WO2023025009A1 - Cellule immunitaire modifiée et son utilisation - Google Patents

Cellule immunitaire modifiée et son utilisation Download PDF

Info

Publication number
WO2023025009A1
WO2023025009A1 PCT/CN2022/113151 CN2022113151W WO2023025009A1 WO 2023025009 A1 WO2023025009 A1 WO 2023025009A1 CN 2022113151 W CN2022113151 W CN 2022113151W WO 2023025009 A1 WO2023025009 A1 WO 2023025009A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
engineered immune
immune cell
antigen
nucleic acid
Prior art date
Application number
PCT/CN2022/113151
Other languages
English (en)
Chinese (zh)
Inventor
邢芸
任江涛
Original Assignee
南京北恒生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京北恒生物科技有限公司 filed Critical 南京北恒生物科技有限公司
Publication of WO2023025009A1 publication Critical patent/WO2023025009A1/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
    • A61K39/464436Cytokines
    • A61K39/46444Interleukins [IL]
    • 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
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464416Receptors for cytokines
    • A61K39/464421Receptors for chemokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • 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
    • A61K2239/27Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by targeting or presenting multiple antigens
    • A61K2239/28Expressing multiple CARs, TCRs or antigens
    • 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
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/54Pancreas

Definitions

  • This disclosure is in the field of immunotherapy. More specifically, the present disclosure relates to an engineered immune cell that expresses a cell surface molecule that specifically recognizes an antigen and an exogenous CCL20 gene. More preferably, the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor.
  • Lymphocytes generally sense chemokines secreted by other cells through chemokine receptors, and then are directed to home to target sites. Despite the large number of chemokines and their receptors, generally only a limited number of chemokine receptors are expressed per lymphocyte. Therefore, when certain tumor cells secrete a certain chemokine in large quantities and lymphocytes (such as engineered CAR-T cells) do not express the receptors paired with the chemokine, the lymphocytes cannot be effectively transported to the tumor The microenvironment affects the therapeutic effect.
  • the present disclosure provides a novel engineered immune cell expressing a cell surface molecule that specifically recognizes an antigen and exogenous CCL20.
  • the engineered immune cells of the present disclosure further express exogenous IL7.
  • the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor or a T cell receptor, preferably a chimeric antigen receptor.
  • the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor comprising an antigen binding region, a transmembrane domain and an intracellular signaling domain comprising costimulatory domain and/or primary signaling domain.
  • the antigen binding region can be selected from IgG, Fab, Fab', F(ab')2, Fd, Fd', Fv, scFv, sdFv, linear antibody, single domain antibody, nanobody, diabody, anticalin and DARPIN.
  • the antigen binding region is selected from scFv, Fab, single domain antibodies and nanobodies.
  • the cell surface molecule that specifically recognizes the antigen binds to one or more targets selected from: CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33, CD37, CD38, CD40, CD40L, CD44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD179a , DR4, DR5, TAC, TEM1/CD248, VEGF, GUCY2C, EGP40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa light chain, TIM3, TSHR, CD19, BAFF-R, CLL-1, EGFRvIII , tEGFR, GD2, GD3, BCMA, Tn antigen, PSMA, ROR1, FLT3, FAP, TAG72, CD44v6, CEA,
  • the transmembrane domain is selected from the transmembrane domains of the following proteins: TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ chain, TCR ⁇ chain, CD3 ⁇ subunit, CD3 ⁇ subunit, CD3 ⁇ subunit, CD3 ⁇ subunit, CD3 ⁇ subunit, CD45, CD4, CD5, CD8a, CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137 and CD154.
  • the transmembrane domain is selected from the transmembrane domains of CD8 ⁇ , CD4, CD28 and CD278.
  • the primary signaling domain is an intracellular region of a protein selected from the group consisting of FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD22, CD79a, CD79b, and CD66d.
  • said primary signaling domain comprises a CD3 ⁇ intracellular region.
  • the co-stimulatory domain comprises one or more intracellular regions selected from the group consisting of CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD7, CD8, CD18, CD27, CD28, CD30, CD40, CD54, CD83, CD134(OX40), CD137(4-1BB), CD270(HVEM), CD272(BTLA), CD276(B7-H3) , CD278 (ICOS), CD357 (GITR), DAP10, DAP12, LAT, NKG2C, SLP76, PD-1, LIGHT, TRIM, ZAP70, and combinations thereof.
  • the co-stimulatory domain is selected from the intracellular region of CD27, CD28, CD134, CD137, DAP10, DAP12 or CD278 or a combination thereof.
  • the immune cells are selected from T cells, macrophages, dendritic cells, monocytes, NK cells or NKT cells.
  • the T cells are CD4+CD8+ T cells, CD4+ helper T cells, CD8+ T cells, CD4-CD8-T cells, tumor infiltrating cells, memory T cells, naive T cells, ⁇ -T cells or ⁇ -T cells.
  • the exogenous expression or activity of CCL20 and/or IL7 is constitutive.
  • the expression or activity of exogenous CCL20 and/or IL7 is conditional.
  • conditional expression is achieved by operably linking the exogenous gene to an inducible, repressible or tissue-specific promoter.
  • CCL20 and/or IL7 can be operably linked to a localization domain, which can localize and express the exogenous gene of the present disclosure on a specific cell location, such as a cell membrane.
  • exogenous genes of the present disclosure such as CCL20 and/or IL7, are operably linked to transmembrane domains, thereby anchoring expression on the surface of engineered immune cells.
  • the present disclosure provides a nucleic acid molecule comprising a nucleic acid sequence encoding a cell surface molecule that specifically recognizes an antigen and a nucleic acid sequence encoding CCL20.
  • the nucleic acid molecule further comprises a nucleic acid sequence encoding IL7.
  • the cell surface molecule that specifically recognizes an antigen is a chimeric antigen receptor or a T cell receptor, more preferably a chimeric antigen receptor.
  • the nucleic acid is DNA or RNA.
  • the present disclosure also provides vectors comprising the nucleic acid molecules described above.
  • the vector is selected from plasmids, retroviruses, lentiviruses, adenoviruses, vaccinia viruses, Rous sarcoma virus (RSV), polyoma virus and adeno-associated virus (AAV).
  • the vector further comprises an origin of autonomous replication in immune cells, a selectable marker, a restriction enzyme cleavage site, a promoter, a polyA tail (polyA), a 3'UTR, a 5'UTR, an enhancer Elements such as promoters, terminators, insulators, operators, selectable markers, reporter genes, targeting sequences and/or protein purification tags.
  • said vector is an in vitro transcribed vector.
  • the present disclosure also provides a pharmaceutical composition, which comprises the engineered immune cells, nucleic acid molecules or vectors described in the present disclosure, and one or more pharmaceutically acceptable excipients.
  • the present disclosure also provides a method of treating a subject suffering from cancer, infection or autoimmune disease, comprising administering to the subject an effective amount of the immune cells according to the present disclosure, Nucleic acid molecule, vector or pharmaceutical composition.
  • the cancer is a solid tumor or a hematological tumor. More specifically, the cancer is selected from the group consisting of: brain glioma, blastoma, sarcoma, leukemia, basal cell carcinoma, biliary tract cancer, bladder cancer, bone cancer, brain and CNS cancer, breast cancer, peritoneal cancer, cervical cancer , choriocarcinoma, colon and rectal cancer, connective tissue cancer, cancer of the digestive system, endometrial cancer, esophageal cancer, eye cancer, head and neck cancer, gastric cancer, glioblastoma (GBM), liver cancer, hepatoma, Intraepithelial neoplasms, kidney cancer, laryngeal cancer, liver tumors, lung cancer, lymphoma, melanoma, myeloma, neuroblastoma, oral cancer, ovarian cancer, pancreatic cancer, prostate cancer, retinoblastoma, rhabdomyosarcoma,
  • the infections include, but are not limited to, infections caused by viruses, bacteria, fungi, and parasites.
  • the autoimmune disease includes, but is not limited to, type 1 diabetes, celiac disease, Graves' disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, Addison Illness, Sjogren's syndrome, Hashimoto's thyroiditis, myasthenia gravis, vasculitis, pernicious anemia and systemic lupus erythematosus, etc.
  • Figure 1 CAR expression levels of CAR-T cells determined by flow cytometry.
  • Figure 2 The expression level of IL7 in CAR-T cells determined by ELISA.
  • Figure 3 The expression level of CCL20 in CAR-T cells determined by ELISA.
  • FIG. 4 IFN- ⁇ release levels after CAR-T cells were co-cultured with target cells and non-target cells.
  • Figure 5 The body weight change curve of mice after pancreatic cancer was treated with CAR-T cells.
  • Figure 6 Tumor growth curves of mice treated with CAR-T cells for pancreatic cancer.
  • the present disclosure provides a novel engineered immune cell expressing a cell surface molecule that specifically recognizes an antigen and exogenous CCL20.
  • the term "cell surface molecule that specifically recognizes an antigen” refers to a molecule expressed on the surface of a cell that is capable of specifically binding to a target molecule (eg, an antigen).
  • a target molecule eg, an antigen
  • Such surface molecules generally comprise an antigen-binding region capable of specifically binding to an antigen, a transmembrane domain that anchors the surface molecule to the cell surface, and an intracellular domain responsible for signal transmission.
  • Common examples of such surface molecules include eg T cell receptors or chimeric antigen receptors.
  • T cell receptor refers to a membrane protein complex that responds to antigen presentation and participates in T cell activation. Stimulation of the TCR is triggered by major histocompatibility complex molecules (MHC) on antigen-presenting cells, which present antigenic peptides to T cells and bind to the TCR complex to induce a series of intracellular signaling.
  • MHC major histocompatibility complex molecules
  • TCR is composed of six peptide chains that form heterodimers, which are generally divided into ⁇ type and ⁇ type. Each peptide chain includes a constant region and a variable region, where the variable region is responsible for binding specific antigens and MHC molecules.
  • the variable region of the TCR may comprise or be operably linked to an antigen binding region, wherein the definition of the antigen binding region is as follows.
  • chimeric antigen receptor refers to an artificially constructed hybrid polypeptide that generally includes an antigen-binding region (such as the antigen-binding portion of an antibody), a transmembrane domain, and an intracellular Signal transduction domains (comprising co-stimulatory domains and/or primary signal transduction domains), each domain is connected by a linker.
  • CARs are able to exploit the antigen-binding properties of monoclonal antibodies to redirect the specificity and reactivity of T cells and other immune cells to a target of choice in a non-MHC-restricted manner.
  • Non-MHC-restricted antigen recognition confers on CAR cells the ability to recognize antigens independently of antigen processing, thus bypassing major mechanisms of tumor escape. Furthermore, CAR advantageously does not dimerize with the alpha and beta chains of the endogenous T cell receptor (TCR) when expressed in T cells.
  • TCR T cell receptor
  • antigen binding region refers to any structure or functional variant thereof that can bind to an antigen.
  • the antigen binding region can be an antibody structure, including but not limited to monoclonal antibody, polyclonal antibody, recombinant antibody, human antibody, humanized antibody, murine antibody, chimeric antibody and functional fragments thereof.
  • antigen binding domains include, but are not limited to, IgG, Fab, Fab', F(ab')2, Fd, Fd', Fv, scFv, sdFv, linear antibodies, single domain antibodies, nanobodies, diabodies, anticalins, DARPIN etc., preferably selected from Fab, scFv, sdAb and Nanobodies.
  • the antigen binding domain may be monovalent or bivalent, and may be a monospecific, bispecific or multispecific antibody.
  • the antigen binding region may also be a specific binding polypeptide or receptor structure of a specific protein, such as PD1, PDL1, PDL2, TGF ⁇ , APRIL and NKG2D.
  • the term "functional variant” or “functional fragment” refers to a variant comprising essentially the amino acid sequence of a parent but containing at least one amino acid modification (i.e. substitution, deletion or insertion) compared to the parent amino acid sequence, provided that the Such variants retain the biological activity of the parent amino acid sequence.
  • the amino acid modification is preferably a conservative modification.
  • conservative modification refers to an amino acid modification that does not significantly affect or alter the binding characteristics of an antibody or antibody fragment comprising the amino acid sequence. These conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into chimeric antigen receptors of the disclosure by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. A conservative amino acid substitution is one in which an amino acid residue is replaced by an amino acid residue with a similar side chain.
  • Families of amino acid residues with similar side chains have been defined in the art and include basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid, ), uncharged polar side chains (e.g. glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), non-polar side chains (e.g. alanine, valine acid, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g.
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid,
  • uncharged polar side chains e.g. glycine, asparagine, glutamine, serine, threonine, tyros
  • threonine valine, isoleucine
  • aromatic side chains eg, tyrosine, phenylalanine, tryptophan, histidine.
  • Conservative modifications can be selected, for example, on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • a “functional variant” or “functional fragment” has at least 75%, preferably at least 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84% of the parent amino acid sequence. %, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity, And retain the biological activity of the parent amino acid, such as binding activity.
  • sequence identity means the degree to which two (nucleotide or amino acid) sequences in an alignment have the same residue at the same position, and is usually expressed as a percentage. Preferably, identity is determined over the entire length of the sequences being compared. Therefore, two copies of the exact same sequence have 100% identity.
  • sequence identity can be determined using standard parameters, such as Blast (Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402), Blast2 (Altschul et al. (1990) J. Mol. Biol. 215:403-410), Smith-Waterman (Smith et al. (1981) J. Mol. Biol. 147:195-197) and Clustal W.
  • an antigen binding region of the present disclosure binds to one or more targets selected from the group consisting of: CD2, CD3, CD4, CD5, CD7, CD8, CD14, CD15, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD30, CD33, CD37, CD38, CD40, CD40L, CD44, CD46, CD47, CD52, CD54, CD56, CD70, CD73, CD80, CD97, CD123, CD126, CD138, CD171, CD 179a, DR4 , DR5, TAC, TEM1/CD248, VEGF, GUCY2C, EGP40, EGP-2, EGP-4, CD133, IFNAR1, DLL3, kappa light chain, TIM3, TSHR, CD19, BAFF-R, CLL-1, EG
  • the CARs of the present disclosure can be designed to include an antigen-binding region specific for that antigen.
  • the target is selected from CD7, CD19, CD20, CD22, CD30, CD33, CD38, CD123, CD138, CD171, MUC1, AFP, Folate receptor alpha, CEA, PSCA, PSMA, Her2, EGFR, IL13Ra2, GD2 , NKG2D, Claudin18.2, ROR1, EGFRvIII, CS1, BCMA, GPRC5D, mesothelin, and any combination thereof.
  • a CD19 antibody can be used as an antigen binding region of the present disclosure.
  • the antigen binding region is an antibody targeting CD19, which has the same CDRs as the antibody shown in SEQ ID NO: 1 or 2.
  • transmembrane domain refers to a polypeptide capable of expressing a chimeric antigen receptor on the surface of an immune cell (such as a lymphocyte, NK cell or NKT cell) and directing a cellular response of the immune cell against a target cell structure.
  • Transmembrane domains can be natural or synthetic and can be derived from any membrane-bound or transmembrane protein. The transmembrane domain is capable of signaling when the chimeric antigen receptor binds to the target antigen.
  • Transmembrane domains particularly suitable for use in the present disclosure may be derived from, for example, TCR alpha chain, TCR beta chain, TCR gamma chain, TCR delta chain, CD3 zeta subunit, CD3 epsilon subunit, CD3 gamma subunit, CD3 delta subunit, CD45, CD4, CD5, CD8 alpha , CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD137, CD154 and functional fragments thereof.
  • the transmembrane domain may be synthetic and may comprise predominantly hydrophobic residues such as leucine and valine.
  • the transmembrane domain is derived from CD28, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% of the amino acid sequence shown in SEQ ID NO:3 or 100% sequence identity; or derived from CD8 ⁇ , it has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% with the amino acid sequence shown in SEQ ID NO: 4 or 5 or 100% sequence identity.
  • the chimeric antigen receptors of the present disclosure may further comprise a hinge region located between the antigen binding region and the transmembrane domain.
  • the term "hinge region” generally refers to any oligopeptide or polypeptide that functions to link a transmembrane domain to an antigen binding region. Specifically, the hinge region is used to provide greater flexibility and accessibility to the antigen binding region.
  • the hinge region may comprise up to 300 amino acids, preferably 10 to 100 amino acids and most preferably 25 to 50 amino acids.
  • the hinge region may be derived in whole or in part from a natural molecule, such as in whole or in part from the extracellular region of CD8, FcyRIIIa receptor, IgG4, IgGl, CD4 or CD28, or in whole or in part from an antibody constant region.
  • the hinge region may be a synthetic sequence corresponding to a naturally occurring hinge sequence, or may be an entirely synthetic hinge sequence.
  • the hinge region comprises a CD28 hinge, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% of the amino acid sequence shown in SEQ ID NO:15.
  • CD8 ⁇ hinge which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity; or comprising an IgG4 hinge having at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% to the amino acid sequence shown in SEQ ID NO: 18 % or 100% sequence identity.
  • intracellular signaling domain refers to the portion of a protein that transduces effector function signals and directs the cell to perform a specified function, which includes a co-stimulatory domain and/or a primary signaling domain.
  • the intracellular signaling domain is responsible for intracellular signal transmission following antigen binding at the antigen binding region, resulting in activation of immune cells and immune responses.
  • the intracellular signaling domain is responsible for activating at least one of the normal effector functions of the immune cell in which the CAR is expressed.
  • the effector function of a T cell can be cytolytic activity or helper activity, including secretion of cytokines.
  • the chimeric antigen receptors of the present disclosure comprise a primary signaling domain, which may be the cytoplasmic sequence of the T cell receptor and co-receptor that function together to elicit primary signaling following antigen receptor binding , and any derivatives or variants of these sequences and any synthetic sequences having the same or similar function.
  • the primary signaling domain can contain many immunoreceptor tyrosine-based activation motifs (Immunoreceptor Tyrosine-based Activation Motifs, ITAM).
  • Non-limiting examples of primary signaling domains of the present disclosure include, but are not limited to, those derived from FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD22, CD79a, CD79b, and CD66d.
  • the primary signaling domain of the disclosed CAR may comprise a CD3 ⁇ intracellular region, and the signaling domain has at least 70% of the amino acid sequence shown in SEQ ID NO: 9, 10 or 11, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • a chimeric antigen receptor of the disclosure comprises one or more co-stimulatory domains.
  • a co-stimulatory domain may be an intracellular functional signaling domain from a co-stimulatory molecule comprising the entire intracellular portion of said co-stimulatory molecule, or a functional fragment thereof.
  • a "costimulatory molecule” refers to a cognate binding partner that specifically binds to a costimulatory ligand on a T cell, thereby mediating a costimulatory response (eg, proliferation) of the T cell. Costimulatory molecules include, but are not limited to, MHC class 1 molecules, BTLA, and Toll ligand receptors.
  • Non-limiting examples of co-stimulatory domains of the present disclosure include, but are not limited to, intracellular regions derived from the following proteins: CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD7, CD8, CD18, CD27, CD28, CD30, CD40, CD54, CD83, CD134(OX40), CD137(4-1BB), CD270(HVEM), CD272(BTLA), CD276(B7-H3) , CD278 (ICOS), CD357 (GITR), DAP10, DAP12, LAT, NKG2C, SLP76, PD-1, LIGHT, TRIM, and ZAP70.
  • CD94 intracellular regions derived from the following proteins: CD94, LTB, TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, CARD11, CD2, CD
  • the co-stimulatory domain of the CAR of the present disclosure is from 4-1BB, CD28, CD27, OX40, ICOS, DAP10, DAP12 or a combination thereof.
  • the CAR of the present disclosure comprises a CD28 co-stimulatory domain, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% of the amino acid sequence shown in SEQ ID NO:6 Or 99% or 100% sequence identity; and/or comprising a 4-1BB costimulatory domain, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% sequence identity.
  • the CAR of the present disclosure may also comprise a signal peptide such that when it is expressed in a cell, such as a T cell, the nascent protein is directed to the endoplasmic reticulum and subsequently to the cell surface.
  • the core of the signal peptide may contain a long stretch of hydrophobic amino acids with a propensity to form a single ⁇ -helix.
  • At the end of the signal peptide there is usually a stretch of amino acids that is recognized and cleaved by the signal peptidase.
  • the signal peptidase can cleave during translocation or after completion to generate a free signal peptide and mature protein. Then, the free signal peptide is digested by specific proteases.
  • Signal peptides useful in the present disclosure are well known to those skilled in the art, such as signal peptides derived from B2M, CD8 ⁇ , IgG1, GM-CSFR ⁇ , and the like.
  • the signal peptide useful in the present disclosure is a B2M signal peptide, which has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% of the amino acid sequence shown in SEQ ID NO: 12. % or 99% or 100% sequence identity; or CD8 ⁇ signal peptide, it has at least 70%, preferably at least 80%, more preferably at least 90%, 95% with the amino acid sequence shown in SEQ ID NO: 13 or 14 , 97% or 99% or 100% sequence identity.
  • the CAR of the present disclosure may also include a switch structure to regulate the expression time of the CAR.
  • the switch structure can be in the form of a dimerization domain, which induces a conformational change upon binding to its corresponding ligand, exposing the extracellular binding domain to allow it to bind to the targeted antigen, thereby activating the signaling pathway.
  • a switch domain can be used to link the binding and signaling domains separately, and the binding and signaling domains can pass through the dimer only when the switch domains are associated with each other (e.g. in the presence of an inducing compound). Link together to activate the signaling pathway.
  • the switch structure can also be in the form of a masking peptide.
  • the masking peptide can mask the extracellular binding domain and prevent it from binding to the targeted antigen.
  • the masking peptide is cleaved by, for example, a protease, the extracellular binding domain can be exposed, making it an "ordinary" CAR structure.
  • Various switch configurations known to those skilled in the art can be used in the present disclosure.
  • the CAR of the present disclosure may also contain a suicide gene, that is, to express a cell death signal that can be induced by an exogenous substance, so as to eliminate CAR cells when necessary (for example, when severe toxic side effects occur).
  • suicide genes can be in the form of inserted epitopes, such as CD20 epitopes, RQR8, etc., and when necessary, CAR cells can be eliminated by adding antibodies or reagents targeting these epitopes.
  • the suicide gene can also be herpes simplex virus thymidine kinase (HSV-TK), which causes cell death induced by ganciclovir treatment.
  • HSV-TK herpes simplex virus thymidine kinase
  • the suicide gene can also be iCaspase-9, and the dimerization of iCaspase-9 can be induced by chemically inducing drugs such as AP1903 and AP20187, thereby activating the downstream Caspase3 molecule and leading to cell apoptosis.
  • chemically inducing drugs such as AP1903 and AP20187
  • the engineered immune cells of the present disclosure also express exogenous CCL20.
  • Chemotactic cytokines can be divided into four subfamilies, CXC, CC, XC and CX3C, according to the arrangement of their amino-terminal cysteines.
  • CCL20 is a member of the CC subfamily, also known as macrophage inflammatory protein 3 ⁇ (MIP-3 ⁇ ).
  • MIP-3 ⁇ macrophage inflammatory protein 3 ⁇
  • CCL20 has a strong chemotactic effect on lymphocytes and dendritic cells.
  • CCL20 plays a role in autoimmune diseases (such as rheumatoid arthritis, psoriasis, etc.) and malignant tumors (such as cancers of the liver, colon, breast, pancreas, stomach, etc.).
  • the CCL20 used in the present disclosure has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% of the amino acid sequence shown in SEQ ID NO: 25 or 27 % sequence identity, or the coding sequence of CCL20 has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% of the nucleic acid sequence shown in SEQ ID NO: 24 or 26 % sequence identity.
  • the engineered immune cells of the present disclosure further express IL7.
  • the IL7 used in the present disclosure has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% of the amino acid sequence shown in SEQ ID NO: 21 or 23 % sequence identity, or the coding sequence of IL7 has at least 70%, preferably at least 80%, more preferably at least 90%, 95%, 97% or 99% or 100% with the nucleic acid sequence shown in SEQ ID NO: 22 or 24 % sequence identity.
  • exogenous genes in the present disclosure can be expressed constitutively or conditionally.
  • the expression of exogenous CCL20 and/or IL7 is conditional.
  • the exogenous gene of the present disclosure can be operably linked with an inducible, repressible or tissue-specific promoter as required, so as to regulate the expression of the introduced exogenous gene at a specific time or in a specific tissue or cell type level.
  • the promoter is an inducible promoter, ie a promoter that initiates transcription only in the presence of specific environmental conditions, developmental conditions or inducers.
  • environmental conditions include, for example, an acidic tumor microenvironment, a hypoxic tumor microenvironment, and the like.
  • Such inducers include, for example, cyclocycline, tetracycline, or analogs thereof.
  • Analogs of tetracycline include, for example, chlortetracycline, oxytetracycline, desmethylchlortetracycline, methacycline, doxycycline, and minocycline.
  • Inducible promoters include, for example, a Lac operator sequence, a tetracycline operator sequence, a galactose operator sequence, or a doxycycline operator sequence, and the like.
  • the promoter is a repressible promoter, ie, in the presence of a repressor specific for the repressible promoter, expression of the foreign gene in the cell is suppressed or not expressed.
  • Repressible promoters include, for example, a Lac repressible element or a tetracycline repressible element.
  • Inducible/repressible expression systems well known to those skilled in the art can be used in the present disclosure, including but not limited to Tet-on system, Tet-off system, Cre/loxP system, etc.
  • CCL20 and/or IL7 can be operably linked to a localization domain, and the localization domain can localize and express the exogenous gene of the present disclosure on a specific cell location, such as the cell membrane and the like.
  • Localization domains include, but are not limited to, nuclear localization signals, leader peptides, transmembrane domains, and the like.
  • the exogenous gene CCL20 and/or IL7 of the present disclosure is operably linked to the transmembrane domain, so as to be expressed on the surface of the engineered immune cells.
  • the exogenous genes in the present disclosure can be wild-type or fusion proteins or mutants with specific properties (eg resistance to proteolysis).
  • the present disclosure also provides a nucleic acid molecule comprising a nucleic acid sequence encoding a cell surface molecule that specifically recognizes an antigen and a nucleic acid sequence encoding CCL20.
  • the nucleic acid molecule further comprises a nucleic acid sequence encoding IL7.
  • the cell surface molecule that specifically recognizes an antigen is a T cell receptor or a chimeric antigen receptor, preferably a chimeric antigen receptor.
  • Chimeric antigen receptors are defined above.
  • nucleic acid molecule includes sequences of ribonucleotides and deoxyribonucleotides, such as modified or unmodified RNA or DNA, each in single- and/or double-stranded form, linear or circular shape, or their mixtures (including hybrid molecules).
  • nucleic acids according to the present disclosure include DNA (such as dsDNA, ssDNA, cDNA), RNA (such as dsRNA, ssRNA, mRNA, ivtRNA), combinations or derivatives thereof (such as PNA).
  • the nucleic acid is DNA or RNA, more preferably mRNA.
  • the present disclosure also provides a vector comprising the nucleic acid as described in the present disclosure.
  • the nucleic acid sequence encoding the cell surface molecule that specifically recognizes the antigen, the nucleic acid sequence encoding CCL20 and the optional nucleic acid sequence encoding IL7 may be located in one or more vectors.
  • vector is a nucleic acid molecule used as a vehicle for the transfer of (exogenous) genetic material into a host cell where it can eg be replicated and/or expressed.
  • Targeting vector is a medium that delivers an isolated nucleic acid to the interior of a cell by, for example, homologous recombination or a hybrid recombinase using a sequence at a specific targeting site.
  • An "expression vector” is a vector used for the transcription of heterologous nucleic acid sequences, such as those encoding chimeric antigen receptor polypeptides of the present disclosure, and translation of their mRNA in a suitable host cell. Suitable vectors that can be used in the present disclosure are known in the art and many are commercially available.
  • vectors of the present disclosure include, but are not limited to, plasmids, viruses such as retroviruses, oncolytic viruses, lentiviruses, adenoviruses, vaccinia virus, Rous sarcoma virus, polyoma virus, and adeno-associated virus (AAV ), etc.), phage, phagemid, cosmid and artificial chromosome (including BAC and YAC).
  • the vector itself is usually a sequence of nucleotides, usually a DNA sequence containing the insert (transgene) and a larger sequence that acts as the "backbone" of the vector.
  • the engineered vector usually also contains an origin of autonomous replication in the host cell (if stable expression of the polynucleotide is desired), a selectable marker, and a restriction enzyme cleavage site (such as a multiple cloning site, MCS).
  • the vector may additionally comprise elements such as a promoter, a polyA tail (polyA), a 3' UTR, an enhancer, a terminator, an insulator, an operator, a selectable marker, a reporter gene, a targeting sequence and/or a protein purification tag.
  • said vector is an in vitro transcribed vector.
  • the present disclosure also provides an engineered immune cell comprising the nucleic acid or vector of the present disclosure.
  • the engineered immune cells of the present disclosure express cell surface molecules that specifically recognize antigens and an exogenous CCL20 gene, and optionally an exogenous IL7 gene.
  • the term "immune cell” refers to any cell of the immune system that has one or more effector functions (eg, cytotoxic cell killing activity, secretion of cytokines, induction of ADCC and/or CDC).
  • the immune cells can be T cells, macrophages, dendritic cells, monocytes, NK cells and/or NKT cells, or immune cells obtained from stem cell sources such as iPSCs and ESCs.
  • the immune cells are T cells.
  • the T cells may be any T cells, such as T cells cultured in vitro, such as primary T cells, or T cells from T cell lines cultured in vitro, such as Jurkat, SupT1, etc., or T cells obtained from a subject.
  • T cells can be obtained from a variety of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors. T cells can also be enriched or purified.
  • T cells can be at any developmental stage, including, but not limited to, CD4+CD8+ T cells, CD4+ helper T cells (such as Th1 and Th2 cells), CD8+ T cells (such as cytotoxic T cells), CD4-CD8-T cells, tumor infiltrating cells, memory T cells, naive T cells, ⁇ -T cells, ⁇ -T cells, etc.
  • the immune cells are human T cells.
  • T cells can be obtained from the blood of a subject using a variety of techniques known to those of skill in the art, such as Ficoll separation.
  • the immune cells of the present disclosure further comprise at least one inactivated gene selected from the group consisting of: CD52, GR, TCR ⁇ , TCR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD247 ⁇ , HLA-I, HLA-II, B2M , immune checkpoint genes such as PD1, CTLA-4, LAG3 and TIM3. More specifically, at least TCR components (including TCR ⁇ , TCR ⁇ genes) or CD3 components (including CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD247 ⁇ ) in immune cells are inactivated. This inactivation renders the TCR-CD3 complex nonfunctional in the cell. This strategy is particularly useful for avoiding graft-versus-host disease (GvHD).
  • GvHD graft-versus-host disease
  • DNA fragmentation is mediated by meganuclease, zinc finger nuclease, TALEN nuclease or Cas enzyme in the CRISPR system, thereby inactivating the gene.
  • the present disclosure also provides a pharmaceutical composition, which comprises the engineered immune cell, nucleic acid molecule or carrier described in the present disclosure as an active agent, and one or more pharmaceutically acceptable excipients. Therefore, the present disclosure also covers the use of the nucleic acid molecule, vector or engineered immune cell in the preparation of a pharmaceutical composition or medicament.
  • the term "pharmaceutically acceptable excipient” means pharmacologically and/or physiologically compatible with the subject and the active ingredient (i.e., capable of eliciting the desired therapeutic effect without causing any adverse desired local or systemic effect), which are well known in the art (see, for example, Remington's Pharmaceutical Sciences. Edited by Gennaro AR, 19th ed. Pennsylvania: Mack Publishing Company, 1995).
  • Examples of pharmaceutically acceptable excipients include, but are not limited to, fillers, binders, disintegrants, coating agents, adsorbents, anti-adhesive agents, glidants, antioxidants, flavoring agents, coloring agents, Sweeteners, solvents, co-solvents, buffers, chelating agents, surfactants, diluents, wetting agents, preservatives, emulsifiers, coating agents, isotonic agents, absorption delaying agents, stabilizers and tonicity regulators .
  • suitable excipients is known to those skilled in the art to prepare the desired pharmaceutical compositions of the present disclosure.
  • excipients for use in pharmaceutical compositions of the present disclosure include saline, buffered saline, dextrose and water.
  • suitable excipients depends inter alia on the active agent used, the disease to be treated and the desired dosage form of the pharmaceutical composition.
  • compositions according to the present disclosure are suitable for a variety of routes of administration. Typically, administration is accomplished parenterally.
  • Parenteral delivery methods include topical, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.
  • the pharmaceutical composition according to the present disclosure can also be prepared in various forms, such as solid, liquid, gaseous or freeze-dried forms, especially ointments, creams, transdermal patches, gels, powders, tablets, solutions, gaseous In the form of aerosols, granules, pills, suspensions, emulsions, capsules, syrups, elixirs, extracts, tinctures or liquid extracts, or in a form especially adapted to the desired method of administration.
  • Processes known in this disclosure for the manufacture of pharmaceuticals may include, for example, conventional mixing, dissolving, granulating, dragee-making, milling, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions comprising immune cells such as those described herein are typically provided in solution, and preferably comprise a pharmaceutically acceptable buffer.
  • compositions according to the present disclosure may also be administered in combination with one or more other agents suitable for the treatment and/or prevention of the disease to be treated.
  • agents suitable for combination include known anticancer drugs such as cisplatin, maytansine derivatives, rachelmycin, calicheamicin, docetaxel, etoposide , gemcitabine, ifosfamide, irinotecan, melphalan, mitoxantrone, sorfimer sodium photofrin II, temozolomide, topotecan, trimetreate glucuronate, Auristatin E, vincristine, and doxorubicin; peptide cytotoxins, such as ricin, diphtheria toxin, Pseudomonas bacterial exotoxin A, DNase, and RNase; radionuclides, such as iodine 131, rhenium 186, indium 111, iridium 90, bismuth 210 and
  • the present disclosure also provides a method of treating a subject suffering from cancer, infection or autoimmune disease, comprising administering to the subject an effective amount of an immune cell or a pharmaceutical composition according to the present disclosure. Therefore, the present disclosure also covers the use of the engineered immune cells in the preparation of a medicament for the treatment of cancer, infection or autoimmune disease.
  • the method of treatment comprises administering to a subject an effective amount of an immune cell and/or a pharmaceutical composition of the present disclosure.
  • the immune cells are autologous or allogeneic cells, preferably T cells, macrophages, dendritic cells, monocytes, NK cells and/or NKT cells, more preferably T cells, NK cells cells or NKT cells.
  • autologous refers to any material derived from an individual that will later be reintroduced into that same individual.
  • allogeneic refers to any material derived from a different animal of the same species or a different patient as the individual into whom the material is introduced. Two or more individuals are considered allogeneic to each other when the genes at one or more loci differ. In some cases, allogeneic material from individuals of the same species may be genetically different enough for antigenic interactions to occur.
  • the term "subject" is a mammal. Mammals can be humans, non-human primates, mice, rats, dogs, cats, horses, or cows, but are not limited to these examples. Mammals other than humans can be advantageously used as subjects representing animal models of cancer. Preferably, the subject is a human.
  • the cancer is a cancer associated with expression of a target bound by the antigen binding region.
  • the cancers include, but are not limited to: brain glioma, blastoma, sarcoma, leukemia, basal cell carcinoma, biliary tract cancer, bladder cancer, bone cancer, brain and CNS cancers, breast cancer, peritoneal cancer, cervical cancer , choriocarcinoma, colon and rectal cancer, connective tissue cancer, cancer of the digestive system, endometrial cancer, esophageal cancer, eye cancer, head and neck cancer, gastric cancer (including gastrointestinal cancer), glioblastoma (GBM), Liver cancer, hepatoma, intraepithelial neoplasia, renal cancer, laryngeal cancer, liver tumors, lung cancer (such as small cell lung cancer, non-small cell lung cancer, adenoid lung cancer, and squamous lung cancer), lymphoma (including Hodgkin lymphoma and non-
  • the diseases that can be treated with the engineered immune cells or pharmaceutical compositions of the present disclosure are selected from: leukemia, lymphoma, multiple myeloma, brain glioma, pancreatic cancer, gastric cancer, and the like.
  • the infections include, but are not limited to, infections caused by viruses, bacteria, fungi, and parasites.
  • the autoimmune disease includes, but is not limited to, type 1 diabetes, celiac disease, Graves' disease, inflammatory bowel disease, multiple sclerosis, psoriasis, rheumatoid arthritis, Addison Illness, Sjogren's syndrome, Hashimoto's thyroiditis, myasthenia gravis, vasculitis, pernicious anemia and systemic lupus erythematosus, etc.
  • the method further comprises administering to the subject one or more additional chemotherapeutic agents, biologics, drugs or treatments.
  • the chemotherapeutic agent, biologic, drug or treatment is selected from radiation therapy, surgery, antibody agents and/or small molecules and any combination thereof.
  • MSCV-mCD19-CAR plasmid which contains CD19-scFv (SEQ ID NO: 2), CD8 ⁇ hinge region (SEQ ID NO: 17), CD8 ⁇ transmembrane region (SEQ ID NO: 5), 41BB co-stimulatory domain ( SEQ ID NO: 7) and the coding sequence of CD3 ⁇ intracellular region (SEQ ID NO: 11).
  • the MSCV-mCD19-CAR-IL7 plasmid was constructed, which further included the coding sequences of T2A (SEQ ID NO: 19) and IL7 (SEQ ID NO: 23) on the basis of the MSCV-mCD19-CAR plasmid.
  • the MSCV-mCD19-CAR-CCL20 plasmid was constructed, which further included the coding sequences of T2A (SEQ ID NO: 19) and CCL20 (SEQ ID NO: 25) on the basis of the MSCV-mCD19-CAR plasmid.
  • Opti-MEM Gibco, Cat. No. 31985-070
  • 45 ⁇ g of prepared retroviral plasmid 15 ⁇ g of packaging vector pCL-Eco (Shanghai Hewu Biotechnology Co., Ltd., Cat. No. P3029)
  • packaging vector pCL-Eco Shanghai Hewu Biotechnology Co., Ltd., Cat. No. P3029
  • 120 ⁇ l of X-treme GENE HP DNA transfection reagent (Roche, Cat. No. 06366236001)
  • the plasmid/vector/transfection reagent mixture was added dropwise into the pre-prepared 293T cell culture flask, and cultured overnight at 37°C and 5% CO 2 . Cultures were harvested 72 hours after transfection and centrifuged (2000g, 4°C, 10 minutes) to obtain retroviral supernatants.
  • T lymphocytes were isolated from mouse spleen, and T cells were activated with DynaBeads CD3/CD28 CTS TM (Gibco, Cat. No. 40203D), and then cultured at 37°C and 5% CO 2 for 1 day.
  • Activated T cells were inoculated into 24-well plates pre-coated overnight with RetroNectin at a density of 3 ⁇ 106 cells/mL per well, then 500 ⁇ L of retrovirus supernatant was added, and complete medium was supplemented to 2 mL.
  • the 24-well plate Place the 24-well plate in a centrifuge for centrifugal infection, and centrifuge at 2000g for 2h at 32°C. Then, immediately place the 24-well plate in a 37°C, CO2 incubator for static culture. Replace the fresh medium the next day, and adjust the cell density to 1 ⁇ 106 cells/mL. Three days after infection, cells were harvested for subsequent analysis. The collected cells are mCD19-CAR cells, mCD19-CAR+CCL20 cells and mCD19-CAR+IL7+CCL20 cells.
  • CAR-T cells Take out 2 ⁇ 10 5 CAR-T cells prepared in Example 1, use Goat Anti-Rat IgG (H&L) Biotin (BioVision, Cat. No. 6910-250) as the primary antibody, and APC Streptavidin (BD Pharmingen, Cat. No. 554067) as the secondary antibody , the expression level of CAR on CAR T cells was detected by flow cytometry, and the results are shown in Figure 1. It can be seen that compared with untreated NT cells, the CAR positive efficiency in mCD19-CAR cells, mCD19-CAR+CCL20 cells and mCD19-CAR+IL7+CCL20 cells were all greater than 60%, indicating that these cells can all be effective Express CAR.
  • H&L Goat Anti-Rat IgG
  • APC Streptavidin BD Pharmingen, Cat. No. 554067
  • the IFN- ⁇ level of CAR-T cells expressing the combination of IL7+CCL20 was also significantly higher than that of CAR-T cells expressing only CCL20, indicating that the addition of IL7 can further improve the killing activity of CAR-T cells.
  • Panc02-mCD19 pancreatic cancer cells were inoculated subcutaneously in the axilla of the left forelimb of healthy C57BL/6 mice.
  • the mice inoculated with pancreatic cancer cells were randomly divided into 3 groups, 5 mice in each group.
  • mice in each group were injected with 1 ⁇ 106 NT cells, mCD19-CAR cells or mCD19-CAR+IL7+CCL20 cells via tail vein. Monitor the mice for body weight and tumor volume changes until the end of the experiment.
  • mice The body weight changes of the mice are shown in Figure 5. It can be seen that after administration of CAR-T cells, the body weight of the mice in each group has no significant difference compared with the control group, indicating that the administration of CAR-T cells will not have obvious toxic side effects on the mice.
  • the present disclosure provides a novel engineered immune cell, which expresses a cell surface molecule that specifically recognizes an antigen and exogenous CCL20.
  • the engineered immune cell of the present disclosure further expresses exogenous IL7.
  • the engineered immune cells have inhibitory or therapeutic effects on cancer, infection or autoimmune diseases.

Landscapes

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

Abstract

L'invention concerne une cellule immunitaire modifiée exprimant une molécule de surface cellulaire reconnaissant spécifiquement un antigène et un CCL20 exogène. L'invention concerne également une utilisation de la cellule immunitaire modifiée dans le traitement de cancers, d'infections ou de maladies auto-immunes. Par comparaison avec les cellules immunitaires modifiées classiques, la cellule immunitaire modifiée présente une activité tumoricide significativement améliorée.
PCT/CN2022/113151 2021-08-26 2022-08-17 Cellule immunitaire modifiée et son utilisation WO2023025009A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110985593.1A CN115725504A (zh) 2021-08-26 2021-08-26 工程化免疫细胞及其用途
CN202110985593.1 2021-08-26

Publications (1)

Publication Number Publication Date
WO2023025009A1 true WO2023025009A1 (fr) 2023-03-02

Family

ID=85289892

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/113151 WO2023025009A1 (fr) 2021-08-26 2022-08-17 Cellule immunitaire modifiée et son utilisation

Country Status (2)

Country Link
CN (1) CN115725504A (fr)
WO (1) WO2023025009A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103228284A (zh) * 2010-08-09 2013-07-31 赛瓦克斯公司 用于预防病况的方法和组合物
CN107523586A (zh) * 2017-10-20 2017-12-29 广州万孚生物技术股份有限公司 免疫质粒及用于检测抗穆勒氏管激素的单抗、杂交瘤细胞及其制备方法和应用
US20180291089A1 (en) * 2015-04-30 2018-10-11 University Of Southern California Secretory tnt car cell immunotherapy
WO2019006467A1 (fr) * 2017-06-30 2019-01-03 Memorial Sloan Kettering Cancer Center Compositions et procédés pour la thérapie cellulaire adoptive
US20200215111A1 (en) * 2017-06-30 2020-07-09 Memorial Sloan Kettering Cancer Center Compositions and methods for adoptive cell therapy for cancer
CN112063588A (zh) * 2020-08-13 2020-12-11 南京北恒生物科技有限公司 工程化免疫细胞及其用途
WO2021034982A1 (fr) * 2019-08-21 2021-02-25 Board Of Regents, The University Of Texas System Cellules immunitaires pour thérapies cellulaires adoptives

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103228284A (zh) * 2010-08-09 2013-07-31 赛瓦克斯公司 用于预防病况的方法和组合物
US20180291089A1 (en) * 2015-04-30 2018-10-11 University Of Southern California Secretory tnt car cell immunotherapy
WO2019006467A1 (fr) * 2017-06-30 2019-01-03 Memorial Sloan Kettering Cancer Center Compositions et procédés pour la thérapie cellulaire adoptive
US20200215111A1 (en) * 2017-06-30 2020-07-09 Memorial Sloan Kettering Cancer Center Compositions and methods for adoptive cell therapy for cancer
CN107523586A (zh) * 2017-10-20 2017-12-29 广州万孚生物技术股份有限公司 免疫质粒及用于检测抗穆勒氏管激素的单抗、杂交瘤细胞及其制备方法和应用
WO2021034982A1 (fr) * 2019-08-21 2021-02-25 Board Of Regents, The University Of Texas System Cellules immunitaires pour thérapies cellulaires adoptives
CN112063588A (zh) * 2020-08-13 2020-12-11 南京北恒生物科技有限公司 工程化免疫细胞及其用途

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HAN YIPING: "Inhibitors of PI3K-AKT-mTOR Signaling Pathway and Cancer Immunotherapy", CHINESE JOURNAL OF CANCER BIOTHERAPY, PUBLISHING HOUSE OF CHINESE JOURNAL OF CANCER BIOTHERAPY, CN, vol. 24, no. 12, 31 December 2017 (2017-12-31), CN , pages 1424 - 1430, XP009543810, ISSN: 1007-385X *
LIU J Y, LI F, WANG L P, CHEN X F, WANG D, CAO L, PING Y, ZHAO S, LI B, THORNE S H, ZHANG B, KALINSKI P, ZHANG Y: "CTL- vs Treg lymphocyte-attracting chemokines, CCL4 and CCL20, are strong reciprocal predictive markers for survival of patients with oesophageal squamous cell carcinoma", BRITISH JOURNAL OF CANCER, NATURE PUBLISHING GROUP UK, LONDON, vol. 113, no. 5, 1 September 2015 (2015-09-01), London, pages 747 - 755, XP093039591, ISSN: 0007-0920, DOI: 10.1038/bjc.2015.290 *

Also Published As

Publication number Publication date
CN115725504A (zh) 2023-03-03

Similar Documents

Publication Publication Date Title
WO2022033537A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2021169977A1 (fr) Nouveau récepteur antigénique chimérique et utilisation associée
JP7462782B2 (ja) 改変免疫細胞とその使用
WO2022048523A1 (fr) Récepteur antigénique chimérique ciblant le récepteur activé par nk
WO2022166365A1 (fr) Nouveau récepteur antigénique chimérique et son utilisation
US20230242661A1 (en) Engineered immune cell expressing nk inhibitory molecule and use thereof
WO2022223049A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2022218226A1 (fr) Cellule immunitaire modifiée et son utilisation
US20230248768A1 (en) Engineered immune cell for allotransplantation
US20240009308A1 (en) Chimeric antigen receptor comprising novel co-stimulatory domain and use thereof
WO2023025009A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2023016284A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2023035947A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2023083003A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2023051361A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2023071811A1 (fr) Cellule immunitaire modifiée et son utilisation
WO2022267983A1 (fr) Cellule immunitaire modifiée et son utilisation
US20230270858A1 (en) Novel co-stimulatory domain and use thereof

Legal Events

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

Ref document number: 22860355

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE