WO2019109980A1 - Protéine chimère, et cellule effectrice immunitaire l'exprimant et application associée - Google Patents

Protéine chimère, et cellule effectrice immunitaire l'exprimant et application associée Download PDF

Info

Publication number
WO2019109980A1
WO2019109980A1 PCT/CN2018/119588 CN2018119588W WO2019109980A1 WO 2019109980 A1 WO2019109980 A1 WO 2019109980A1 CN 2018119588 W CN2018119588 W CN 2018119588W WO 2019109980 A1 WO2019109980 A1 WO 2019109980A1
Authority
WO
WIPO (PCT)
Prior art keywords
receptor
domain
acid sequence
amino acid
tgf
Prior art date
Application number
PCT/CN2018/119588
Other languages
English (en)
Chinese (zh)
Inventor
李宗海
王益
Original Assignee
科济生物医药(上海)有限公司
上海市肿瘤研究所
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 科济生物医药(上海)有限公司, 上海市肿瘤研究所 filed Critical 科济生物医药(上海)有限公司
Publication of WO2019109980A1 publication Critical patent/WO2019109980A1/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
    • A61K39/464474Proteoglycans, e.g. glypican, brevican or CSPG4
    • 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
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • 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
    • 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
    • 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
    • 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
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/53Liver
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment

Definitions

  • the present invention belongs to the field of adoptive cell therapy; in particular, the present invention relates to a chimeric protein and a genetically engineered immune effector cell, which can significantly enhance the proliferation and survival of immune cells and enhance their cell killing ability. .
  • adoptive cell therapy such as CAR-T therapy and TCR-T therapy have shown good results in the field of hematoma, but the treatment of solid tumors is not good, because cancer cells in solid tumors can be around them.
  • a tumor microenvironment is formed to support the growth and metastasis of cancer cells.
  • Immunosuppressive cytokines such as IL-4, IL-10, TGF- ⁇ , etc., which are abundantly expressed in the tumor microenvironment, inhibit the antitumor activity of CAR-T cells.
  • TGF- ⁇ also induces T cell differentiation into regulatory T cells (Treg), and regulatory T cells further inhibit T cell killing.
  • the object of the present invention is to significantly enhance the ability of immune cells to proliferate and survive, and to enhance their cell killing ability, and the object of the present invention is achieved by a specific chimeric protein and genetically engineered immune effector cells.
  • the invention provides an immune effector cell expressing a chimeric protein comprising an extracellular domain of a TGF-beta receptor and an intracellular signal domain of a receptor for an IL-2 family protein.
  • the immune effector cell further expresses a chimeric receptor capable of recognizing a target antigen; preferably, the chimeric receptor is a chimeric antigen receptor (CAR), a T cell receptor (TCR) a T cell fusion protein (TFP), or a T cell antigen coupler (TAC); more preferably, the chimeric receptor is a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • TCR T cell receptor
  • TFP T cell fusion protein
  • TAC T cell antigen coupler
  • the chimeric receptor is a chimeric antigen receptor (CAR).
  • the extracellular domain of the TGF-beta receptor comprises an extracellular domain of TGF-beta receptor I and/or an extracellular domain of TGF-beta receptor II; preferably a cell of TGF-beta receptor II Outland.
  • the receptor intracellular signal domain of the IL-2 family protein is selected from the group consisting of an IL-2 receptor, an IL-4 receptor, an IL-7 receptor, an IL-9 receptor, and an IL- An intracellular signal domain of any of the 15 receptors, IL-21 receptors, or a combination of at least two;
  • an intracellular signal domain selected from any of the IL-2 receptor, the IL-7 receptor, and the IL-21 receptor;
  • the receptor intracellular signal domain of the IL-2 family protein is the intracellular signal domain of the IL-7 receptor or the intracellular signal domain of the IL-21 receptor.
  • the receptor intracellular signal domain of the IL-2 family protein comprises an intracellular domain of IL-2RG.
  • the receptor intracellular signal domain of the IL-2 family protein comprises an intracellular domain of IL-2R ⁇ , IL-7R or IL-21R.
  • the receptor intracellular domain of the IL-2 family protein comprises the intracellular domain of IL-2RG, and the intracellular domain of IL-2R ⁇ , IL-7R, or IL-21R.
  • the chimeric protein comprises an extracellular domain of TGF-beta receptor II and an intracellular signal domain of the alpha subunit of a receptor of any of the IL-2 family proteins;
  • the alpha subunit of the receptor of the IL-2 family protein is selected from the group consisting of IL-2R ⁇ , IL-7R, or IL-21R; more preferably IL-7R or IL-21R.
  • the extracellular domain and the intracellular signal domain have a transmembrane domain; preferably, the transmembrane domain is a transmembrane domain of a receptor for an IL-2 family protein.
  • the chimeric protein comprises a first chimeric protein and a second chimeric protein
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular domain of an intracellular signal domain selected from the intracellular signal domain of IL-2RG or the alpha subunit of a receptor of an IL-2 family protein ;with
  • the second chimeric protein comprises an extracellular domain of TGF-beta receptor II and an intracellular domain of an intracellular signal domain selected from the intracellular signal domain of IL-2RG or the alpha subunit of a receptor of an IL-2 family protein .
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular signal domain of IL-2RG;
  • the second chimeric protein comprises an extracellular domain of TGF-beta receptor II and an intracellular signal domain of the alpha subunit of the receptor of the IL-2 family protein.
  • the intracellular signal domain of the alpha subunit of the receptor for the IL-2 family protein is selected from the intracellular signal domain of IL-2R ⁇ , IL-7R, or IL-21R.
  • the extracellular domain of the TGF-beta receptor and the intracellular signal domain of the receptor of the IL-2 family protein are wild-type or mutant.
  • the amino acid sequence of the extracellular domain of TGF-beta receptor I has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:4;
  • the amino acid sequence of the extracellular domain of TGF-beta receptor II has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 12;
  • the amino acid sequence of the intracellular domain of IL-2RG has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 8.
  • the amino acid sequence of the intracellular domain of IL-2R ⁇ has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 16;
  • amino acid sequence of the intracellular domain of IL-7R has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:20;
  • the amino acid sequence of the intracellular domain of IL-21R has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 24.
  • the amino acid sequence of the first chimeric protein has at least 90% homology with the amino acid sequence set forth in SEQ ID NO: 27; the amino acid sequence of the second chimeric protein is The amino acid sequence set forth in any one of SEQ ID NO: 28, 29, or 30 has at least 90% homology.
  • the immune effector cell is selected from any one of T cells, natural killer cells, natural killer T cells, DNT cells, mast cells, or bone marrow-derived phagocytic cells, or a combination thereof; preferably The immune effector cells are selected from the group consisting of T cells.
  • the target antigen is a tumor antigen or a pathogenic microorganism antigen.
  • the target antigen is a pathogenic microorganism antigen, the pathogenic microorganism including a virus, a bacterium, a fungus, a protozoa or a parasite; more preferably, the pathogenic microorganism is a virus; or more preferably, The pathogenic microorganism is selected from the group consisting of a cytomegalovirus, an Epstein-Barr virus, a human immunodeficiency virus, and an influenza virus;
  • the target antigen is a tumor antigen, and preferably, the tumor antigen comprises:
  • Prostate-specific membrane antigen PSMA
  • carcinoembryonic antigen CEA
  • IL13Ralpha HER-2
  • CD19 NY-ESO-1
  • HIV-1 Gag Lewis Y, MART-1, gp100, tyrosinase, WT- I, hTERT, mesothelin, EGFR, EGFRvIII, phosphatidylinositol 3, EphA2, HER3, EpCAM, MUC1, MUC16, CLDN18.2, folate receptor, CLDN6, CD30, CD138, ASGPR1, CDH16, GD2 , 5T4, 8H9, ⁇ v ⁇ 6 integrin, B cell mature antigen (BCMA), B7-H3, B7-H6, CAIX, CA9, CD20, CD22, kappa light chain, CD33, CD38, CD44, CD44v6, CD44v7/8, CD70, CD123, CD171, CSPG4, EGP2, EGP40, ERBB3,
  • the target antigen is an antigen of a solid tumor.
  • amino acid sequence of the extracellular antigen recognition domain of the chimeric antigen receptor has at least 90% homology to the sequence set forth in any one of SEQ ID NOs: 31-35.
  • the present invention provides a pharmaceutical composition comprising the immune effector cell of the first aspect.
  • the present invention provides the use of the immune effector cell of the first aspect or the pharmaceutical composition of the second aspect for the preparation of a medicament for preventing or treating a tumor or a pathogenic microorganism infection.
  • the invention provides a chimeric protein comprising an extracellular domain of a TGF-beta receptor and an intracellular signal domain of a receptor for an IL-2 family protein.
  • the extracellular domain of the TGF-beta receptor comprises an extracellular domain of TGF-beta receptor I and/or an extracellular domain of TGF-beta receptor II, preferably a cell of TGF-beta receptor II Outland;
  • the receptor intracellular signal domain of the IL-2 family protein is selected from the group consisting of an IL-2 receptor, an IL-4 receptor, an IL-7 receptor, an IL-9 receptor, and an IL-
  • the intracellular signal domain of any of the receptors; more preferably, the receptor intracellular signal domain of the IL-2 family protein is the intracellular signal domain of the IL-7 receptor or the cell of the IL-21 receptor Internal signal domain.
  • the chimeric protein comprises the extracellular domain of TGF-beta receptor II and the intracellular signal domain of the alpha subunit of the receptor of any of the IL-2 family proteins.
  • the alpha subunit of the receptor for the IL-2 family protein is selected from the group consisting of IL-2R ⁇ , IL-7R, or IL-21R; preferably, IL-7R or IL-21R.
  • the chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular signal domain of IL-2RG.
  • the extracellular domain of the TGF-beta receptor and the intracellular signal domain of the receptor of the IL-2 family protein are wild-type or mutant.
  • the amino acid sequence of the extracellular domain of TGF-beta receptor I has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:4;
  • the amino acid sequence of the extracellular domain of TGF-beta receptor II has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:12.
  • the amino acid sequence of the intracellular domain of IL-2RG has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:8.
  • the amino acid sequence of the intracellular domain of IL-2R ⁇ has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 16;
  • amino acid sequence of the intracellular domain of IL-7R has at least 90% homology to the amino acid sequence set forth in SEQ ID NO:20;
  • the amino acid sequence of the intracellular domain of IL-21R has at least 90% homology to the amino acid sequence set forth in SEQ ID NO: 24.
  • the extracellular domain and the intracellular signal domain have a transmembrane domain
  • the transmembrane domain is a transmembrane domain of a receptor for an IL-2 family protein.
  • the amino acid sequence of the chimeric protein has at least 90% homology to the amino acid sequence set forth in any one of SEQ ID NO: 27, 28, 29, or 30;
  • amino acid sequence of the chimeric protein is as set forth in any one of SEQ ID NOs: 27, 28, 29, or 30.
  • the chimeric protein comprises a first chimeric protein and a second chimeric protein
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular domain of an intracellular signal domain selected from the intracellular signal domain of IL-2RG or the alpha subunit of a receptor of an IL-2 family protein ;with
  • the second chimeric protein comprises an extracellular domain of TGF-beta receptor II and an intracellular domain of an intracellular signal domain selected from the intracellular signal domain of IL-2RG or the alpha subunit of a receptor of an IL-2 family protein .
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular signal domain of IL-2RG;
  • the second chimeric protein comprises an extracellular domain of TGF-beta receptor II and an intracellular signal domain of the alpha subunit of the receptor of the IL-2 family protein.
  • Figure 1 is a schematic diagram showing the structure of a chimeric protein.
  • Figure 2A is a plasmid map of plasmid 1 expressing the first chimeric protein and IL-2R ⁇
  • Figure 2B is a plasmid map of plasmid 2 expressing the first chimeric protein and IL-7RA
  • Figure 2C is the expression of the first chimeric protein and Plasmid map of plasmid 3 of IL-21R.
  • Figure 3 shows the results of Western blot analysis of STAT3/5 phosphorylation levels.
  • Figure 4A is a Foxp3 flow pattern in the CD4+CD25+ population and Figure 4B is a histogram representation of the Foxp3+ population ratio.
  • Figure 5 shows the sustained killing ability of chTR7/21 cells against tumor cells under TGF- ⁇ stimulation.
  • Figure 6 is a flow diagram of the preparation of Huh7-TGF ⁇ .
  • Figure 7 shows the results of in vitro killing when the target cells are Huh7 and SK-hep1-GPC3 cells.
  • Figure 8 shows the in vitro killing results when the target cells are Huh7-TGF ⁇ and SK-hep1-GPC3-TGF ⁇ .
  • the present invention can be completed not only by inhibiting TGF- ⁇ in the tumor microenvironment but also by increasing the proliferation and survival levels of immune cells.
  • immune effector cells refers to cells involved in an immune response, for example, to promote an immune effector response.
  • immune effector cells include T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, DNT cells, mast cells, and bone marrow-derived phagocytic cells.
  • T cells may be T cells directly derived from peripheral blood, or may be subtypes of T cells, such as CD8+ T cells, CD4+ T cells, ⁇ / ⁇ T cells, ⁇ / ⁇ T cells, and the like.
  • CAR chimeric antigen receptor
  • CAR refers to a group of polypeptides which, when administered in an immune effector cell, confer specificity to a target cell target antigen (eg, a tumor antigen) and have cells.
  • the internal signal is generated.
  • CAR typically includes at least one extracellular antigen binding domain, a transmembrane domain (also known as a transmembrane domain), and a cytoplasmic signaling domain (also referred to herein as an "intracellular domain”).
  • signaling domain refers to a functional portion of a protein that functions by transmitting information within a cell for regulating cells via a defined signaling pathway by generating a second messenger or by acting as an effector in response to such a messenger. Activity.
  • TCR T cell receptor
  • TCR a characteristic marker on the surface of all T cells, binds to CD3 with a non-covalent bond to form a TCR-CD3 complex.
  • the role of TCR is to recognize antigens.
  • TCR is a heterodimer composed of two different peptide chains, consisting of two peptide chains, ⁇ and ⁇ . Each peptide chain can be further divided into variable region (V region), constant region (C region), transmembrane. The region and the cytoplasmic region are several parts; it is characterized by a short cytoplasmic region.
  • the TCR molecule belongs to the immunoglobulin superfamily, and its antigen specificity exists in the V region; the V region (V ⁇ , V ⁇ ) has three hypervariable regions CDR1, CDR2, and CDR3, among which the CDR3 mutation is the largest, which directly determines the TCR antigen. Binding specificity. When the TCR recognizes the MHC-antigen peptide complex, CDR1, CDR2 recognizes and binds to the side wall of the MHC molecule antigen binding groove, and CDR3 binds directly to the antigen peptide.
  • TCR is divided into two categories: TCR1 and TCR2; TCR1 consists of two chains of ⁇ and ⁇ , and TCR2 consists of two chains of ⁇ and ⁇ .
  • T Cell Fusion Protein includes various polypeptide-derived recombinant polypeptides that constitute a TCR, which are capable of i) binding to a surface antigen on a target cell, and ii) complex TCR complexes
  • TFP consists of an antigen binding domain consisting of a TCR subunit and a human or humanized antibody domain, wherein the TCR subunit comprises at least a portion of the TCR extracellular domain, the transmembrane domain, and the TCR intracellular domain.
  • the TCR subunit and the antibody domain are operably linked, wherein the extracellular, transmembrane, intracellular signal domain of the TCR subunit is derived from CD3 epsilon or CD3 gamma, and the TFP is integrated TCR expressed on T cells.
  • an “effective link” is meant a functional link between a regulatory sequence and a heterologous nucleic acid sequence that results in expression of a heterologous nucleic acid sequence.
  • the first nucleic acid sequence is located in a functional regulatory region of the second nucleic acid sequence, the first nucleic acid sequence is operably linked to the second nucleic acid sequence.
  • this promoter can be operably linked to the coding sequence.
  • T Cell Antigen Coupler includes three functional domains: a tumor targeting domain, including a single chain antibody, a designed ankyrin repeat protein (DARPin). Or other targeting group; 2 extracellular domain domain, is a single-chain antibody that binds to CD3, such that the TAC receptor is adjacent to the TCR receptor; 3 the transmembrane region and the intracellular region of the CD4 co-receptor, wherein The intracellular domain is linked to the protein kinase LCK, which catalyzes the phosphorylation of immunoreceptor tyrosine activation motifs (ITAMs) of the TCR complex as an initial step in T cell activation.
  • ITAMs immunoreceptor tyrosine activation motifs
  • tumor refers to the growth and proliferation of all neoplastic cells, whether malignant or benign, as well as all precancerous and cancerous cells and tissues.
  • tumor microenvironment refers to any and all elements of the tumor environment, including elements that create a structural and/or functional environment for a malignant process to survive and/or expand and/or spread.
  • TGF- ⁇ and TGFb have the same meaning herein, and the term "extracellular domain of TGF-beta receptor” includes the extracellular domain of the native TGF-beta receptor and the extracellular domain of the mutant TGF-beta receptor. .
  • sequence of the extracellular domain of a "native” or “wild-type” TGF-beta receptor primarily refers to the extracellular domain sequence of a human TGF-beta receptor, whether purified from a natural source or using recombinant techniques.
  • the extracellular domain of the TGF-beta receptor comprises a fragment that may be shorter or longer than the extracellular domain protein of the native TGF-beta receptor, as long as the extracellular domain protein fragment of the TGF-beta receptor remains The ability to bind TGF- ⁇ . It is also to be understood that the disclosure encompasses nucleic acid molecules encoding TGF-[beta] receptors described herein or known in the art, including, but not limited to, RNA sequences corresponding to the DNA sequences described herein.
  • TGF- ⁇ receptor II also referred to herein as TGFbRII
  • TGFbRII TGF- ⁇ receptor II
  • TGFbRI TGF- ⁇ receptor I
  • IL-2 family protein includes IL-2, IL-4, IL-7, IL-9, IL-15, IL-21, etc., and the "receptor of IL-2 family protein” contains ⁇ .
  • the chain (IL-2RG, also referred to herein as IL2RG, whose sequence is set forth in SEQ ID NO: 43) acts as a cytokine for its receptor subunit, and is also shared by each specific R ⁇ (eg, IL-2 and IL-15).
  • IL-2R ⁇ also referred to herein as IL2RB
  • IL-7R also referred to herein as IL7R
  • IL-21R also referred to herein as IL21R
  • IL-2RG shared gamma chain subunit
  • antigen refers to a molecule that can provoke an immune response.
  • the immune response can involve the production of antibodies, or activation of specific immunocompetent cells, or both.
  • any macromolecule including substantially all proteins or peptides, can serve as an antigen.
  • the antigen may be derived from recombinant DNA or genomic DNA. It will be apparent to those skilled in the art that any DNA comprising a nucleotide sequence or a partial nucleotide sequence encoding a protein that can elicit an immune response, thus encoding an "antigen" (as the term is used herein).
  • the antigen need not be encoded only by the full length nucleotide sequence of the gene. It will be readily apparent that the invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene, which may be arranged in various combinations to encode a polypeptide that elicits a desired immune response. Furthermore, it will be apparent to those skilled in the art that the antigen need not be encoded by a "gene”. It will be readily apparent that the antigen may be produced synthetically, or may be derived from a biological sample, or may be a macromolecule other than a polypeptide.
  • the biological sample can include, but is not limited to, a tissue sample, a tumor sample, a cell or a liquid, and other biological components.
  • expression refers to the transcription and/or translation of a particular nucleotide sequence driven by a promoter.
  • Lentivirus refers to the genus of the family Lentiviridae. Lentiviruses are unique in retroviruses and are capable of infecting non-dividing cells; they are capable of delivering a significant amount of genetic information into the DNA of a host cell, whereby they are one of the most efficient methods of gene delivery vectors. HIV, SIV and FIV are all examples of lentiviruses.
  • homologous or identical refers to between two polymer molecules, for example between two nucleic acid molecules, for example between two DNA molecules or two RNA molecules, or between two polypeptide molecules.
  • Unit sequence identity When a subunit position is occupied by the same monomer subunit in two molecules, for example when two DNA molecules are occupied by adenosine at one position, they are homologous or identical at that position.
  • the homology between two sequences is a direct function of the number of matched or homologous positions; for example, when half of the positions in the two sequences (for example, 5 positions in a polymer of 10 subunits) are homologous
  • the two sequences are 50% homologous; if 90% of the positions (eg, 9 out of 10) are matched or homologous, then the two sequences are 90% homologous.
  • transfected or “transformed” or “transduced” refers to the process of transferring or introducing an exogenous nucleic acid into a host cell.
  • a “transfected” or “transformed” or “transduced” cell is one that has been transfected, transformed or transduced with an exogenous nucleic acid.
  • the cells include primary test cells and their progeny.
  • the present invention provides a chimeric protein comprising an extracellular domain of a TGF-beta receptor and an intracellular signal domain of a receptor for an IL-2 family protein. Co-expression of this chimeric protein and a receptor recognizing the target antigen on immune effector cells can inhibit TGF- ⁇ in the tumor microenvironment and increase the proliferation and survival of immune cells.
  • the extracellular domain of the TGF-beta receptor is the extracellular domain of TGF-beta receptor I and/or the extracellular domain of TGF-beta receptor II.
  • the receptor intracellular signal domain of the IL-2 family protein is selected from the group consisting of an IL-2 receptor, an IL-4 receptor, an IL-7 receptor, an IL-9 receptor, An intracellular signal domain of any of the IL-15 receptor, IL-21 receptor, or a combination of at least two; preferably, selected from the group consisting of an IL-2 receptor, an IL-7 receptor, and an IL-21
  • the intracellular signal domain of any of the receptors; more preferably, the receptor intracellular signal domain of the IL-2 family protein is the intracellular signal domain of the IL-7 receptor or the IL-21 receptor Intracellular signal domain.
  • the extracellular domain of the TGF-beta receptor is preferably the extracellular domain of TGF-beta receptor II
  • the receptor intracellular signal domain of the IL-2 family protein is the IL-2 family.
  • the intracellular signal domain of the alpha subunit of the protein receptor is selected from the group consisting of IL-2R ⁇ , IL-7R, or IL-21R; preferably IL-7R or IL-21R.
  • polypeptides having certain amino acid sequence identity or homology to the extracellular domain or intracellular domain described above may also have the same or similar functions.
  • the amino acid sequence of the intracellular domain of 2R ⁇ , the intracellular domain of IL-7R, and the intracellular domain of IL-21R is SEQ ID NO: 4, SEQ ID NO: 12, and SEQ ID NO: 8, respectively.
  • amino acid sequence set forth in SEQ ID NO: 16, SEQ ID NO: 20, SEQ ID NO: 24 has at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% Or 99% homology; and the amino acid sequence of the chimeric protein of the invention has at least 90%, 91%, 92%, 93 with the amino acid sequence set forth in any one of SEQ ID NO: 27, 28, 29, or %, 94%, 95%, 96%, 97%, 98% or 99% homology.
  • the chimeric proteins of the invention also have a transmembrane domain between the extracellular domain and the intracellular signal domain.
  • the "transmembrane domain” may include a transmembrane domain of a plurality of natural receptor proteins, as long as it functions to connect the extracellular and intracellular regions of the receptor and anchor to the cell membrane, and thus, the incorporation of the present invention
  • the transmembrane domain in the protein is not restricted to any particular transmembrane domain, including but not limited to the TGF-beta receptor, the transmembrane region of the receptor of the IL-2 family protein; preferably, the transmembrane domain is The transmembrane domain of the receptor for the IL-2 family of proteins.
  • the chimeric protein of the present invention comprises the first chimeric protein and the second chimeric protein, it can have more excellent technical effects.
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and a cell selected from the intracellular signal domain of IL-2RG or the alpha subunit of the receptor of the IL-2 family protein An intracellular domain of an internal signal domain; said second chimeric protein comprising an extracellular domain of TGF-beta receptor II and an alpha subunit selected from the group consisting of an intracellular signal domain of IL-2RG or a receptor of an IL-2 family protein The intracellular domain of the intracellular signal domain.
  • the first chimeric protein comprises an extracellular domain of TGF-beta receptor I and an intracellular domain of IL-2RG;
  • the second chimeric protein comprises an extracellular domain of TGF-beta receptor II and IL-2
  • CAR Chimeric Antigen Receptor
  • Chimeric antigen receptors typically comprise a (fine) extracellular antigen binding region.
  • the extracellular antigen binding region can be fully human. In other cases, the extracellular antigen binding region can be humanized. In other instances, the extracellular antigen binding region can be murine or the chimera in the extracellular antigen binding region consists of amino acid sequences from at least two different animals. In some embodiments, the extracellular antigen binding region can be non-human.
  • the extracellular antigen binding region comprises a hinge or spacer.
  • the terms hinge and spacer are used interchangeably.
  • the hinge can be considered as part of a CAR for providing flexibility to the extracellular antigen binding region.
  • the hinge can be used to detect CAR on the cell surface of a cell, particularly when detecting antibodies to the extracellular antigen binding region are ineffective or available.
  • the length of the hinge derived from an immunoglobulin may need to be optimized, depending on the location of the extracellular antigen binding region that targets the epitope on the target.
  • the transmembrane domain of CAR can anchor the CAR to the plasma membrane of the cell, such as the transmembrane domain of CD8, the transmembrane domain of CD28, and the like.
  • the skilled person can replace it according to the known transmembrane domain.
  • the intracellular signal domain of CAR may be responsible for activating at least one of the effector functions of the immune response cells into which the CAR has been placed.
  • CAR can induce effector functions of T cells, for example, the effector function is cytolytic activity or helper activity, including secretion of cytokines.
  • the term "intracellular signal domain” refers to a portion of a protein that transduces an effector function signal and directs the cell to perform a specific function. Although the entire intracellular signaling region can generally be used, in many cases it is not necessary to use the entire chain of the signal domain. In some cases, a truncated portion of the intracellular signaling region is used. In some instances, the term “intracellular signal domain” includes any truncated portion of an intracellular signaling region sufficient to transduce an effector function signal.
  • the intracellular signaling domain of CAR can be selected from any of the domains of Table 1.
  • the intracellular signaling region of CAR may further comprise one or more costimulatory domains.
  • the intracellular signaling region may comprise a single costimulatory domain, such as an ⁇ chain (first generation CAR) or it is with CD28 or 4-1BB (second generation CAR).
  • the intracellular signaling region can comprise two costimulatory domains, such as CD28/OX40 or CD28/4-1BB (third generation).
  • signals generated by the CAR may be combined with an auxiliary or costimulatory signal.
  • costimulatory signaling domains chimeric antigen receptor-like complexes can be designed to contain several possible costimulatory signal domains.
  • T cell activation Several receptors have been reported to provide co-stimulation for T cell activation including, but not limited to, CD28, OX40, CD27, CD2, CD5, ICAM-1, LFA-1 (CD11a/CD18), 4-1BBL, MyD88, and 4- 1BB.
  • the signaling pathways used by these costimulatory molecules work synergistically with the primary T cell receptor activation signal.
  • the signals provided by these costimulatory signaling regions can act synergistically with primary effect activation signals derived from one or more ITAM motifs (eg, the CD3zeta signal transduction domain) and can fulfill the requirements for T cell activation.
  • ITAM motifs eg, the CD3zeta signal transduction domain
  • Chimeric proteins or immune effector cells expressing chimeric proteins can be analyzed using standard methods known in the art or described herein.
  • Immune effector cells comprising a chimeric protein as described herein and a receptor that recognizes a target antigen can be used in a variety of therapeutic applications.
  • the immune effector cells described herein can be used in the treatment or prevention of any disease, disorder, or condition involving a cell that expresses TGF- ⁇ and that would benefit from inhibition of cell proliferation or promotion of cell death. In some embodiments, it can be used to induce apoptosis or cell death, or to treat disorders associated with abnormal apoptosis or cell proliferation, such as cancer.
  • cancer refers to a cell that has the ability to grow automatically (eg, an abnormal state or condition characterized by a proliferation of cells that are proliferating). Hyperproliferative or neoplastic disease states can be classified as pathological types (eg, because they deviate from normal but are not associated with disease states). Thus, “cancer” or “tumor” refers to any unwanted growth of a cell that has no physiological function.
  • cancer includes cell growth that is technically benign but may present a risk of becoming malignant.
  • Malignant refers to the abnormal growth of any cell type or tissue.
  • malignant includes cell growth that is technically benign but at risk of becoming malignant.
  • the term also includes any cancer, cancer, neoplasm, tumor formation or tumor. Thus, these terms are meant to include all types of cancer growth or tumorigenesis processes, metastatic tissues or malignant transformed cells, tissues or organs, whether of histopathological type or invasive stage.
  • cancer which is the majority of cancers and epidermal cells or covers organs, glands, or other body structures (eg, skin, uterus, lung cancer, breast cancer, prostate cancer). , cancer of the outer or inner surface of the stomach, intestines, and often metastasis; sarcoma, which is derived from connective tissue or supporting tissue (eg, bone, cartilage, tendons, ligaments, fat, muscle); and blood Tumors, which are derived from bone marrow and lymphoid tissues.
  • connective tissue or supporting tissue eg, bone, cartilage, tendons, ligaments, fat, muscle
  • Tumors which are derived from bone marrow and lymphoid tissues.
  • Examples of cancer include, but are not limited to, cancer, sarcoma, and hematological tumor formation disorders such as leukemia.
  • the cancer can be an adenocarcinoma (which is typically in an organ or gland that can be secreted, such as the breast, lung, colon, prostate, or bladder), or can be a squamous cell carcinoma (which is derived from the squamous epithelium and is generally large in the body). Part of the area is formed).
  • adenocarcinoma which is typically in an organ or gland that can be secreted, such as the breast, lung, colon, prostate, or bladder
  • a squamous cell carcinoma which is derived from the squamous epithelium and is generally large in the body. Part of the area is formed).
  • Sarcoma can be osteosarcoma or osteogenic sarcoma (bone), chondrosarcoma (cartilage), leiomyosarcoma (smooth muscle), striated muscle (skeletal muscle), mesothelioma or mesothelioma (membranous lining of body cavity), fibrosarcoma (fibrous tissue), angiosarcoma or hemangioendothelial blood vessels), liposarcoma (fat), glioma or astrocytoma (found in the brain's neurogenic connective tissue), mucinous sarcoma (primary embryonic connective tissue) or between Leaf cell tumor or mesodermal mixed tumor (mixed connective tissue type).
  • Hematopoietic tumor-forming disorders include proliferative/neoplastic cells involved in the origin of hematopoiesis, for example, derived from the myeloid, lymphoid or erythroid cell lines or their precursor cells.
  • the disease is derived from poorly differentiated acute leukemia (eg, erythroblastic leukemia and acute megakaryoblastic leukemia).
  • Additional exemplary myeloid disorders include, but are not limited to, acute promyelocytic leukemia (APML), acute myeloid leukemia (AML), and chronic myelogenous leukemia (CML); lymphoid malignancies include, but are not limited to, acute lymphoblastic leukemia (ALL), including B-line acute lymphoblastic leukemia and T-line acute lymphoblastic leukemia, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia, and Waldens Waldenstrom's macroglobulinemia.
  • ALL acute lymphoblastic leukemia
  • ALL including B-line acute lymphoblastic leukemia and T-line acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • hairy cell leukemia and Waldens Waldenstrom's macroglobulinemia.
  • malignant lymphoma include, but are not limited to, non-Hodgkin's lymphoma and its variants, peripheral T-cell lymphoma, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymph Cellular leukemia (LGF), Hodgkin's disease, and Ris-Sick disease.
  • non-Hodgkin's lymphoma and its variants include, but are not limited to, non-Hodgkin's lymphoma and its variants, peripheral T-cell lymphoma, adult T-cell leukemia/lymphoma (ATL), cutaneous T-cell lymphoma (CTCL), large granular lymph Cellular leukemia (LGF), Hodgkin's disease, and Ris-Sick disease.
  • cancers can also be named according to the organ from which the cancer is derived, ie, the "primary site", such as the breast, brain, lung, liver, skin, prostate, testis, bladder, colon and rectum, cervix, uterus, and the like. This name is adhered to even if the cancer is transferred to another part of the body that is different from the original site.
  • Cancers named according to the primary site can be associated with histological classification.
  • lung cancer is generally small cell lung cancer and non-small cell lung cancer, which may be squamous cell carcinoma, adenocarcinoma, large cell carcinoma; skin cancer is usually basal cell carcinoma, squamous cell carcinoma or melanoma. Lymphoma may occur in lymph nodes associated with the head, neck, and chest as well as in abdominal lymph nodes or axillary or inguinal lymph nodes.
  • Solid tumor refers to tumors and/or metastases other than lymphoma (wherever they are), such as the brain or other central nervous system tumors (eg meningioma, brain tumor, myeloma, cranial neuroma, and central nervous system) Other parts of the tumor, such as malignant glioma or medulloblastoma; head and / or neck cancer; breast tumors; circulatory tumors (such as the heart, mediastinum and pleura, and other organs in the thoracic tumor, hemangiomas and Tumors associated with vascular tissue; excretory system tumors (eg, kidney, renal pelvis, ureter, bladder, other and urinary organs not specifically indicated); gastrointestinal tumors (eg, esophagus, stomach, small intestine, colon, colorectum, recto sigmoid colon) Combination point, rectum, anus, anal canal), other liver and intrahepatic bile duct, gallbladder,
  • metastasis of the primary organ or tissue and/or any other site is also indicated, either alone or simultaneously, regardless of where the tumor and/or metastases are located. .
  • compositions in accordance with the present disclosure may comprise the immune effector cells provided herein and one or more non-toxic pharmaceutically acceptable carriers, diluents, excipients, and adjuvants. These compositions may be suitable for use in the treatment of the therapeutic indications described herein.
  • the immune effector cells can be administered in a therapeutically effective amount to treat one or more cancers or in combination with other therapies.
  • the immune effector cells can be administered before, during or after treatment with an anti-tumor or other therapy.
  • An "anti-cancer treatment” is a compound, composition or treatment (eg, surgery) that prevents or delays the growth and/or metastasis of cancer cells.
  • anti-cancer therapies include, but are not limited to, surgery (eg, removal of all or part of a tumor), chemotherapy drug therapy, radiation, gene therapy, hormone control, immunotherapy (eg, therapeutic antibodies and cancer vaccines), and antisense or RNAi Oligonucleotide treatment.
  • chemotherapeutic agents include, but are not limited to, hydroxyurea, busulphan, cisplatin, carboplatin, chlorambucil, melphalan, cyclophosphamide, ifosfamide, daunubicin ), doxorubicin, epirubicin, mitoxantrone, vincristine, vinblastine, vinorelbine, etoposide, teniposide, paclitaxel, docetaxel, gemcitabine, cytosine, arabinose Cytidine, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C, avastin, fluorouracil, temozolamide, and the like.
  • the immune effector cells are also suitable for use by standard combination therapies using two or more chemotherapeutic drugs. It should be understood that anti-cancer therapies include new compounds or treatments developed in the future.
  • the immune effector cells can also be used in combination with a radiation sensitizer such as a radiotherapy sensitizer.
  • a sensitizer is any agent capable of increasing the activity of the immune effector cells.
  • a sensitizer will increase the ability of the fusion protein to inhibit cancer cell growth or kill cancer cells.
  • exemplary sensitizers include antibodies against IL-10, bone morphogenetic proteins, and HDAC inhibitors (see, for example, Sakariassen et al., Neoplasia 9(11): 882-92, 2007).
  • the immune effector cells may be used as part of a neoadjuvant therapy (to primary therapy) as part of an adjuvant therapy regimen in which the goal is to cure cancer in the subject.
  • the immune effector cells can also be administered at different stages of tumorigenesis and progression, including in advanced and/or invasive neoplasms (eg, by topical treatment in a subject (eg, a dominant disease that cannot be cured by surgery or radiotherapy) , metastatic disease.
  • Local phase disease and/or refractory tumor eg, treatment of cancer or tumor that does not respond to treatment
  • Primary therapy refers to the initial diagnosis of cancer in a subject. First-line treatment.
  • Exemplary primary therapy may involve surgery, a wide range of chemotherapy, and radiation therapy.
  • “Auxiliary therapy” refers to a therapy that follows a primary therapy and is administered at a risk of recurrence.
  • the adjuvant systemic treatment begins soon after the primary therapy, for example at 2, 3, 4, 5, or 6 weeks after the last primary therapy treatment to delay recurrence, prolong survival or cure the subject.
  • the immune effector cells can be used alone or as part of an adjuvant therapy with one or more Other chemotherapeutic drugs are used in combination.
  • the combination of the immune effector cells and standard chemotherapeutic agents can enhance the efficacy of chemotherapy and, therefore, can be used to improve standard cancer therapy.
  • a "subject” can be a mammal in need of treatment, such as a human or veterinary patient (eg, a rodent such as a mouse or rat, a cat, a dog, a cow, a horse, a sheep, a goat, or other animal).
  • a "subject” can be a clinical patient, a clinical trial volunteer, an experimental animal, and the like.
  • the subject may be suspected of having a disease characterized by cell proliferation or having a disease characterized by cell proliferation, being diagnosed as having a disease characterized by cell proliferation, or being confirmed not to have a cell Control subjects for proliferative diseases, as described herein, diagnostic methods for diseases characterized by cell proliferation and clinical division of such diagnosis are known to those skilled in the art.
  • the composition may be a liquid solution, suspension, emulsion, sustained release formulation or powder, and may be formulated with a pharmaceutically acceptable carrier.
  • the composition can be formulated as a suppository using conventional binders and carriers such as triglycerides.
  • “Pharmaceutically acceptable carrier” refers to a carrier matrix or vehicle that does not interfere with the effectiveness of the biological activity of the active ingredient and which does not confer toxicity to the host or subject.
  • the immune effector cells can be delivered with a pharmaceutically acceptable vehicle.
  • the vehicle can enhance stability and/or delivery properties.
  • Vehicles such as artificial membrane vesicles (including liposomes, nonionic surfactant noisome, nanolipid vesicles, etc.), microparticles or microcapsules, or colloidal formulations comprising pharmaceutically acceptable polymers.
  • a pharmaceutical composition comprising one or more immune effector cells can be formulated as a sterile injectable aqueous or oil according to methods known in the art and using one or more suitable dispersing or wetting agents and/or suspending agents.
  • the sterile injectable preparation may be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent.
  • Example 1 Construction of a vector expressing a chimeric protein
  • the plasmid expressing the chimeric protein comprising the structure shown in Fig. 1 was constructed as follows.
  • Human TGF- ⁇ receptor I signal peptide (SEQ ID NO: 2), extracellular domain of TGF- ⁇ receptor I (SEQ ID NO: 4) and human IL-2RG transmembrane region (SEQ ID NO: 6)
  • the nucleotide sequence of the coding sequence of the IL-2RG intracellular domain (SEQ ID NO: 8) is ligated to construct the nucleotide sequence of the first chimeric protein chIL2RG (SEQ ID NO: 37, which encodes the amino acid sequence as SEQ. ID NO: 27)).
  • the second chimeric protein TGF ⁇ RII-IL21R (SEQ ID NO: 30) was ligated with the coding sequence of the cleavage peptide F2A (SEQ ID NO: 26) and inserted into the pWPT lentiviral expression vector (purchased from Addgene).
  • Plasmid 1 expressing the first chimeric protein and IL-2RB (the plasmid map is shown in Figure 2A)
  • plasmid 2 expressing the first chimeric protein and IL-7RA (the plasmid map is shown in Figure 2B)
  • the expression A chimeric protein and plasmid 3 of IL-21R (plasmid pattern is shown in Figure 2C).
  • the constructed lentiviral plasmid 1, lentiviral plasmid 2, lentiviral plasmid 3 were co-transfected into the HEK-293T cells with the lentiviral packaging plasmid, respectively, and the corresponding lentiviruses were prepared. The following were recorded as lentivirus 1 and lentivirus 2, respectively. Lentivirus 3.
  • Human PBMC were cultured in AIM-V medium, 2% human AB type serum was added, 500 U/mL recombinant human IL-2 was added, and CD3/CD28 antibody was added to activate magnetic beads for 48 h to obtain activated T cells. After T cells were activated, they were infected with lentivirus 1, lentivirus 2, and lentivirus 3, respectively, to obtain chimeric T cells chTR2, chTR7, and chTR21.
  • T cell UTD untransfected T cells, Untransfected
  • UTD, chTR2, chTR7, and chTR21 cells were used to divide into TGF- ⁇ 1 positive group (+) and negative group (-).
  • the culture was carried out, and TGF- ⁇ 1 stimulation was not used in the negative group culture, and the recombinant group was stimulated with recombinant human TGF- ⁇ 1 (5 ng/mL) for 30 min.
  • Cell extracts were collected for Western blot analysis to analyze changes in STAT3/5 phosphorylation levels.
  • Example 3 Effect of chimeric protein-expressing T cells on the level of Treg differentiation stimulated by recombinant human TGF- ⁇ 1
  • Human PBMC were cultured in AIM-V medium, 2% human AB type serum and 500 U/mL recombinant human IL-2 were added to obtain the corresponding cell culture medium.
  • the T cells chTR7, chTR21 and empty control T cell UTD obtained in Example 2 were cultured in the above cell culture medium, cultured for 4 days with recombinant human TGF- ⁇ 1 (5 ng/mL), and cells were collected to label Treg markers with antibodies.
  • CD4, CD25, and Foxp3 were subjected to flow detection, and a test group not treated with recombinant human TGF- ⁇ 1 was used as a control (CTRL).
  • the selected CAR is a GPC-targeting CAR having the scFv set forth in SEQ ID NO: 31, the amino acid sequence of the CAR is set forth in SEQ ID NO: 36, and the nucleotide sequence is SEQ ID NO: 44. Shown.
  • Lentiviral packaging was carried out by molecular biology methods conventional in the art, and a plasmid containing the gene of CAR (SEQ ID NO: 44) was infected with 293T cells to obtain a lentivirus comprising a second generation GPC3-28z-CAR targeting GPC3. .
  • T cell activation Human PBMC were cultured in AIM-V medium, 2% human AB type serum and 500 U/mL recombinant human IL-2 were added, and CD3/CD28 antibody was added to activate magnetic beads for 48 h.
  • T cells were activated, infected with lentivirus 2 and the second generation GPC3-28z-CAR lentivirus targeting GPC3, and T cells expressing chTR7 and CAR were obtained, which were recorded as chTR7-CAR; infected with lentivirus 3 and targeted GPC3 Generation of the LPC of GPC3-28z-CAR, T cells expressing chTR21 and CAR, designated chTR21-CAR, infected with the second generation GPC3-28z-CAR targeting GPC3 (the amino acid sequence of which is shown in SEQ ID NO: 36)
  • the chTR7-CAR, chTR21-CAR and GPC3-CAR-T were divided into control group (CTRL) and rTGF- ⁇ 1 group, CTRL group was cultured under normal conditions, and rTGF- ⁇ 1 group was further added with recombinant human TGF- ⁇ 1 (5 ng/mL). Treatment culture. After 4 days, the target cell was incubated with the target cell Huh7 for 48 h. After the first round of target cell killing, the collected T cells continued to be incubated with the target cell Huh7 for a second round for 48 h. The dead cells were washed with PBS, and the target cell killing was observed under the microscope. The results are shown in Fig. 5.
  • chTR7-CAR and GPC3-CAR-T prepared in Example 4 were selected for in vitro culture under different conditions:
  • ChTR7-CAR and GPC3-CAR-T were cultured under the culture conditions of AMIV medium + 2% human AB serum, without adding IL2 and not co-incubating with tumor cells, the results showed that chTR7-CAR and GPC3-CAR There was no significant difference in proliferative capacity compared to T cells.
  • Huh7-TGF ⁇ is prepared by carrying a lentiviral expression vector plasmid pWPT-EGFP-F2A carrying TGF ⁇ 1 (the nucleotide sequence is shown in SEQ ID NO: 40, the amino acid sequence is shown as SEO ID NO: 41).
  • -hTGFb1 Rev packaging plasmid, RRE packaging plasmid and VSV-G envelope protein pellet were mixed with transfection reagent polyethyleneimine (PEI, Polysciences) and transfected into HEK-293T cells, and transfected to obtain recombinant lentivirus solution.
  • PEI transfection reagent polyethyleneimine
  • the GFP-positive cells were sorted by flow to obtain Huh7-TGF ⁇ , and the flow chart is shown in Fig. 6.
  • Example 4 hepatoma cell lines Huh7 and SK-hep1-GPC3 cells were used as target cells, and in vitro killing of chTR7-CAR and GPC3-CAR-T under different culture conditions was performed at a target ratio of 3:1. The situation was carried out using the CytoTox96 non-radioactive cytotoxicity test kit (Promega). The specific method is described in the CytoTox 96 non-radioactive cytotoxicity test kit.
  • a mouse subcutaneous tumor model was constructed using a liver cancer cell line (PLC/PRF/5-TGF ⁇ 1) overexpressing TGF ⁇ 1.
  • the preparation method of PLC/PRF/5-TGF ⁇ 1 is similar to the preparation method of Huh7-TGF ⁇ of Example 5.
  • the PLC/PRF/5 cell line (purchased from the Shanghai Institute of Biosciences, Chinese Academy of Sciences) was transfected with HEK-293T cells carrying the TGF ⁇ 1 gene to obtain PLC/PRF/5-TGF ⁇ 1 cells.
  • a G-cell-targeting CAR-T cell is employed, and those skilled in the art can employ CAR-T cells targeting other targets, such as targeting EGFR, in accordance with the teachings of the present application.
  • CAR-T cells exemplary, the sequence of the extracellular scFv of EGFR-targeting CAR-T cells is shown in SEQ ID NO: 32), such as CAR-T cells targeting CLD18A2 (exemplary, target)
  • the sequence of the extracellular scFv to the CAR-T cells of CLD18A2 is as shown in SEQ ID NO: 33), such as CAR-T cells targeting CD19 (exemplary, extracellular to CAR-T cells targeting CD19)
  • the sequence of the scFv is as set forth in SEQ ID NO: 34), such as a CAR-T cell targeting BCMA (exemplary, the sequence of the extracellular scFv of the BCMA-targeting CAR-T cell is set forth in SEQ ID NO: 35 ).

Landscapes

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

Abstract

La présente invention concerne une cellule effectrice immunitaire exprimant une protéine chimère et une application associée. La cellule effectrice immunitaire exprime une protéine chimère contenant un domaine extracellulaire d'un récepteur TGF-β et un domaine de signal intracellulaire d'un récepteur d'une protéine de la famille IL-2. L'invention concerne en outre une composition pharmaceutique contenant la cellule effectrice immunitaire et une utilisation de la cellule effectrice immunitaire ou de la composition pharmaceutique pour la préparation d'un médicament pour la prévention ou le traitement de tumeurs ou d'une infection par un micro-organisme pathogène. La cellule effectrice immunitaire exprimant la protéine chimère de la présente invention est capable de transformer la stimulation du TGF-β en un signal positif d'IL-2, d'IL-7 ou d'IL-21, et d'inhiber la différenciation des Treg induite par TGF-β, ce qui favorise la survie et la prolifération de cellules immunitaires, et est également capable de réduire l'effet de TGF-β sur la capacité de destruction de cellules.
PCT/CN2018/119588 2017-12-06 2018-12-06 Protéine chimère, et cellule effectrice immunitaire l'exprimant et application associée WO2019109980A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711279705.1 2017-12-06
CN201711279705 2017-12-06

Publications (1)

Publication Number Publication Date
WO2019109980A1 true WO2019109980A1 (fr) 2019-06-13

Family

ID=66750790

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/119588 WO2019109980A1 (fr) 2017-12-06 2018-12-06 Protéine chimère, et cellule effectrice immunitaire l'exprimant et application associée

Country Status (2)

Country Link
CN (1) CN109880803B (fr)
WO (1) WO2019109980A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020257823A3 (fr) * 2019-06-21 2021-02-18 Kite Pharma, Inc. RÉCEPTEURS DE TGF-β ET LEURS MÉTHODES D'UTILISATION
WO2022214089A1 (fr) 2021-04-08 2022-10-13 克莱格医学有限公司 Utilisation d'immunothérapie cellulaire
WO2023288281A3 (fr) * 2021-07-15 2023-02-23 Fred Hutchinson Cancer Center Polypeptides chimériques
US11708401B2 (en) 2018-06-22 2023-07-25 Kite Pharma, Inc. Chimeric transmembrane proteins and uses thereof
WO2024099990A1 (fr) * 2022-11-07 2024-05-16 Leibniz-Institut Für Immuntherapie (Lit) Cellules car t de récepteur de commutateur tgf-ss

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114929863A (zh) * 2019-08-09 2022-08-19 克莱格医学有限公司 共表达趋化因子受体的免疫效应细胞
CN112390892B (zh) * 2019-08-14 2024-01-12 博源润生医药(杭州)有限公司 一种嵌合抗原受体以及表达该嵌抗原受体的免疫效应细胞
CN110564695A (zh) * 2019-09-23 2019-12-13 华东师范大学 靶向前列腺癌的增强型car-t细胞及其制备方法和药物
JP7488337B2 (ja) * 2019-11-27 2024-05-21 ジーアイ・セル・インコーポレイテッド Il-2タンパク質及びcd80タンパク質を含む融合タンパク質及びnk細胞を含む抗癌治療用組成物
CN111171160B (zh) * 2020-02-14 2022-05-17 四川大学华西医院 基于TGF-β改造的嵌合抗原受体及其修饰的免疫细胞
CN111218426A (zh) * 2020-02-26 2020-06-02 华东师范大学 一种克服肿瘤微环境car-t细胞的制备及其用途
CN111269326A (zh) * 2020-02-28 2020-06-12 南京北恒生物科技有限公司 新型嵌合抗原受体及其用途
CN111690070A (zh) * 2020-05-13 2020-09-22 深圳市众循精准医学研究院 一种sPD-1-Fc-sTGFβRII融合蛋白及其应用
CN115698038A (zh) * 2020-06-01 2023-02-03 上海君赛生物科技有限公司 信号转换受体及其用途
CN112210543B (zh) * 2020-07-30 2023-01-03 山东省成体细胞产业技术研究院有限公司 一种针对实体瘤克服TGF-β免疫抑制的CAR-T细胞
CN116194125A (zh) 2020-08-07 2023-05-30 克莱格医学有限公司 工程化改造的细胞以及工程化改造细胞的方法
CN111978412B (zh) * 2020-08-13 2021-04-30 南京凯地生物科技有限公司 武装靶向TGF-β的特异性嵌合抗原受体细胞及其制备方法和应用
CN116390940A (zh) * 2020-09-23 2023-07-04 海正生物制药有限公司 Tgfbr2-ecd突变体及包含其的融合蛋白与应用
CN117460741A (zh) 2021-06-29 2024-01-26 科济生物医药(上海)有限公司 调控细胞生理活动的嵌合多肽
CN115873802A (zh) * 2021-09-29 2023-03-31 亘喜生物科技(上海)有限公司 嵌合抗原受体免疫细胞及其制法和应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103582699A (zh) * 2011-04-08 2014-02-12 贝勒医学院 使用嵌合细胞因子受体逆转肿瘤微环境的影响
US20160361265A1 (en) * 2012-04-12 2016-12-15 Yale University Nanoparticle-mediated delivery of cytokines for maintenance of the regulatory t cell phenotype

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6578271B2 (ja) * 2013-04-17 2019-09-18 ベイラー カレッジ オブ メディスンBaylor College Of Medicine 免疫抑制TGF−βシグナルコンバーター
MA41538A (fr) * 2014-10-17 2017-12-26 Baylor College Medicine Cellules immunitaires bipartites et tripartites de signalisation
CA2975147A1 (fr) * 2015-01-31 2016-08-04 Yangbing Zhao Compositions et procedes d'administration a des cellules t de molecules therapeutiques

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103582699A (zh) * 2011-04-08 2014-02-12 贝勒医学院 使用嵌合细胞因子受体逆转肿瘤微环境的影响
US20160361265A1 (en) * 2012-04-12 2016-12-15 Yale University Nanoparticle-mediated delivery of cytokines for maintenance of the regulatory t cell phenotype

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CLAUDIA PENAFUERTEL ET AL.: "B Effector Cells Activated by a Chimeric Protein Consisting of IL -2 and the Ectodomain of TGF-b Receptor II Induce Potent Antitumor Immunity", CANCER RESEARCH, vol. 72, no. 5, 12 January 2012 (2012-01-12), pages 1210 - 1220, XP055505176, ISSN: 0008-5472, doi:10.1158/0008-5472.CAN-11-1659 *
STEPHAN GASSER ET AL.: "Constitutive Expression of a Chimeric Receptor That Delivers IL -2/ IL -15 Signals Allows Antigen-Independent Proliferation of CD 8 + CD 44high But Not Other T Cells", THE JOURNAL OF IMMUNOLOGY, vol. 164, 20 March 2000 (2000-03-20), pages 5659 - 5667, XP002783502, ISSN: 1550-6606, DOI: doi:10.4049/jimmunol.164.11.5659 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11708401B2 (en) 2018-06-22 2023-07-25 Kite Pharma, Inc. Chimeric transmembrane proteins and uses thereof
WO2020257823A3 (fr) * 2019-06-21 2021-02-18 Kite Pharma, Inc. RÉCEPTEURS DE TGF-β ET LEURS MÉTHODES D'UTILISATION
CN114391021A (zh) * 2019-06-21 2022-04-22 凯德药业股份有限公司 TGF-β受体和使用方法
AU2020296878B2 (en) * 2019-06-21 2023-04-20 Kite Pharma, Inc. TGF-beta receptors and methods of use
WO2022214089A1 (fr) 2021-04-08 2022-10-13 克莱格医学有限公司 Utilisation d'immunothérapie cellulaire
WO2023288281A3 (fr) * 2021-07-15 2023-02-23 Fred Hutchinson Cancer Center Polypeptides chimériques
WO2024099990A1 (fr) * 2022-11-07 2024-05-16 Leibniz-Institut Für Immuntherapie (Lit) Cellules car t de récepteur de commutateur tgf-ss

Also Published As

Publication number Publication date
CN109880803B (zh) 2024-06-07
CN109880803A (zh) 2019-06-14

Similar Documents

Publication Publication Date Title
WO2019109980A1 (fr) Protéine chimère, et cellule effectrice immunitaire l'exprimant et application associée
Ahmed et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors
US11980640B2 (en) Bicistronic chimeric antigen receptors and their uses
JP6767347B2 (ja) Pd−l1によって誘導される免疫寛容の低減
EP3586852B1 (fr) Expansion de cellules modifiées et leurs utilisations
JP6887492B6 (ja) 遺伝子改変細胞で使用するための共刺激ドメイン
TWI688651B (zh) Car表現載體及car表現t細胞
JP7000315B2 (ja) 高結合活性hpv t細胞受容体
WO2017219936A1 (fr) Lymphocyte t exprimant le récepteur car capable d'exprimer avec efficacité et stabilité un anticorps activé, et ses utilisations
KR20190101979A (ko) 합성 면역 수용체 및 이의 사용 방법
WO2019052562A1 (fr) Protéine de fusion d'une il-4r et son utilisation
WO2017159736A1 (fr) Cellule immunocompétente et vecteur d'expression exprimant des facteurs de régulation de la fonction immunitaire
TWI811278B (zh) 表現特異性辨識人類間皮素之細胞表面分子、il-7、及ccl19之免疫活性細胞
CN113677353A (zh) 修饰细胞的扩增及其用途
Zhang et al. Genetic engineering with T cell receptors
WO2019210863A1 (fr) Cellules effectrices immunitaires et leur utilisation
BR112019019941A2 (pt) receptor de antígeno quimérico
CN114867490A (zh) 细胞因子调控表达的免疫效应细胞
WO2022016114A1 (fr) Molécules chimériques fournissant une co-stimulation ciblée pour une thérapie cellulaire adoptive
WO2022095903A1 (fr) Immunothérapie antitumorale ciblant pd-1h (vista)
CN110819596A (zh) 具有增强的迁移能力的修饰的细胞
WO2021244654A1 (fr) Production de cytokine induite par activation dans des cellules immunitaires
WO2022214089A1 (fr) Utilisation d'immunothérapie cellulaire
WO2022037562A1 (fr) Cellules immunoréactives modifiées et leurs utilisations
CN110540997B (zh) 靶向bcma嵌合抗原受体、核酸序列、载体及应用

Legal Events

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

Ref document number: 18887083

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: 18887083

Country of ref document: EP

Kind code of ref document: A1