived from molecules responsible for primary or antigen-dependent stimulation.
  • the intracellular signaling domain may comprise a costimulatory intracellular domain.
  • Exemplary costimulatory intracellular signaling domains include those derived from molecules responsible for costimulatory signaling or antigen-independent stimulation.
  • the primary intracellular signaling domain may comprise the cytoplasmic sequence of the T cell receptor and co-stimulate intracellular signaling Domains may comprise cytoplasmic sequences from co-receptors or co-stimulatory molecules.
  • the primary intracellular signaling domain may include a signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM.
  • ITAM-containing primary cytoplasmic signaling sequences include, but are not limited to, those derived from CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD5, CD22, CD79a, CD79b, CD278 ("ICOS"), Fc ⁇ RI, CD66d, DAP10 and DAP12.
  • costimulatory molecule refers to a cognate binding partner on a T cell that specifically binds to a costimulatory ligand to mediate a costimulatory response of the T cell, such as, but not limited to, proliferation.
  • Costimulatory molecules are cell surface molecules other than antigen receptors or their ligands that are required for an effective immune response.
  • Costimulatory molecules include but are not limited to MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activation molecules (SLAM proteins), activated NK cell receptors, BTLA , Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM -1, ICOS(CD278), GITR, BAFFR, LIGHT, HVEM(LIGHTR), KIRDS2, SLAMF7, NKp80(KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8 ⁇ , CD8 ⁇ , IL2R ⁇ , IL2R ⁇ , IL7R ⁇ , ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49
  • a costimulatory intracellular signaling domain refers to the intracellular portion of a costimulatory molecule.
  • the intracellular signaling domain may include the entire intracellular portion or the entire native intracellular signaling domain of the molecule, or a functional fragment thereof.
  • 4-1BB refers to a member of the TNFR superfamily having an amino acid sequence as provided in GenBank Acc. No. AAA62478.2, or equivalent residues from non-human species such as mouse, rodent, monkey, ape, etc.
  • 4-1BB costimulatory domain is defined as amino acid residues 214-255 of GenBank Acc.No..AAA62478.2, or equivalents from non-human species such as mouse, rodent, monkey, ape, etc. Residues.
  • a "4-1BB costimulatory domain” is a sequence as provided in SEQ ID NO. 5 or equivalent residues from a non-human species such as mouse, rodent, monkey, ape, etc.
  • immune effector cell refers to a cell that is involved in an immune response, eg, promotes an immune effector response.
  • immune effector cells include T cells, eg, alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells, and myeloid-derived phagocytes.
  • immune effector function or immune effector response refers to an immune effector cell, eg, a function or response that enhances or facilitates the immune attack of a target cell.
  • immune effector function or response refers to the properties of T cells or NK cells that promote killing of target cells or inhibit growth or proliferation.
  • T cells primary stimulation and costimulation are examples of immune effector functions or responses.
  • a population of cells comprises, eg, T cells or populations of T cells at various stages of differentiation. Stages of T cell differentiation ranging from least to most differentiated include naive T cells, stem central memory T cells, central memory T cells, effector memory T cells, and terminal effector T cells. Following antigen exposure, naive T cells proliferate and differentiate into memory T cells, such as stem central memory T cells and central memory T cells, and then differentiate into effector memory T cells. Memory T cells further differentiate into terminal effector T cells after receiving appropriate T cell receptors, co-stimulatory and inflammatory signals.
  • Naive T cells are characterized by the expression pattern of the following cell surface markers: CCR7+, CD62L+, CD45RO-, CD95-.
  • Stem cell central memory T cells are characterized by the expression pattern of the following cell surface markers: CCR7+, CD62L+, CD45RO-, CD95+.
  • Central memory T cells are characterized by the following expression pattern of cell surface markers: CCR7+, CD62L+, CD45RO+, CD95+.
  • Effector memory T cells were characterized by the expression pattern of the following cell surface markers: CCR7-, CD62L-, CD45RO+, CD95+.
  • Terminal effector T cells are characterized by the following expression pattern of cell surface markers: CCR7-, CD62L-, CD45RO-, CD95+.
  • Fresh T cells from healthy donors are typically divided into four subgroups based on CD45RA and CD62L expression: 1) naive T cells (CD45RA+CD62L+, termed Tn), 2) central memory T cells (CD45RA-CD62L+, termed Tcm) ), 3) effector memory T cells (CD45RA-CD62L-, referred to as Tem) and 4) CD45RA positive effector T cells (CD45RA+CD62L-, referred to as Temra).
  • Tn naive T cells
  • CD45RA-CD62L+ termed Tcm
  • Tem effector memory T cells
  • Temra CD45RA positive effector T cells
  • T cell subsets were positive for CD95, while only a small fraction of Tn expressed CD95 (3.6 ⁇ 1.4% in CD4+ and 3.7 ⁇ 1.3% in CD8+ T cells). This small population also co-expresses CD27, CD28 and CCR7 and is considered memory stem cells (Tscm).
  • effector function refers to a specialized function of a cell.
  • the effector function of T cells can be cytolytic activity or helper activity, including secretion of cytokines.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable pharmaceutically acceptable carriers are described in the latest edition of Remington's Pharmaceutical Sciences, a standard bibliography in the art, which is incorporated herein by reference. Preferred examples of such carriers or diluents include, but are not limited to, water, saline, Ringer's solution, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles, such as fixed oils, can also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. In addition to any conventional media or agent incompatibility with the chimeric antigen receptor, its use in the composition is envisaged.
  • treatment includes prevention, treatment and/or cure. Prevention refers to preventing an underlying disease and/or preventing the worsening of symptoms or the development of a disease. Treatment also includes any of the chimeric antigen receptors provided and any pharmaceutical uses of the compositions provided herein.
  • efficacy refers to an effect resulting from a treatment in an individual that alters, generally ameliorates or ameliorates the symptoms of a disease or condition, or cures the disease or condition.
  • terapéuticaally effective amount refers to an amount of a substance, compound, material or composition comprising a compound that is at least sufficient to produce a therapeutic effect after administration to a subject. Thus, it is an amount necessary to prevent, cure, ameliorate, block or partially block the symptoms of the disease or disorder.
  • prophylactically effective amount refers to an amount of a substance, compound, material or composition comprising a compound that, when administered to a subject, will have the desired prophylactic effect, eg, preventing or delaying the onset of a disease or symptom or Relapse, reducing the likelihood of disease or symptoms developing or recurring.
  • a fully prophylactically effective dose need not occur by administering one dose, and may occur only after administering a series of doses. Thus, a prophylactically effective amount can be administered in one or more administrations.
  • patient refers to a mammal, such as a human.
  • the present disclosure provides a chimeric antigen receptor comprising a first fusion peptide and a second fusion peptide, wherein:
  • the first fusion peptide comprises an antigen binding domain and a transmembrane domain
  • the second fusion peptide comprises a transmembrane domain, a cytoplasmic domain and a costimulatory domain.
  • the transmembrane domain of the second fusion peptide interacts with the transmembrane domain of the first fusion peptide through charge interactions, or the second fusion peptide interacts with phosphorylated ITAM within the cytoplasmic domain
  • the sequence interacts with the signaling molecule.
  • the transmembrane domain of the first fusion peptide is a KIR transmembrane domain; preferably, the KIR transmembrane domain is selected from the group consisting of KIR2DS2, KIR2DL3, KIR2DL1, KIR2DL2, KIR2DL4, KIR2DL5A, KIR2DL5B, KIR2DS1, KIR2DS3, KIR2DS4, KIR2DS5, KIR3DL1, KIR3DS1, KIR3DL2, KIR3DL3, KIR2DP1 and KIR3DP1; more preferably, the KIR transmembrane domain is KIRS2 or KIR2DS2; preferably, the KIRS2 comprises the An amino acid sequence showing that the amino acid sequence has 80% or more identity, preferably an amino acid sequence with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, more preferably 98% or 99% More preferably, the amino acid sequence of the KIRS2 is as shown in SEQ ID NO
  • the transmembrane domain of the second fusion peptide is a DAP12 transmembrane domain; preferably, the transmembrane domain of DAP12 comprises 80% or more of the amino acid sequence shown in SEQ ID NO. Amino acid sequences with the above identity, preferably amino acid sequences with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, more preferably amino acid sequences with 98% or more identity ; More preferably, the amino acid sequence of the transmembrane domain of the DAP12 is shown in SEQ ID NO.14.
  • the first fusion peptide comprises a signal peptide
  • the signal peptide is a CD8 ⁇ signal peptide
  • the CD8 ⁇ signal peptide comprises 80% or more identical to the amino acid sequence shown in SEQ ID NO. 4 amino acid sequence, preferably an amino acid sequence with more than 85%, 90%, 95%, 96%, 97%, 98%, 99% identity, more preferably an amino acid sequence with 98% or more identity; more
  • the amino acid sequence of the CD8 ⁇ signal peptide is shown in SEQ ID NO.4.
  • the second fusion peptide comprises a signal peptide
  • the signal peptide is a DAP12 signal peptide or a CD8 ⁇ signal peptide
  • the DAP12 signal peptide comprises an amino acid sequence having 80°C with the amino acid sequence shown in SEQ ID NO.1 % or more identical amino acid sequences, preferably amino acid sequences with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identities, more preferably 98% or more.
  • the amino acid sequence of the DAP12 signal peptide is shown in SEQ ID NO.1; preferably, the CD8 ⁇ signal peptide comprises 80% or more identity with the amino acid sequence shown in SEQ ID NO.4 amino acid sequence, preferably amino acid sequence with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, more preferably amino acid sequence with 98% or more identity; more preferably Typically, the amino acid sequence of the CD8 ⁇ signal peptide is shown in SEQ ID NO.4.
  • the costimulatory domain is selected from the group consisting of 4-1BB, CD28, CD27, CD28, 4-1BB, OX40, CD30, CD40, PD-1, ICOS, LFA-1, CD2, CD7, LIGHT, NKG2C, B7-H3.
  • the costimulatory domain is 4-1BB; preferably, the 4-1BB comprises an amino acid sequence that is 80% or more identical to the amino acid sequence shown in SEQ ID NO. 5, preferably 85 %, 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the 4-1BB The amino acid sequence of is shown in SEQ ID NO.5.
  • the cytoplasmic domain is selected from the cytoplasmic domains of DAP12 and KIR.
  • the cytoplasmic domain is the cytoplasmic domain of DAP12; preferably, the cytoplasmic domain of DAP12 comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO.15 Amino acid sequence, preferably amino acid sequence with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, more preferably amino acid sequence with 98% or more identity; more preferably , the amino acid sequence of the cytoplasmic domain of the DAP12 is shown in SEQ ID NO.15.
  • the antigen binding domain comprises an antibody or antigen binding fragment thereof, preferably the antigen binding domain comprises the heavy chain CDR1, CDR2 and CDR3 of the antibody and the CDR1, CDR2 and CDR3 of the light chain; preferably Preferably, the antigen binding domain comprises a heavy chain variable region and a light chain variable region of an antibody; preferably, the antigen binding domain comprises Fab, Fab', F(ab') 2 , single-chain Fv (scFv ), Fv, dsFv, diabodies, Fd and Fd' fragments.
  • the antigen is a tumor-associated antigen.
  • the tumor-associated antigen is selected from the group consisting of: CD19, mesothelin, GD-2, CD20, CD22, CD30, CD33, CD38, CD123, CD138, CEA, CTLA4, BCMA, CS1, c-Met, EPCAM, EGFR/EGFRvIII, gp100, GPC3, IGF1R, IGF-I receptor, MAGE A3, B7-H3, MUC1, NY-ESO-1, HER2, PD1, PSMA, ROR1, WT1, glycolipid F77 or any other tumor antigen or other modifier types and any combination thereof.
  • the tumor-associated antigen is selected from the group consisting of CD19, mesothelin, and GD-2
  • the antigen binding domain is selected from the group consisting of CD19 antibody or antigen-binding fragment thereof, mesothelin antibody or antigen-binding fragment thereof, and GD2 antibody or antigen-binding fragment thereof.
  • the CD19 antibody or antigen-binding fragment thereof comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 7, preferably 85%, 90%, 95%, 96% , 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the CD19 scFv is shown in SEQ ID NO.7 .
  • the mesothelin antibody or antigen-binding fragment thereof comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 19 or SEQ ID NO. 20, preferably 85%, An amino acid sequence of 90%, 95%, 96%, 97%, 98%, 99% or more identity, more preferably an amino acid sequence of 98% or more identity; more preferably, the amino acid sequence of the SS1 scFv As shown in SEQ ID NO.19 or SEQ ID NO.20.
  • the GD2 antibody or antigen-binding fragment thereof comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 25, preferably 85%, 90%, 95%, 96% , 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or 99% or more identical amino acid sequences; more preferably, the amino acid sequence of the GD2 scFv is shown in SEQ ID NO.25 .
  • the first fusion peptide comprises CD19 scFv and KIRS2, or comprises CD19 scFv and KIR2DS2.
  • the CD19 scFv comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 7, preferably 85%, 90%, 95%, 96%, 97%, 98 %, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the CD19 scFv is shown in SEQ ID NO.7.
  • the amino acid sequence of the KIRS2 is shown in SEQ ID NO.8; preferably, the amino acid sequence of the KIR2DS2 is shown in SEQ ID NO.9.
  • the second fusion peptide comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and a costimulatory domain 4-1BB, or comprises a truncated DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and Costimulatory domain 4-1BB; preferably, the DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise an amino acid sequence having 80% or more identity with the amino acid sequence shown in SEQ ID NO.2, preferably 85% , 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the DAP12 transmembrane structure
  • the amino acid sequence of the domain and the DAP12 cytoplasmic domain is shown in SEQ ID NO.2; preferably, the truncated DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise the amino acid sequence shown in SEQ ID
  • amino acid sequence of preferably 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical
  • amino acid sequence of more preferably, the amino acid sequence of the truncated DAP12 transmembrane domain and DAP12 cytoplasmic domain is shown in SEQ ID NO.3.
  • the chimeric antigen receptor is a CD19-KIRS2/Dap12-BB chimeric antigen receptor
  • the CD19-KIRS2/Dap12-BB chimeric antigen receptor is formed by cleavage of a fusion protein by a T2A peptide
  • the fusion protein comprises DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIRS2;
  • the fusion protein comprises An amino acid sequence with 80% or more identity with the amino acid sequence shown in SEQ ID NO.10, preferably an amino acid sequence with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, More preferably, the amino acid sequence with 98% or more identity; more preferably, the amino acid sequence of the fusion protein is shown in SEQ ID NO.10.
  • the chimeric antigen receptor is a CD19-KIRS2/tDap12-BB chimeric antigen receptor
  • the CD19-KIRS2/tDap12-BB chimeric antigen receptor is formed by cleavage of a fusion protein by a T2A peptide
  • the fusion protein consists of CD8 ⁇ signal peptide, truncated DAP12 (tDap-12, including truncated transmembrane and cytoplasmic domains), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIRS2; preferably, the fusion protein comprises an amino acid sequence with 80% or more identity with the amino acid sequence shown in SEQ ID NO.
  • amino acid sequence of the fusion protein is shown in SEQ ID NO.11.
  • the chimeric antigen receptor is a CD19-KIR2DS2/Dap12-BB chimeric antigen receptor
  • the CD19-KIR2DS2/Dap12-BB chimeric antigen receptor is formed by cleavage of a fusion protein with a T2A peptide
  • the fusion protein consists of DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIR2DS2;
  • the fusion protein comprises An amino acid sequence with 80% or more identity with the amino acid sequence shown in SEQ ID NO.12, preferably an amino acid sequence with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity, More preferably, the amino acid sequence with 98% or more identity; more preferably, the amino acid sequence of the fusion protein is shown in SEQ ID NO.12.
  • the T2A cleavage site comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 6, preferably 85%, 90%, 95%, 96%, 97% , 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the T2A cleavage site is shown in SEQ ID NO.6.
  • the first fusion peptide comprises SS1 scFv and KIRS2, or comprises SS1 scFv and KIR2DS2.
  • the SS1 scFv comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 19 or SEQ ID NO. 20, preferably 85%, 90%, 95%, 96% %, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or 99% or more identical amino acid sequences; more preferably, the amino acid sequence of the SS1 scFv is as shown in SEQ ID NO.19 or shown in SEQ ID NO.20.
  • the amino acid sequence of the KIRS2 is shown in SEQ ID NO.8; preferably, the amino acid sequence of the KIR2DS2 is shown in SEQ ID NO.9.
  • the second fusion peptide comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and a costimulatory domain 4-1BB, or comprises a truncated DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and Costimulatory domain 4-1BB; preferably, the DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise an amino acid sequence having 80% or more identity with the amino acid sequence shown in SEQ ID NO.2, preferably 85% , 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the DAP12 transmembrane structure
  • the amino acid sequence of the domain and the DAP12 cytoplasmic domain is shown in SEQ ID NO.2; preferably, the truncated DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise the amino acid sequence shown in SEQ ID
  • amino acid sequence of preferably 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical
  • amino acid sequence of more preferably, the amino acid sequence of the truncated DAP12 transmembrane domain and DAP12 cytoplasmic domain is shown in SEQ ID NO.3.
  • the chimeric antigen receptor is an SS1 scFv1-KIRS2/Dap12-BB chimeric antigen receptor
  • the SS1 scFv1-KIRS2/Dap12-BB chimeric antigen receptor is cleaved by a fusion protein via a T2A peptide
  • the fusion protein comprises DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, SS1 scFv1 and KIRS2; preferably, the fusion The protein comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO.
  • amino acids with 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity sequence more preferably an amino acid sequence with 98% or more identity; more preferably, the amino acid sequence of the fusion protein is shown in SEQ ID NO.21.
  • the chimeric antigen receptor is an SS1 scFv2-KIRS2/Dap12-BB chimeric antigen receptor
  • the SS1 scFv2-KIRS2/Dap12-BB chimeric antigen receptor is cleaved from a fusion protein by a T2A peptide
  • the fusion protein comprises CD8 ⁇ signal peptide, truncated DAP12 (tDap-12, including truncated transmembrane and cytoplasmic domains), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, SS1 scFv2 and KIRS2;
  • the fusion protein comprises an amino acid sequence with 80% or more identity with the amino acid sequence shown in SEQ ID NO.23, preferably 85%, 90%, 95%, 96%, 97%, 98 %, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid
  • the T2A cleavage site comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 6, preferably 85%, 90%, 95%, 96%, 97% , 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the T2A cleavage site is shown in SEQ ID NO.6.
  • the first fusion peptide comprises GD2 scFv and KIRS2, or comprises GD2 scFv and KIR2DS2.
  • the GD2 scFv comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 25, preferably 85%, 90%, 95%, 96%, 97%, 98% %, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the GD2 scFv is shown in SEQ ID NO.25.
  • the amino acid sequence of the KIRS2 is shown in SEQ ID NO.8; preferably, the amino acid sequence of the KIR2DS2 is shown in SEQ ID NO.9.
  • the second fusion peptide comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and a costimulatory domain 4-1BB, or comprises a truncated DAP12 transmembrane domain, a DAP12 cytoplasmic domain, and Costimulatory domain 4-1BB; preferably, the DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise an amino acid sequence having 80% or more identity with the amino acid sequence shown in SEQ ID NO.2, preferably 85% , 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the DAP12 transmembrane structure
  • the amino acid sequence of the domain and the DAP12 cytoplasmic domain is shown in SEQ ID NO.2; preferably, the truncated DAP12 transmembrane domain and the DAP12 cytoplasmic domain comprise the amino acid sequence shown in SEQ ID
  • amino acid sequence of preferably 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical
  • amino acid sequence of more preferably, the amino acid sequence of the truncated DAP12 transmembrane domain and DAP12 cytoplasmic domain is shown in SEQ ID NO.3.
  • the chimeric antigen receptor is a GD2 scFv-KIRS2/Dap12-BB chimeric antigen receptor
  • the GD2 scFv-KIRS2/Dap12-BB chimeric antigen receptor is cleaved from a fusion protein by a T2A peptide
  • the fusion protein comprises DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, GD2 scFv1 and KIRS2; preferably, the fusion protein Contains an amino acid sequence with 80% or more identity with the amino acid sequence shown in SEQ ID NO. , more preferably an amino acid sequence with 98% or more identity; more preferably, the amino acid sequence of the fusion protein is shown in SEQ ID NO.26.
  • the T2A cleavage site comprises an amino acid sequence with 80% or more identity to the amino acid sequence shown in SEQ ID NO. 6, preferably 85%, 90%, 95%, 96%, 97% , 98%, 99% or more identical amino acid sequences, more preferably 98% or more identical amino acid sequences; more preferably, the amino acid sequence of the T2A cleavage site is shown in SEQ ID NO.6.
  • the present disclosure provides a nucleic acid encoding the aforementioned chimeric antigen receptor; preferably, the nucleic acid comprises SEQ ID NO.16, SEQ ID NO.17, SEQ ID NO.18, SEQ ID NO.22,
  • the nucleotide sequences shown in SEQ ID NO.24, SEQ ID NO.27 and SEQ ID NO.29 have 80% or more identity, preferably 85%, 90%, 95%, 96%, A nucleotide sequence of 97%, 98%, 99% or more identity, more preferably a nucleotide sequence of 98% or more identity;
  • the nucleic acid is selected from: SEQ ID NO.16, SEQ ID NO.16, SEQ ID NO.16, SEQ ID NO.
  • the present disclosure provides vectors comprising the aforementioned nucleic acids.
  • the present disclosure provides cells comprising the aforementioned nucleic acids or vectors.
  • the present disclosure provides a composition comprising the aforementioned chimeric antigen receptor, nucleic acid, vector and/or cell and a pharmaceutically acceptable carrier.
  • the present disclosure provides a method for preparing cells, the method comprising introducing the aforementioned nucleic acid and vector into immune effector cells.
  • the present disclosure provides use of the aforementioned chimeric antigen receptors, nucleic acids, vectors and/or cells in the manufacture of a medicament for the treatment and/or prevention of a disease or disorder.
  • the use is in the manufacture of a medicament for the treatment and/or prevention of a disease or disorder associated with aberrant CD19 expression, activity and/or signaling.
  • the disease or disorder comprises a tumor, preferably, the tumor is a cancer disease; preferably, the cancer disease is selected from the group consisting of: brain cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, Liver cancer, kidney cancer, lymphoma, leukemia, lung cancer, melanoma, metastatic melanoma, mesothelioma, neuroblastoma, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, skin cancer, thymoma, sarcoma, non- Hodgkin lymphoma, Hodgkin lymphoma or uterine cancer and any combination thereof.
  • the chimeric antigen receptor can be used as a therapeutic agent.
  • agents will typically be used to treat, alleviate and/or prevent a disease or pathology associated with aberrant mesothelin (MSLN) expression, activity and/or signaling in a subject.
  • Treatment can be carried out using standard methods by identifying a subject, such as a human patient having (or at risk or developing) a disease or disorder associated with aberrant MSLN expression, activity and/or signaling, such as cancer or other neoplastic disorders Program.
  • An antibody preparation preferably one with high specificity and high affinity for its target antigen, is administered to a subject and will generally have an effect due to its binding to the target.
  • the administered chimeric antigen receptor can abrogate or inhibit or interfere with the expression, activity and/or signaling function of the target (eg, MSLN).
  • the administered chimeric antigen receptor can eliminate or inhibit or prevent the target (eg, MSLN) from binding to the endogenous ligand to which it naturally binds.
  • a chimeric antigen receptor binds to a target and modulates, blocks, inhibits, reduces, antagonizes, neutralizes, or otherwise interferes with MSLN expression, activity, and/or signaling.
  • a chimeric antigen receptor having heavy and light chain CDRs can be administered to a subject.
  • the disease or disorder associated with aberrant MSLN expression may be cancer.
  • diseases or disorders associated with aberrant MSLN expression, activity and/or signaling include solid tumors.
  • the mesothelin expression-related disease is selected from the group consisting of mesothelioma, epithelial-like malignant pleural mesothelioma, ovarian cancer, lung cancer, esophageal cancer, pancreatic cancer, gastric cancer, biliary tract cancer, endometrial cancer, thymic cancer, colon cancer cancer, breast cancer.
  • the chimeric antigen receptor can be used as a therapeutic agent.
  • Such agents will typically be used to treat, alleviate and/or prevent a GD2-related disease or pathology in a subject.
  • Treatment regimens can be implemented using standard methods by identifying a subject, eg, a human patient having (or at risk or developing) a disease or disorder associated with GD2, eg, cancer or other neoplastic disorder.
  • An antibody preparation preferably one with high specificity and high affinity for its target antigen, is administered to a subject and will generally have an effect due to its binding to the target.
  • the administered chimeric antigen receptor can abrogate or inhibit or interfere with the expression, activity and/or signaling function of the target (eg, GD2).
  • the administered chimeric antigen receptor can eliminate or inhibit or prevent the target (eg, GD2) from binding to the endogenous ligand to which it naturally binds.
  • a chimeric antigen receptor binds to a target and modulates, blocks, inhibits, reduces, antagonizes, neutralizes, or otherwise interferes with GD2 expression, activity, and/or signaling.
  • a chimeric antigen receptor having heavy and light chain CDRs can be administered to a subject.
  • the disease or disorder associated with abnormal GD2 may be cancer.
  • a GD2-related disease or disorder is a GD2-positive tumor.
  • a GD2-positive tumor is one that is associated with elevated levels of GD2 expression, including, for example, neuroblastoma, glioblastoma, medulloblastoma, astrocytoma, melanoma, small cell lung cancer , desmoplastic small round cell tumor, osteosarcoma, rhabdomyosarcoma, Ewing's sarcoma or liposarcoma, fibrosarcoma, leiomyosarcoma and other soft tissue sarcomas in adults.
  • the patient has primary refractory or recurrent high-risk neuroblastoma, or minimal residual disease with high-risk neuroblastoma.
  • the patient may have been previously treated or concurrently treated with another therapy such as surgery, chemotherapy, radiation therapy, stem cell transplantation, cytokine therapy (eg, with IL-2 and/or GM-CSF), and/or tretinoin (for example, the use of isotretinoin) to treat.
  • another therapy such as surgery, chemotherapy, radiation therapy, stem cell transplantation, cytokine therapy (eg, with IL-2 and/or GM-CSF), and/or tretinoin ( For example, the use of isotretinoin) to treat.
  • Symptoms associated with cancer and other neoplastic disorders include, for example, inflammation, fever, general malaise, fever, pain, often localized inflammation, loss of appetite, weight loss, swelling, headache, fatigue, rash, anemia, muscle weakness, muscle fatigue and abdominal Symptoms such as abdominal pain, diarrhea or constipation.
  • the present disclosure provides a method of providing anti-tumor immunity in a mammal, comprising administering to the mammal an effective amount of a chimeric antigen receptor, nucleic acid, vector, cell and/or composition comprising the foregoing .
  • the present disclosure provides a method of treating a mammal having a disease or disorder, comprising administering to the mammal an effective amount of the aforementioned chimeric antigen receptor, nucleic acid, vector, cell, and/or composition .
  • compositions of the embodiments are formulated to be compatible with their intended route of administration.
  • routes of administration include parenteral, eg, intravenous, intradermal, subcutaneous, oral (eg, inhalation), transdermal (ie, topical), transmucosal, and rectal.
  • Solutions or suspensions for parenteral, intradermal or subcutaneous administration may include the following components: sterile injectable diluents such as water, saline solutions, fixed oils, polyethylene glycols, glycerol, propylene glycol or other synthetic solvents; Antibacterial agents such as benzyl alcohol or methylparaben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetate, citrate Or phosphate, and agents to adjust osmotic pressure, such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be packaged in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (herein water-soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable pharmaceutically acceptable carriers include physiological saline, bacteriostatic water, Cremophor EL TM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that it is easy to inject. It must be stable under the conditions of manufacture and storage and must be protected against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • Proper fluidity can be maintained, for example, by the use of coatings such as lecithin to maintain the desired particle size in the case of dispersions, and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents such as sugars, polyols (such as mannitol, sorbitol), sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the chimeric antigen receptor in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filter sterilization.
  • dispersions are prepared by incorporating the chimeric antigen receptor into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the methods of preparation are vacuum drying and freeze-drying to obtain a powder containing the active ingredient and any additional desired ingredient from a sterile-filtered solution of those previously enumerated. .
  • the compounds are delivered as an aerosol spray from a pressurized container or dispenser containing a gas of a suitable propellant, such as carbon dioxide, or a nebulizer.
  • a gas of a suitable propellant such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the permeation barrier are used in the formulation.
  • penetrants are generally known in the art and include, for example, detergents, bile salts and fusidic acid derivatives for transmucosal administration.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • one or more of the chimeric antigen receptors can be formulated into an ointment, ointment, gel, or cream as generally known in the art.
  • the compounds can also be prepared for rectal delivery in the form of suppositories (eg, with conventional suppository bases such as cocoa butter or other glycerides) or retention enemas.
  • suppositories eg, with conventional suppository bases such as cocoa butter or other glycerides
  • retention enemas e.g., retention enemas.
  • the chimeric antigen receptor can be prepared with a carrier that will prevent it from being rapidly eliminated by the body, such as a sustained/controlled release formulation, including implants and microencapsulated delivery systems.
  • a sustained/controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparing such formulations will be apparent to those skilled in the art.
  • Dosage unit form refers to physically discrete units suitable as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier one or more of the chimeric antigen receptors.
  • the specifications for the dosage unit forms of the embodiments are indicated by and are directly dependent on the unique characteristics of the chimeric antigen receptor and the particular therapeutic effect to be achieved, and the field of formulation of such chimeric antigen receptors used to treat the individual. inherent limitations.
  • the pharmaceutical composition can be placed in a container, pack, or dispenser with instructions for administration.
  • compositions described herein may also contain more than one such chimeric antigen receptor, preferably those that have complementary activities but do not negatively affect each other, depending on the particular condition to be treated.
  • the composition may, for example, comprise an agent that enhances its function, such as a cytotoxic agent, a cytokine, a chemotherapeutic agent, or a growth inhibitory agent.
  • a cytotoxic agent such as a cytotoxic agent, a cytokine, a chemotherapeutic agent, or a growth inhibitory agent.
  • the production of the cytokine IL-6 is reduced or inhibited. In another embodiment, the production of the cytokine IL-10 is reduced or inhibited.
  • cytokine release syndrome is graded.
  • Level 1 describes a cytokine release syndrome wherein symptoms are not life threatening and only symptomatic treatment is required, eg, fever, nausea, fatigue, headache, myalgia, malaise.
  • Grade 2 symptoms require and respond to moderate intervention, such as oxygen, fluids, or vasopressors for hypotension.
  • grade 3 symptoms require and respond to aggressive intervention.
  • the grade 4 symptoms are life-threatening symptoms requiring a ventilator and the patient exhibits organ toxicity.
  • one or more of the chimeric antigen receptors may be administered in combination therapy, ie, with other agents such as therapeutic agents (which are useful in the treatment of pathological conditions or disorders, such as various forms of cancer, autoimmune disorders and inflammatory diseases).
  • agents such as therapeutic agents (which are useful in the treatment of pathological conditions or disorders, such as various forms of cancer, autoimmune disorders and inflammatory diseases).
  • the term "combination” as used herein refers to the administration of the agents substantially simultaneously, simultaneously or sequentially. If administered sequentially, at the start of administration of the second compound, the first of the two compounds is still preferably detected at an effective concentration at the treatment site.
  • “combination” can also be the simultaneous inclusion of a chimeric antigen receptor of the present disclosure and other therapeutic agents in a kit.
  • a combination therapy can comprise one or more chimeric antigen receptors described herein with one or more additional therapeutic agents (eg, one or more cytokine and growth factor inhibitors, immunosuppressants, anti-inflammatory agents) , metabolic inhibitors, enzyme inhibitors, and/or cytotoxins or cytostatics, as described in more detail below) are co-formulated and/or co-administered.
  • additional therapeutic agents eg, one or more cytokine and growth factor inhibitors, immunosuppressants, anti-inflammatory agents
  • metabolic inhibitors e.g., one or more cytokine and growth factor inhibitors, immunosuppressants, anti-inflammatory agents
  • enzyme inhibitors e.g., enzyme inhibitors, and/or cytotoxins or cytostatics, as described in more detail below
  • the present disclosure uses DAP12 in series with 4-1BB to increase the costimulatory signal molecular elements, and designs a chimeric antigen receptor with better anti-tumor effect.
  • Its anti-tumor beneficial effect has the following four aspects: 1. (2) lower secretion of immunosuppressive factors in vivo (reduced secretion of IL-10); (3) good clinical safety and good efficacy (low-grade CRS, durable remission).
  • the present disclosure increases costimulatory signaling molecular elements through DAP12 in series with 4-1BB, links different anti-CD19, anti-mesothelin or anti-GD2 scFV antibodies, and designs anti-CD19, anti-mesothelin or anti-GD2 expression-positive solid tumors.
  • Chimeric antigen receptors forming a chimeric antigen receptor structure in parallel with a fusion peptide comprising an antigen binding domain and a transmembrane domain and another fusion peptide comprising a transmembrane domain, a cytoplasmic domain and a costimulatory domain.
  • the chimeric antigen receptor has good killing activity on CD19, mesothelin or GD2 positive tumor cells, and can promote the secretion of cytokines (including IL-2 and INF ⁇ ).
  • cytokines including IL-2 and INF ⁇ .
  • the CAR structure disclosed in the present disclosure is expected to greatly improve the therapeutic effect of CAR-T cells in the clinical application of MSLN-positive solid tumors in terms of the low efficacy of CAR-T in the clinical treatment of solid tumors.
  • the CD19-KIRS2/Dap12-BB chimeric antigen receptor comprises a first fusion peptide CD19-KIRS2 and a second fusion peptide Dap12-BB, wherein:
  • the first fusion peptide CD19-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is a CD19 scFv, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide Dap12-BB comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • the CD19-KIRS2/Dap12-BB chimeric antigen receptor is formed by the cleavage of DPK01 protein by T2A peptide, and the DPK01 fusion protein comprises DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4- 1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIRS2, the amino acid sequence of the DPK01 fusion protein is shown in SEQ ID NO.10.
  • DPK01 DAP12 signal peptide+DAP12+4-1BB+T2A+CD8 ⁇ signal peptide+CD19 scFv+KIRS2
  • the CD19-KIRS2/tDap12-BB chimeric antigen receptor comprises a first fusion peptide CD19-KIRS2 and a second fusion peptide tDap12-BB, wherein:
  • the first fusion peptide CD19-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is a CD19 scFv, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide tDap12-BB comprises a truncated DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • the CD19-KIRS2/tDap12-BB chimeric antigen receptor is formed by the cleavage of the DPK02 protein by the T2A peptide, and the DPK02 fusion protein comprises the CD8 ⁇ signal peptide, the truncated DAP12 (tDap12, including the transmembrane domain and the cytoplasmic structure). domain), 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIRS2, the amino acid sequence of the DPK02 fusion protein is shown in SEQ ID NO.11.
  • DPK02 CD8 ⁇ signal peptide+tDap12+4-1BB+T2A+CD8 ⁇ signal peptide+CD19 scFv+KIRS2
  • the CD19-KIR2DS2/Dap12-BB chimeric antigen receptor comprises a first fusion peptide CD19-KIR2DS2 and a second fusion peptide Dap12-BB, wherein:
  • the first fusion peptide CD19-KIR2DS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is a CD19 scFv, and the transmembrane domain is a KIR2DS2 transmembrane domain;
  • the second fusion peptide Dap12-BB comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • the CD19-KIR2DS2/Dap12-BB chimeric antigen receptor is formed by the cleavage of DPK03 protein by T2A peptide, and the DPK03 fusion protein comprises DAP12 signal peptide, DAP12 (including transmembrane domain and cytoplasmic domain), 4- 1BB, T2A cleavage site, CD8 ⁇ signal peptide, CD19 scFv and KIR2DS2, the amino acid sequence of the DPK03 fusion protein is shown in SEQ ID NO.12.
  • DPK03 DAP12 signal peptide+DAP12+4-1BB+T2A+CD8 ⁇ signal peptide+CD19 scFv+KIR2DS2
  • the CD19-KIRS2/Dap12 chimeric antigen receptor comprises a first fusion peptide CD19-KIRS2 and a second fusion peptide Dap12, wherein:
  • the first fusion peptide CD19-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is a CD19 scFv, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide Dap12 comprises a DAP12 transmembrane domain and a DAP12 cytoplasmic domain.
  • the CD19-KIRS2/Dap12 chimeric antigen receptor is formed by the cleavage of the pKT011 protein by the T2A peptide, and the pKT011 fusion protein comprises a DAP12 signal peptide, DAP12 (including a transmembrane domain and a cytoplasmic domain), and a T2A cleavage site.
  • CD8 ⁇ signal peptide, CD19 scFv and KIRS2 the amino acid sequence of the pKT011 fusion protein is shown in SEQ ID NO.13.
  • pKT011 DAP12 signal peptide+DAP12+T2A+CD8 ⁇ signal peptide+CD19 scFv+KIRS2
  • the amino acid sequence of the DAP12 signal peptide is as follows:
  • the amino acid sequence of DAP12 includes the DAP12 transmembrane domain and the DAP12 cytoplasmic domain, and the specific sequence is as follows:
  • the truncated amino acid sequence of DAP12 includes a truncated DAP12 transmembrane domain and a DAP12 cytoplasmic domain, and the specific sequence is as follows:
  • the amino acid sequence of the CD8 ⁇ signal peptide is as follows:
  • amino acid sequence of 4-1BB is as follows:
  • the amino acid sequence of the T2A cleavage site is as follows:
  • the amino acid sequence of CD19 scFv is as follows:
  • KIRS2 The amino acid sequence of KIRS2 is as follows:
  • amino acid sequence of KIR2DS2 is as follows:
  • the amino acid sequence of DPK01 is as follows:
  • the amino acid sequence of DPK02 is as follows:
  • the amino acid sequence of DPK03 is as follows:
  • amino acid sequence of pKT011 is as follows:
  • the amino acid sequence of the transmembrane domain of DAP12 is as follows:
  • the amino acid sequence of the cytoplasmic domain of DAP12 is as follows:
  • the nucleic acid sequence of DPK01 is as follows:
  • the nucleic acid sequence of DPK02 is as follows:
  • the nucleic acid sequence of DPK03 is as follows:
  • the cells were observed, and the cell density reached approximately 80%-90%, at which point the transfection started.
  • plasmid DNA and calcium chloride solution take a T150 bottle as an example, 28 ⁇ g pRSV.rev (purchased from Invitrogen Company), 28 ⁇ g pGAG-Pol (purchased from Invitrogen Company), 11 ⁇ g pVSVG (purchased from Invitrogen Company), 23 ⁇ g of lentiviral expression plasmids (plasmids DPK01, DPK02, DPK03, pKT011) were added to 1.5 ml of calcium chloride solution and mixed.
  • the DNA-calcium chloride-borate buffer mixture (purchased from Shanghai Biyuntian Biotechnology Co., Ltd.) was uniformly added dropwise to the T150 bottle with a 5ml pipette. Incubate in a 37°C cell incubator with 5% carbon dioxide, and change the medium for 6h.
  • the supernatants from the two collections were pooled and filtered through a 0.45 ⁇ m filter to remove cell debris.
  • the supernatant was poured out, fresh 5% (wt) FBS DMEM medium was added to resuspend, and the virus was subpackaged (denoted as DPK01, DPK02, DPK03, pKT011), and quickly stored in a -80°C refrigerator for later use.
  • the CAR lentivirus (DPK01, DPK02, DPK03, pKT011) prepared in Example 2 was added to transfect T cells, and the lentivirus was removed after 48h of infection.
  • D4 On day 4 (D4), the cells were observed in supplemental culture every 1-2 days, and the cell density was maintained at 0.8 ⁇ 10 6 cells/mL.
  • Activation medium was used on days 4-5 (D4-D5), and expansion medium (X-VIVO15, 300 IU/ml IL-2) was used after day 5 (D5).
  • Four kinds of CAR-T cells (DPK01, DPK02, DPK03, pKT011) were obtained by continuous culture until the 11th day (D11).
  • CAR-T cell subset detection antibodies anti-human CD3 (APC-Cy7), anti-human CCR7 antibody (BV421), anti-human CD45RO antibody (APC), CD4-BB515, CD8-BV510, CD45RA (PE- Cy7), CD62L-PE.
  • MCF-7 and MC-7-CD19 were cultured in DMEM medium (DMEM+10% FBS+1% Penicillin/Streptomycine); Nalm6 and L428 were cultured in 1640 medium (1640+10 %FBS+1% penicillin/streptomycin).
  • the target cells were digested and counted, and the target cell suspension with a density of 2 ⁇ 10 5 cells/ml was adjusted with culture medium (DMEM+10% FBS);
  • RTCA real-time label-free dynamic cell analysis technology
  • the monitoring time shall not exceed 24h.
  • NTD non-transduced T cells
  • DPK01, DPK02, DPK03, pKT011 CAR-T were co-cultured with Nalm6 (positive target cells) and L428 (negative target cells) for 24 hours, respectively; IFN- ⁇ .
  • CAR-T was cultured to the 8th day (D8) to demagnetize the beads, and continued to culture for 2 days with IL-2-free medium;
  • Count the target cells (Nalm-6 and 293T), and adjust the cell suspension with a density of 1 ⁇ 10 6 /ml with a culture medium (1640+10% FBS+300UI/ml IL-2) for use;
  • Tumor cells Nalm6 (1 ⁇ 10 7 cells)
  • Tumor formation method subcutaneous tumor formation
  • Quantification of soluble cytokines was performed using Luminex bead array technology and kits purchased from Life technologies (Invitrogen). Assays were performed using an 8-point standard curve generated by 3-fold serial dilutions according to the manufacturer's protocol. Each standard spot and sample was evaluated in duplicate at a 1:3 dilution; the calculated %CV was less than 15% for both measurements. The standard curve quantification range was determined by the 80-120% (observed/expected) range. Individual analyte quantification ranges are reported in the figure captions. DPK01, DPK02 and DPK03 of the present disclosure significantly decreased the levels of endocrine IL-6 in mice (Fig. 9), indicating that the present disclosure can achieve better clinical safety, and IL-10 was also significantly decreased, indicating that the production of lower immunosuppressive factors, Promote clinical efficacy.
  • T cells were transfected with DPK01 lentivirus after 24 h, and the lentivirus was removed after 48 h of infection.
  • D4 On day 4 (D4), the cells were observed in supplemental culture every 1-2 days, and the cell density was maintained at 0.8 ⁇ 10 6 cells/mL.
  • Activation medium was used on days 4-5 (D4-D5), and expansion medium (X-VIVO15, 300 IU/ml IL-2) was used after day 5 (D5). Culture was continued until days 7-12 (D7-D12).
  • the CAR clinical-grade vector was manufactured in Nanjing Aide Immunotherapy Research Institute Co., Ltd.
  • the cells are cryopreserved in an injectable freezing medium.
  • Administer CAR-T cells in accordance with the dose administered.
  • Each bag contains an aliquot of freezing medium (volume depends on dose), and cryopreservation solution is CS5.
  • Bags (10-100ml) containing CD19 CAR-T cells were stored in a -135°C liquid nitrogen box under test. Cryopreservation bags are stored in the freezer until needed.
  • the dose of the first reinfused cells was given in divided doses on Day 0 (D0) and Day 1 (Dl), with approximately 30% cells on Day 0 and 70% cells on Day 1.
  • Frozen cells are shipped to the laboratory or patient on dry ice. Using a water bath maintained at 37°C, cells were thawed and massaged gently until cells were just thawed. There should be no frozen pieces left in the container. If the CD19 CAR-T cell product appears to be in a damaged or leaking pocket, it should not be reinfused.
  • the CD19 CAR-T preparation should not be left at room temperature for too long after warming, so the warming time needs to be determined after all preparations for the subject's treatment are completed.
  • CD19-positive relapsed and refractory B-cell hematological malignancies (2) under 70 years of age; (3) KPS score ⁇ 60, expected survival period ⁇ 3 months; (4) absolute platelet count ⁇ 30 ⁇ 10 9 /L; (5) the absolute number of lymphocytes ⁇ 0.15 ⁇ 10 9 /L; (6) serum ALT ⁇ 100U/L, AST ⁇ 100U/L; (7) total bilirubin ⁇ 30 ⁇ mol; (8) creatinine ⁇ 200 ⁇ mol/L; (9) Women of childbearing age who had a negative urine pregnancy test before the start of administration, and agreed to take effective contraceptive measures during the trial period until the last follow-up; (10) Voluntary enrollment, good compliance, and able to cooperate with the trial observed, and signed written informed consent.
  • Figure 9 shows the flow chart of the clinical treatment protocol of CD19 CAR-T cells.
  • the experimental drug DPK01 was administered according to the clinical situation, 1.2 ⁇ 10 6 CAR-T cells/kg intravenous drip was administered in two doses, 30% was administered on the 0th day (D0), and 70% was administered on the 1st day (D1). %.
  • the above dosing regimen was taken as a course of treatment.
  • Example 6 Structural design of a chimeric antigen receptor targeting mesothelin
  • MSLN CARs mesothelin-targeting chimeric antigen receptors
  • the MSLN SS1 scFv1-KIRS2/Dap12-BB chimeric antigen receptor comprises the first fusion peptide MSLN SS1 scFv1-KIRS2 and the second fusion peptide Dap12-BB, wherein:
  • the first fusion peptide MSLN SS1 scFv1-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is MSLN SS1 scFv1, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide Dap12-BB comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • the MSLN SS1 scFv1-KIRS2/Dap12-BB chimeric antigen receptor is formed by the cleavage of the pKT032 fusion protein by the T2A peptide, and the pKT032 fusion protein comprises the DAP12 signal peptide, DAP12 (including the transmembrane domain and the cytoplasmic domain) , 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, MSLN SS1 scFv1 and KIRS2, the amino acid sequence of the pKT032 fusion protein is shown in SEQ ID NO.21, and its encoding nucleic acid is shown in SEQ ID NO.22.
  • pKT032 DAP12 signal peptide+DAP12+4-1BB+T2A+CD8 ⁇ signal peptide+SS1scFv1+KIRS2
  • the MSLN SS1 scFv2-KIRS2/Dap12-BB chimeric antigen receptor comprises the first fusion peptide MSLN SS1 scFv2-KIRS2 and the second fusion peptide Dap12-BB, wherein:
  • the first fusion peptide MSLN SS1 scFv2-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is MSLN SS1 scFv2, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide Dap12-BB comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • the MSLN SS1 scFv2-KIRS2/Dap12-BB chimeric antigen receptor is formed by the cleavage of the pKT108 fusion protein by the T2A peptide, and the pKT0108 fusion protein comprises the DAP12 signal peptide, DAP12 (including the transmembrane domain and the cytoplasmic domain) , 4-1BB, T2A cleavage site, CD8 ⁇ signal peptide, MSLN SS1 scFv2 and KIRS2, the amino acid sequence of the pKT108 fusion protein is shown in SEQ ID NO.23, and its encoding nucleic acid is shown in SEQ ID NO.24.
  • pKT108 DAP12 signal peptide+DAP12+41BB+T2A+CD8 ⁇ signal peptide+SS1 scFv2+KIRS2
  • amino acid sequence of SS1 scFv1 is as follows:
  • amino acid sequence of SS1 scFv2 is as follows:
  • amino acid sequence of pKT032 is as follows:
  • the nucleotide sequence of pKT032 is as follows:
  • amino acid sequence of pKT108 is as follows:
  • the nucleotide sequence of pKT108 is as follows:
  • the cells were observed, and the cell density reached approximately 80%-90%, at which point the transfection started.
  • plasmid DNA and calcium chloride solution take a T150 bottle as an example, 28 ⁇ g pRSV.rev (purchased from Invitrogen Company), 28 ⁇ g pGAG-Pol (purchased from Invitrogen Company), 11 ⁇ g pVSVG (purchased from Invitrogen Company), 23 ⁇ g of lentiviral expression plasmids (plasmids pKT032, pKT108, synthesized by Shanghai Sangong) were added to 1.5 ml of calcium chloride solution and mixed.
  • the DNA-calcium chloride-borate buffer mixture (purchased from Shanghai Biyuntian Biotechnology Co., Ltd.) was evenly added dropwise to the T150 bottle. Incubate in a 37°C cell incubator with 5% carbon dioxide, and change the medium for 6h.
  • the supernatants from the two collections were pooled and filtered through a 0.45 ⁇ m filter to remove cell debris.
  • lentivirus pKT032, pKT108 packaging
  • lentivirus pKT032, pKT108 packaging
  • CAR-T cells pKT032, pKT108 were obtained by continuous culture until the 11th day (D11).
  • CAR-T cell subset detection antibodies anti-human CD3 (APC-Cy7), anti-human CCR7 antibody (BV421), anti-human CD45RO antibody (APC), CD4-BB515, CD8-BV510, CD45RA (PE- Cy7), CD62L-PE.
  • SK-OV-3 cells were cultured using Myco5A medium (Myco5A+10%FBS+1% Penicillin/Streptomycine); OVCAR-3 cells were cultured using 1640 medium (1640+10%FBS+1 % penicillin/streptomycin).
  • RTCA real-time label-free dynamic cell analysis technology
  • the monitoring time should not exceed 48h.
  • NTD non-transduced T cells
  • CAR-T cells were cultured to the 8th day (D8) with demagnetized beads, and continued to culture for 2 days with IL-2-free medium;
  • Count the target cells (SK-OV-3 and OVCAR-3), and adjust the cell suspension to a density of 1 ⁇ 10 6 /ml with culture medium (Myco5A or 1640+10%FBS+300UI/ml IL-2). ,spare;
  • Example 9 In vivo functional test of MSLN CAR-T cells
  • Tumor cells SKOV-3 (5 ⁇ 10 6 cells)
  • Tumor formation method subcutaneous tumor formation
  • T cells were transfected with pKT032 lentivirus after 24 hours, and the lentivirus was removed after 48 hours of infection.
  • D4 On the 4th day (D4), the cell culture was observed every 1-2 days, and the cell density was maintained at 0.8 ⁇ 10 6 cells/mL.
  • Activation medium was used on days 4-5 (D4-D5), and expansion medium (X-VIVO15, 300 IU/ml IL-2) was used after day 5 (D5). Culture was continued until days 7-12 (D7-D12).
  • the CAR clinical-grade vector was manufactured in Nanjing Aide Immunotherapy Research Institute Co., Ltd.
  • the cells are cryopreserved in an injectable freezing medium.
  • Administer CAR-T cells in accordance with the dose administered.
  • Each bag contains an aliquot of freezing medium (volume depends on dose), and cryopreservation solution is CS5.
  • Bags (10-100 ml) containing mesothelin CAR-T cells were stored in a -135°C liquid nitrogen box under test. Cryopreservation bags are stored in the freezer until needed.
  • the first dose was given in divided doses on days 0 and 1, with approximately 30% cells on day 0 and 70% cells on day 1.
  • Frozen cells are shipped to the laboratory or patient on dry ice. Using a water bath maintained at 37°C, cells were thawed and massaged gently until cells were just thawed. There should be no frozen pieces left in the container. If the mesothelin CAR-T cell product appears to be in a damaged or leaking pocket, it should not be reinfused.
  • the mesothelin CAR-T preparation should not be left at room temperature for too long after warming, so the warming time needs to be determined after the subject's treatment preparations are completed.
  • Figure 21 shows a flow chart of the MSLN CAR-T cell clinical treatment protocol.
  • the experimental drug pKT032 was administered by intravenous infusion of 3 ⁇ 10 6 CAR-T cells/kg once according to the clinical situation.
  • the above dosing regimen was taken as a course of treatment.
  • Remarks: 1+ represents the date of statistics; 2 Caused by pretreatment; PR, partial remission; SD, stable disease.
  • the solid tumor patients treated with pKT032 CAR-T cells are all relapsed and refractory. They have been treated with more than 4 kinds of treatments before, but the disease cannot be controlled. For example, 2A patients were enrolled after 40 times of chemotherapy, chemotherapy and targeted drug resistance. CAR-T therapy. According to reports in the literature, the median survival time of patients with relapsed and refractory ovarian cancer was 4.8 months, and the median survival time of patients with solid tumors treated with pKT032 CAR-T cells was 11.6 months ( Figure 22). The median progression-free survival in solid tumors treated with pKT032 CAR-T cells was 7 months ( Figure 23), compared with 2.1 months in the first phase of Penn CAR-T cell therapy (Mol Ther. 2019 Nov 6;27(11):1919-1929) data have obvious benefits.
  • Example 11 Structural Design of a Chimeric Antigen Receptor Targeting GD2
  • the GD2-KIRS2/Dap12-BB chimeric antigen receptor comprises a first fusion peptide GD2-KIRS2 and a second fusion peptide Dap12-BB, wherein:
  • the first fusion peptide GD2-KIRS2 comprises an antigen binding domain and a transmembrane domain, the antigen binding domain is a GD2 scFv, and the transmembrane domain is a KIRS2 transmembrane domain;
  • the second fusion peptide Dap12-BB comprises a DAP12 transmembrane domain, a DAP12 cytoplasmic domain and a costimulatory domain 4-1BB.
  • Described GD2-KIRS2/Dap12-BB chimeric antigen receptor is formed by pKT081 fusion protein after T2A peptide cleavage, described pKT081 fusion protein comprises DAP12 signal peptide, DAP12 (comprising transmembrane domain and cytoplasmic domain), 4 -1BB, T2A cleavage site, CD8 ⁇ signal peptide, GD2 scFv and KIRS2, the amino acid sequence of the pKT081 fusion protein is shown in SEQ ID NO.12, and its encoding nucleic acid sequence is shown in SEQ ID NO.13.
  • pKT081 DAP12 signal peptide+DAP12+4-1BB+T2A+CD8 ⁇ signal peptide+GD2 scFv+KIRS2
  • the GD2-41BB-CD3 ⁇ chimeric antigen receptor comprises a fusion peptide GD2-CD8-CD8Hinge-CD8TM-41BB-CD3 ⁇ , which comprises an antigen binding domain, a transmembrane domain, a costimulatory Domain 4-1BB and signaling domain CD3 ⁇ , wherein the antigen binding domain is GD2 scFv, and the transmembrane domain is CD8TM transmembrane domain.
  • the GD2-41BB-CD3delta chimeric antigen receptor is formed from a pKT082 fusion protein comprising CD8 (including a transmembrane domain and a cytoplasmic domain), 4-1BB, CD3delta and GD2, the pKT082 fusion
  • the amino acid sequence of the protein is shown in SEQ ID NO.14, and its encoding nucleic acid sequence is shown in SEQ ID NO.15.
  • amino acid sequence of GD2 scFv is as follows:
  • amino acid sequence of pKT081 is as follows:
  • nucleic acid sequence of pKT081 is as follows:
  • amino acid sequence of pKT082 is as follows:
  • the nucleic acid sequence of pKT082 is as follows:
  • the cells were observed, and the cell density reached approximately 80%-90%, at which point the transfection was started.
  • plasmid DNA and calcium chloride solution take a T150 bottle as an example, 28 ⁇ g pRSV.rev (purchased from Invitrogen Company), 28 ⁇ g pGAG-Pol (purchased from Invitrogen Company), 11 ⁇ g pVSVG (purchased from Invitrogen Company), 23 ⁇ g of lentiviral expression plasmids (plasmids pKT075, pKT094, pKT095, synthesized by Shanghai Sangong) were added to 1.5 ml of calcium chloride solution and mixed.
  • the DNA-calcium chloride-borate buffer mixture (purchased from Shanghai Biyuntian Biotechnology Co., Ltd.) was evenly added dropwise to the T150 bottle. Incubate in a 37°C cell incubator with 5% carbon dioxide, and change the medium for 6h.
  • the supernatants from the two collections were pooled and filtered through a 0.45 ⁇ m filter to remove cell debris.
  • lentivirus pKT081, pKT082 packaging
  • Example 13 In vitro functional test of GD2 CAR-T
  • CAR-T cells pKT081, pKT082 Two kinds of GD2-targeting CAR-T cells (CAR-T cells pKT081, pKT082) were obtained by continuous culture until the 11th day (D11).
  • CAR-T cell subset detection antibodies anti-human CD3 (APC-Cy7), anti-human CCR7 antibody (BV421), anti-human CD45RO antibody (APC), CD4-BB515, CD8-BV510, CD45RA (PE- Cy7), CD62L-PE.
  • LAN-1 culture uses DMEM medium (DMEM+10%FBS+1% Penicillin/Streptomycine); H1299 culture uses 1640 medium (1640+10%FBS+1% Penicillin/Streptomycine) .
  • the target cells were digested and counted, and the target cell suspension was adjusted to a density of 2 ⁇ 10 5 cells/ml with culture medium (DMEM or 1640+10% FBS);
  • RTCA real-time label-free dynamic cell analysis technology
  • the monitoring time should not exceed 48h.
  • CAR-T cells were cultured to the 8th day (D8) with demagnetized beads, and continued to culture for 2 days with IL-2-free medium;
  • the CFSE fluorescence intensities of the two CAR-Ts (pKT081, pKT082) were detected by flow cytometry (Fig. 30), compared with the NTD control group, after stimulation of GD2-positive target cells, the fluorescence intensities of the CFSEs of the two CAR-Ts shifted to the left Attenuated, it was confirmed that the positive target cells were able to stimulate the proliferation of these two CAR-Ts.

Landscapes

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

Abstract

L'invention concerne un récepteur chimérique contenant un DAP 12 et un domaine de signal de molécule de signal co-stimulateur, ainsi qu'un procédé d'utilisation correspondant. Le récepteur antigénique chimérique comprend un premier peptide de fusion et un second peptide de fusion. Le premier peptide de fusion comprend un domaine de liaison à l'antigène et un domaine transmembranaire. Le second peptide de fusion comprend un domaine transmembranaire, un domaine cytoplasmique et un domaine co-stimulateur.
PCT/CN2021/124728 2020-10-19 2021-10-19 Récepteur chimérique contenant un dap 12 et domaine de signal de molécule de signal co-stimulateur, et son procédé d'utilisation WO2022083590A1 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN202011122767.3 2020-10-19
CN202011122767.3A CN114369168A (zh) 2020-10-19 2020-10-19 包含dap12和共刺激信号分子信号域的嵌合受体及其使用方法
CN202110560490.0A CN115368471A (zh) 2021-05-21 2021-05-21 靶向间皮素的嵌合受体及其用途
CN202110560554.7A CN115368472A (zh) 2021-05-21 2021-05-21 靶向gd2的嵌合受体及其用途
CN202110560554.7 2021-05-21
CN202110560490.0 2021-05-21

Publications (1)

Publication Number Publication Date
WO2022083590A1 true WO2022083590A1 (fr) 2022-04-28

Family

ID=81289680

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/124728 WO2022083590A1 (fr) 2020-10-19 2021-10-19 Récepteur chimérique contenant un dap 12 et domaine de signal de molécule de signal co-stimulateur, et son procédé d'utilisation

Country Status (1)

Country Link
WO (1) WO2022083590A1 (fr)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014145252A2 (fr) * 2013-03-15 2014-09-18 Milone Michael C Ciblage de cellules cytotoxiques par des récepteurs chimériques pour une immunothérapie adoptive
US20170081411A1 (en) * 2014-03-15 2017-03-23 Novartis Ag Regulatable chimeric antigen receptor
CN107580628A (zh) * 2014-09-17 2018-01-12 诺华股份有限公司 用于过继免疫疗法的具有嵌合受体的靶向细胞毒性细胞
CN108752482A (zh) * 2018-06-12 2018-11-06 南京卡提医学科技有限公司 携带截短或未截短的髓样细胞触发性受体信号结构的嵌合抗原受体及其应用
CN108822216A (zh) * 2018-06-08 2018-11-16 南京卡提医学科技有限公司 携带截短或未截短的自然细胞毒性受体信号结构的嵌合抗原受体及其应用
CN109328230A (zh) * 2016-04-15 2019-02-12 宾夕法尼亚大学董事会 嵌合同种异体抗原受体t细胞的组合物和方法
WO2019115818A2 (fr) * 2017-12-14 2019-06-20 Celyad S.A. Domaines de signalisation de regroupement et de co-stimulation dans une conception de car flexible
US20200085868A1 (en) * 2018-09-17 2020-03-19 China Medical University Hospital Method For Treating Cancer
CN113493525A (zh) * 2020-04-07 2021-10-12 南京卡提医学科技有限公司 一种增效且抵抗耗竭的嵌合抗原受体t细胞及其在制备治疗肿瘤药物中的用途

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014145252A2 (fr) * 2013-03-15 2014-09-18 Milone Michael C Ciblage de cellules cytotoxiques par des récepteurs chimériques pour une immunothérapie adoptive
US20170081411A1 (en) * 2014-03-15 2017-03-23 Novartis Ag Regulatable chimeric antigen receptor
CN107580628A (zh) * 2014-09-17 2018-01-12 诺华股份有限公司 用于过继免疫疗法的具有嵌合受体的靶向细胞毒性细胞
CN109328230A (zh) * 2016-04-15 2019-02-12 宾夕法尼亚大学董事会 嵌合同种异体抗原受体t细胞的组合物和方法
WO2019115818A2 (fr) * 2017-12-14 2019-06-20 Celyad S.A. Domaines de signalisation de regroupement et de co-stimulation dans une conception de car flexible
CN108822216A (zh) * 2018-06-08 2018-11-16 南京卡提医学科技有限公司 携带截短或未截短的自然细胞毒性受体信号结构的嵌合抗原受体及其应用
CN108752482A (zh) * 2018-06-12 2018-11-06 南京卡提医学科技有限公司 携带截短或未截短的髓样细胞触发性受体信号结构的嵌合抗原受体及其应用
US20200085868A1 (en) * 2018-09-17 2020-03-19 China Medical University Hospital Method For Treating Cancer
CN113493525A (zh) * 2020-04-07 2021-10-12 南京卡提医学科技有限公司 一种增效且抵抗耗竭的嵌合抗原受体t细胞及其在制备治疗肿瘤药物中的用途

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
E. WANG, L.-C. WANG, C.-Y. TSAI, V. BHOJ, Z. GERSHENSON, E. MOON, K. NEWICK, J. SUN, A. LO, T. BARADET, M. D. FELDMAN, D. BARRETT,: "Generation of Potent T-cell Immunotherapy for Cancer Using DAP12-Based, Multichain, Chimeric Immunoreceptors", CANCER IMMUNOLOGY RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 3, no. 7, 1 July 2015 (2015-07-01), US , pages 815 - 826, XP055241621, ISSN: 2326-6066, DOI: 10.1158/2326-6066.CIR-15-0054 *
WANG ENXIU; WANG LIANG-CHUAN; TSAI CHING-YI; BHOJ VIJAY; MOON EDMUND; ALBELDA STEVEN; MILONE MICHAEL C: "A Chimeric Antigen Receptor (CARs) Based Upon a Killer Immunoglobulin-Like Receptor (KIR) Triggers Robust Cytotoxic Activity in Solid Tumors", MOLECULAR THERAPY, ELSEVIER INC., US, vol. 22, no. Suppl.1, 1 May 2014 (2014-05-01), US , pages S57, XP009179562, ISSN: 1525-0016 *

Similar Documents

Publication Publication Date Title
JP7034934B2 (ja) キメラ抗原及びt細胞受容体、並びに使用方法
US11541076B2 (en) Chimeric antigen receptors targeting TIM-1
JP2020054387A (ja) 免疫療法のための組成物および方法
US20230192805A1 (en) Chimeric receptor polypeptides and uses thereof
JP2022523781A (ja) Dll3標的化キメラ抗原受容体および結合剤
CN112512575A (zh) 双特异性抗体组合物及其使用方法
CA3205637A1 (fr) Traitement du cancer par des cellules nk et un anticorps ciblant cd20
CN112930199A (zh) 免疫效应细胞治疗肿瘤的方法
CA3180613A1 (fr) Traitement du cancer cd30 positif
CA3216024A1 (fr) Recepteur antigenique chimerique comprenant un anticorps anti-her2 ou un fragment de liaison a l'antigene de celui-ci et cellules tueuses naturelles le comprenant
CA3150884A1 (fr) Lymphocyte t de recepteur antigenique chimerique stabilisant une synapse immunologiqueogique
WO2022214089A1 (fr) Utilisation d'immunothérapie cellulaire
WO2022083590A1 (fr) Récepteur chimérique contenant un dap 12 et domaine de signal de molécule de signal co-stimulateur, et son procédé d'utilisation
US20240060046A1 (en) Expanded and stimulated natural killer cells
CN111542547A (zh) 对bdca2抗原具有特异性的嵌合抗原受体
CN116783288A (zh) 用于cd70定向癌症免疫疗法的基因修饰的自然杀伤细胞
EP4103610A1 (fr) Anticorps bispécifiques anti-cd44-ctla4
JP2022525318A (ja) Cd147キメラ抗原受容体および使用の方法
CN115368471A (zh) 靶向间皮素的嵌合受体及其用途
US20230242666A1 (en) Methods and Compositions for the Reduction of Chimeric Antigen Receptor Tonic Signaling
WO2022268162A1 (fr) Méthode et composition de traitement de tumeurs
CN114369168A (zh) 包含dap12和共刺激信号分子信号域的嵌合受体及其使用方法
CN115368470A (zh) 靶向b7-h3的嵌合受体及其用途
CN115368472A (zh) 靶向gd2的嵌合受体及其用途
US20240181055A1 (en) Cellular immunotherapy use

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: 21882006

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21882006

Country of ref document: EP

Kind code of ref document: A1