[go: up one dir, main page]

WO2024241208A1 - Cd3-targeting switchable car-t cells of which cd3 gene is edited - Google Patents

Cd3-targeting switchable car-t cells of which cd3 gene is edited Download PDF

Info

Publication number
WO2024241208A1
WO2024241208A1 PCT/IB2024/054913 IB2024054913W WO2024241208A1 WO 2024241208 A1 WO2024241208 A1 WO 2024241208A1 IB 2024054913 W IB2024054913 W IB 2024054913W WO 2024241208 A1 WO2024241208 A1 WO 2024241208A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
car
tag
cells
seq
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/IB2024/054913
Other languages
French (fr)
Korean (ko)
Inventor
최경호
김효리
김지환
이아늠
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
SNU R&DB Foundation
Original Assignee
Seoul National University R&DB Foundation
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 Seoul National University R&DB Foundation filed Critical Seoul National University R&DB Foundation
Priority to KR1020257015183A priority Critical patent/KR20250088543A/en
Publication of WO2024241208A1 publication Critical patent/WO2024241208A1/en
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/11T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • 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
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2809Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
    • 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
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • 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
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/99Coculture with; Conditioned medium produced by genetically modified cells

Definitions

  • CD3 gene-edited CD3 targeting switchable CAR-T cells ⁇ CD3 targeting switchable CAR-T cells whose CD3 genes are edited ⁇
  • the present invention relates to a CD3 gene-edited CD3-targeted switchable CAR-T cell (chimeric antigen receptor T cell) and its use in treating T cell-derived tumors.
  • a CD3 gene-edited CD3-targeted switchable CAR-T cell chimeric antigen receptor T cell
  • the CAR-T cell can be effectively used in treating T cell-derived tumors.
  • Chimeric antigen receptor (CAR) gene T-cell therapy is a gene-transferred T-cell therapy that converts T cells into cells that attack the desired target by loading an antibody gene (Chimeric Antigen Receptor) that recognizes cancer antigens and injects it into the patient.
  • CAR-T therapy is a gene-transferred T-cell therapy that converts T cells into cells that attack the desired target by loading an antibody gene (Chimeric Antigen Receptor) that recognizes cancer antigens and injects it into the patient.
  • CAR-T therapy is a gene-transferred T-cell therapy that converts T cells into cells that attack the desired target by loading an antibody gene (Chimeric Antigen Receptor) that recognizes cancer antigens and injects it into the patient.
  • Chimeric Antigen Receptor Chimeric Antigen Receptor
  • T-cell lymphoma is a T cell-derived lymphoma and is known to be a cancer with a very poor prognosis with a 5-year survival rate of approximately 30%, but an appropriate treatment method has not been developed. Therefore, T-cell lymphoma may be an important target for the development of novel CAR-T cell therapies.
  • the ideal target antigen for CARs for T-cell lymphoma is CD3, which is widely expressed in T-cell lymphoma.
  • CD3-targeting CAR genes there is a technical barrier that CAR-T cells cannot be produced because they induce cellular suicide (fratricide) by attacking the CAR-T cells themselves.
  • CD3-targeting CAR-T cells another technical barrier to CD3-targeting CAR-T cells is that when these cells are introduced into a patient's body, they may kill not only T-cell lymphoma but also normal T cells of the patient, causing immunodeficiency side effects in the patient.
  • a conjunctive CD3-targeting CAR-T cell is produced, which performs CD3-targeting function only during tumor killing and does not bind to CD3 after the killing function is finished.
  • Patent Document 0001 Korean Patent Publication No. 2018-0031727 (Publication Date: 2018.03.28)
  • the present invention provides an adapter molecule that regulates the activity of a chimeric antigen receptor T cell (CAR-T cell), comprising (a) a first moiety that binds to CD3 on a target cell; and (b) a second moiety that specifically binds to an extracellular domain of a chimeric antigen receptor.
  • CAR-T cell chimeric antigen receptor T cell
  • the present invention also provides a use of the adapter molecule for inducing activation of a chimeric antigen receptor T cell.
  • the present invention also provides a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to the second moiety of the adapter molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain.
  • the present invention seeks to provide a complex comprising a chimeric antigen receptor T cell with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell.
  • the present invention seeks to provide a kit for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell.
  • the present invention seeks to provide a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell.
  • the present invention seeks to provide a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell with reduced or eliminated expression of CD3 and co-administered with an adapter molecule regulating the activity of the CAR-T cell.
  • the present invention provides a method for preventing or treating a T cell-derived tumor, comprising a step of administering the complex or pharmaceutical composition to a subject.
  • the present invention provides a method for producing a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or eliminated, comprising (a) a step of down-regulating the expression of CD3 or a T cell receptor (TCR) in a T cell, and (b) a step of introducing a chimeric antigen receptor (CAR) comprising an extracellular domain, a transmembrane domain, and an intracellular signaling domain, wherein the steps (a) and (b) are performed regardless of the order.
  • CAR-T cell a chimeric antigen receptor T cell
  • TCR T cell receptor
  • One aspect of the present invention provides an adapter molecule that regulates the activity of a chimeric antigen receptor T cell (CAR-T cell), comprising (a) a first moiety that binds to CD3 on a target cell; and (b) a second moiety that specifically binds to an extracellular domain of a chimeric antigen receptor.
  • CAR-T cell chimeric antigen receptor T cell
  • Another aspect of the present invention provides a use of the adapter molecule to induce activation of a chimeric antigen receptor T cell.
  • the term "adaptor molecule” means a molecule that binds to the extracellular domain of a CAR and, at the same time, binds to a target cell, for example, a target tumor cell, in a T cell therapy using the chimeric antigen receptor, i.e., a cell therapy agent called CAR-T.
  • the adapter molecule Since the adapter molecule has a specific binding ability to target T cell-mediated tumor cells or cancer, the tumor target attacked by the CAR-T cell can be changed by replacing the adapter molecule, and when side effects occur due to excessive activation of the CAR-T cell, the toxicity of the CAR cell can be alleviated by stopping the administration of the adapter molecule or reducing the administration dose (Cao et al., Angew Chem Int Ed Engl. 2016 June 20; 55(26) : 7520-7524.).
  • the term “antibody” refers to an antibody that specifically binds to a specific antigen, and includes not only a complete antibody form but also an antigen binding fragment of the antibody molecule.
  • a complete antibody has a structure having two full-length light chains and two full-length heavy chains, and each light chain is connected to a heavy chain by a disulfide bond.
  • the heavy chain constant region has gamma (X), mu (U), alpha (U), delta (6), and epsilon ( ⁇ ) types and has subclasses gamma 1 (X1), gamma 2 (x2), gamma 3 (x3), gamma 4 (x4), alpha l (al), and alpha 2 (a2).
  • the light chain constant region has kappa (K) and lambda ( ⁇ ) types.
  • the term “antigen binding fragment” means a fragment having antigen binding function, and includes Fab, F(ab'), and Fv, scFv, or sdAB (si ng 1 e-doma in antibody).
  • Fab fragment antigen binding
  • Fab fragment antigen binding
  • Fab' differs from Fab in that it has a hinge region containing one or more cysteine residues at the C-terminus of the heavy chain CH1 domain.
  • Fv is the minimum antibody fragment having only heavy chain variable region and light chain variable region, and recombinant technology for producing Fv fragments is disclosed in PCT International Patent Publication Nos. W0 88/10649, W0 88/106630, W0 88/07085, W0 88/07086, and W0 88/09344.
  • the term “heavy chain” refers to a full-length heavy chain and fragments thereof comprising an antibody variable domain VH and three constant domains CHI, CH2 and CH3, which comprise an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen.
  • the term “light chain” as used herein refers to a full-length light chain and fragments thereof comprising an antibody variable domain VL and constant domain CL, which comprise an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen.
  • CDR complementarity determining region
  • the heavy chain (CDRH1, CDRH2 and CDRH3) and the light chain (CDRL1, CDRL2 and CDRL3) each contain three CDRs.
  • the first moiety may mean a protein having a specific binding ability to CD3. More specifically, the first moiety includes a protein that specifically binds to CD3 expressed on the surface of a target cell, for example, a T cell-derived tumor cell.
  • the protein having a specific binding ability to CD3 may be any one selected from the group consisting of an antibody, an antigen-binding fragment, an affibody, a diabody and an aptamer.
  • the protein that specifically binds to CD3 is a protein that binds to a CD3 molecule on the surface of a T cell-mediated tumor cell, and the protein may be an antibody, specifically, any one of a monoclonal antibody, a polyclonal antibody, or a recombinant antibody.
  • the antibody may be a full-length antibody or an antibody fragment.
  • the antibody fragment may include a part of an anti-CD3 antibody having the ability to bind to CD3.
  • the antibody fragment may be Fab, Fab', F(ab')2, Fv, scFv, or a single-domain antibody (sdAB).
  • the first moiety may be a known antibody or antigen fragment binding having the ability to specifically bind to CD3.
  • CDR complementarity determining region
  • CDR1 CDR2, or CDR3
  • a heavy chain variable region selected from the known anti-CD3 antibodies 0KT3, UCHT1, Tep 1 itechnischab, Otel ixizumab, Vi si 1 itechnischab, and Foralumab.
  • the known anti-CD3 is only an example and is not limited thereto, and the antibody or binding fragment thereof may include a combination of CDR1, CDR2, or CDR3 of each of the known anti-CD3 antibodies.
  • the first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 23 to 25, respectively, or a heavy chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 26 to 28, respectively. Additionally, it may include a light chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and/or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 30. In one embodiment of the present invention, a linker may be included between the light chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and the heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 30.
  • the linker may have an amino acid sequence of SEQ ID NO: 1 ((G4Sh linker), but the type of the linker is not limited thereto.
  • the first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 32 to 34, respectively, or CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 35 to 37, respectively.
  • it may include a light chain variable region comprising an amino acid sequence of SEQ ID NO: 38 and/or a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 39.
  • the first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 67 to 69, respectively, or a heavy chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 70 to 72, respectively. In addition, it ... A light chain variable comprising the amino acid sequence of 73 A heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 75 may be included. In one embodiment of the present invention, a linker may be included between the light chain variable region comprising the amino acid sequence of SEQ ID NO: 73 and the heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 75.
  • the linker may have the amino acid sequence of SEQ ID NO: 74, but the type of the linker is not limited thereto.
  • the first moiety may be a scFv fragment having the amino acid sequences of SEQ ID NOs: 13, 22, 31, and 40, or a Fab fragment comprising the amino acid sequences of SEQ ID NOs: 41 and 42.
  • the anti-CD3 antibody or binding fragment thereof can comprise an amino acid sequence of SEQ ID NOS: 13, 22, 31, 40, 41, and 42, a portion thereof, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% sequence identity thereto.
  • the second moiety can be a peptide tag or a hapten tag.
  • the tag can be selected from the group consisting of a His tag, a Myc tag, a cotinine tag, a FITC tag, a Biotin tag, a Leucin zipper tag, a Flag tag, an Xpress tag, an Avi tag, a calmodulin binding peptide (CBP) tag, a polyglutamate tag, an HA tag, a Strep tag, a Softag 1, a Softag 3, and a V5 tag.
  • the first moiety may be an anti-CD3 antibody or an antigen-binding fragment and/or the second moiety may be a His tag.
  • the adapter molecule regulating the activity of a chimeric antigen receptor T cell comprising the first and second moieties may be a scFv fragment having amino acid sequences of SEQ ID NOs: 13, 22, 31, and 40, or a Fab fragment comprising amino acid sequences of SEQ ID NOs: 41 and 42, wherein 6XHis (HHHHHH) is further added.
  • the adapter molecule may include a Fab in a form in which SEQ ID NOs: 42 and 44 are linked, a Fab in a form in which SEQ ID NOs: 46 and 48 are linked, a Fab in a form in which SEQ ID NOs: 50 and 52 are linked, or a Fab in a form in which SEQ ID NOs: 54 and 56 are linked.
  • the present invention is not limited thereto.
  • the linker may be a polypeptide consisting of any amino acid of 1 to 400, 1 to 200, or 2 to 200.
  • the peptide linker may include Gly, Asn, and Ser residues, and may also include neutral amino acids such as Thr and Ala. Suitable amino acid sequences for the peptide linker are known in the art. Optimization of the linker may also be performed to achieve appropriate separation between functional moieties or to maintain essential inter-moiety interactions. The copy number " n " can be controlled by considering.
  • Other flexible linkers are known in the art, for example, G and S linkers which add amino acid residues such as T and A to maintain flexibility as well as adding polar amino acid residues to improve solubility.
  • the linker can be a flexible linker comprising G, S, and/or T residues.
  • a more specific example of a linker is (GGGGS) n wherein n is an integer from 1 to 20, or from 1 to 10.
  • the light chain variable region and the heavy chain variable region used a (G4S)3 linker (SEQ ID NO: 1).
  • Another aspect of the present invention provides the use of the adapter molecule to induce activation of a chimeric antigen receptor T cell.
  • CAR T cell therapy after CAR T cells injected into a subject's body are used to selectively kill a tumor, and after the tumor is effectively reduced, the remaining residual CAR T cells show a side effect of killing normal cells. This is called CAR T immunodeficiency toxicity or normal T cell toxicity.
  • the adapter molecule showed the effect of CAR T cell immunodeficiency toxicity or normal T cell toxicity evasion ability by regulating the activation of chimeric antigen receptor T cells.
  • the CAR T cell unlike existing CAR Ts that directly target existing tumors, the CAR T cell itself binds to the adapter molecule and binds to the tumor cell targeted by the adapter molecule, so that even when the CAR T cell was injected in the absence of the adapter molecule, it did not show toxicity to the target cell or tumor. That is, CAR T cells induce activation only in the presence of the adapter molecule, and thus show toxicity to the target cell or tumor. Therefore, the adapter molecule can induce activation or inactivation of CAR T cells. For this reason, the adapter molecule is also called a switchable molecule.
  • the use of the adapter molecule to induce activation of chimeric antigen receptor T cells includes the use of preventing immunodeficiency by alleviating cytotoxicity of residual CAR T cells to normal cells and the resulting side effect of killing normal T cells after CAR T cell therapy.
  • Another aspect of the present invention provides a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to a second moiety of an adaptor molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain.
  • extracellular domain means a domain that protrudes outside a cell and binds to a ligand or the like.
  • intracellular domain as used herein means a domain that is located inside the cell membrane of a cell, i.e., in the cytoplasm, and transmits a signal transmitted by binding between the extracellular domain and the ligand into the cell.
  • the chimeric antigen receptor refers to a protein that fuses a cell membrane or intracellular signaling site of a T-cell activation protein (CD3-zeta chain, CD28, 41BBL, 0x40, I COS, high-affinity receptor for IgE (FCERI) and other T-cell activation proteins) with an antigen-binding site (single chain Fv fragment) of a cancer antigen-specific antibody.
  • the first-generation CAR includes an extracellular domain including an antigen recognition site specifically expressed in cancer cells, a transmembrane domain and an intracellular signaling domain, and may use only CD3 as the signaling domain.
  • the second-generation CAR has a structure that combines a co-stimulatory domain (CD28 or CD137/4-1BB) and CD3 ⁇ to enhance responsiveness to immune cells, and may increase the number of immune cells containing CAR remaining in the body compared to the first-generation CAR.
  • the third-generation CAR may have a structure that combines a co-stimulatory domain with 4-1BB, CD28 or 0X40, etc., using two or more co-stimulatory domains to achieve expansion and persistence of immune cells containing CAR in the body.
  • the fourth-generation CAR may include an additional gene encoding a cytokine, such as IL-12 or IL-15, to enable additional expression of a CAR-based immune protein of the cytokine, and the fifth-generation CAR may additionally include an interleukin receptor chain, for example, IL-2RP, to strengthen immune cells.
  • the chimeric antigen receptor may have the structure of a first-generation CAR to a fifth-generation CAR.
  • the extracellular connecting portion of the chimeric antigen receptor may further include a hinge domain.
  • the hinge domain may be characterized by being composed of an oligopeptide or a polypeptide and including 1 to 100 amino acid residues, specifically, 10 to 70 amino acid residues, but is not limited thereto.
  • the intracellular signaling domain of the chimeric antigen receptor may be a portion located inside the cell membrane of an immune cell, i.e., in the cytoplasm, and may refer to a portion that transmits a signal within the cell when the antigen binding domain included in the extracellular domain binds to a target antigen and activates an immune response of the immune cell.
  • the intracellular signaling domain may be, but is not limited to, one or more intracellular signaling domains selected from the group consisting of CD3 zeta ( ⁇ ), CD3 gamma (Y), CD3 delta (6), CD3 epsilon (E), FcR gamma, FcR beta, CD5, CD22, CD79a, CD79b and CD66d, and in one embodiment, the intracellular signaling domain of the chimeric antigen receptor is CD3 zeta ( ⁇ ). In one specific embodiment, the intracellular signaling domain may be characterized by further comprising, but is not limited to, a costimulatory domain.
  • the co-stimulatory domain according to the present invention may include one or more co-stimulatory domains selected from the group consisting of ligands that specifically bind to CD2, CD7, CD27, CD28, CD30, CD40, 4-lBB (CD137), 0X40 (CD134), I COS, LFA-1, GITR, MyD88, DAP1, PD-1, LIGHT, NKG2C, B7-H3 and CD83, but is not limited thereto.
  • the chimeric antigen receptor may be characterized by comprising one or more intracellular signaling domains and one or more co-stimulatory domains.
  • the co-stimulatory domain may be 4-lBB.
  • the reduction or elimination of CD3 expression of the chimeric antigen receptor T cells may be achieved by removing CD3, specifically endogenous CD3, of the T cells or CAR-T cells introduced with the CAR gene before introducing the CAR gene, or by removing the TCR.
  • the reduction or elimination of CD3 expression may be achieved by directly removing the CD3 gene or indirectly reducing, suppressing, or eliminating CD3 expression by removing the TCR.
  • Reducing or eliminating the expression of the CD3 or TCR may be performed by a genome editing technique including a CRISPR system, specifically, CRISPR/Cas9, TALEN, Zinc fi nger nuclease, base-editing and prime-editing, or by a nucleic acid selected from the group consisting of antisense RNA, antagomir RNA, si RNA, shRNA and miRNA.
  • the CAR T cell may be one that binds to the second moiety of the adaptor molecule. More specifically, the CAR T cell may exhibit cytotoxicity against a target cell or a tumor cell by binding to the second moiety of the adaptor molecule.
  • the extracellular domain of the CAR T cell may include an antibody or an antigen-binding fragment thereof having binding ability to the second moiety of the adaptor molecule.
  • the second moiety may be a His tag. Therefore, the CAR T cell, more specifically, the extracellular domain of the CAR T cell may include a protein having a specific binding ability to His tag, specifically, an antibody or an antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof that specifically binds to the His tag may include an amino acid sequence of SEQ ID NO: 63, a part thereof, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identity therewith.
  • homology and identity refer to the degree of relationship between two given base sequences and can be expressed as a percentage.
  • the terms homology and identity are often used interchangeably. Whether any two sequences have homology or identity can be determined by, for example, using the default parameters such as Pearson et al (1988) [Proc. Natl. Acad. Sci. USA 85]: 2444 It can be determined using known computer algorithms such as the "FASTA" program.
  • the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needleman program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al. , 2000, Trends Genet . 16: 276— 277) (version 5.0.0 or later) can be determined using the GCG program package (Devereux, J. , et al. , Nucleic Acids Research 12: 387 (1984)), BLASTP, BLASTN, FASTA (Atschul, [S.] [F. ,] [ET AL, J MOLEC BIOL 215]: 403 (1990): Guide to Huge Computers , Martin J .
  • sequence homology or identity can be determined using BLAST of the National Center for Biotechnology Information Database, or ClustalW.
  • Another aspect of the present invention provides a complex comprising a chimeric antigen receptor T cell in which the expression of CD3 is reduced or deleted and an adapter molecule that regulates the activity of the CAR-T cell.
  • kits for preventing or treating a T cell-derived tumor comprising a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or deleted and an adapter molecule that regulates the activity of the CAR-T cell.
  • CAR-T cell chimeric antigen receptor T cell
  • the "CAR T cell”, the "first moiety”, the “second moiety” or the “adapter molecule” are as described above.
  • Another aspect of the present invention provides a composition comprising a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or eliminated and an adapter molecule that regulates the activity of the CAR-T cell.
  • the composition may be a pharmaceutical composition.
  • the composition may be a pharmaceutical composition for preventing or treating a T cell-derived tumor.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell in which the expression of CD3 is reduced or eliminated and is administered in combination with an adapter molecule that regulates the activity of the CAR-T cell.
  • the "CAR T cell”, the “first moiety”, the “second moiety” or the “adapter molecule” As mentioned above.
  • the term “combination therapy” or “combined treatment” or “in combination” refers to any form of simultaneous or concurrent treatment using at least two separate therapeutic agents.
  • the components of the combination therapy may be administered simultaneously, sequentially, in reverse order, or in any order.
  • the components may be administered in different dosages, at different administration frequencies, or via different routes in an appropriate manner.
  • administration means introducing a given substance into a subject in an appropriate manner
  • subject means any living organism, including humans, such as rats, mice, and livestock, that may have a metabolic disease or adverse effects of a metabolic regulator. As a specific example, it may be a mammal, including humans.
  • administered simultaneously as used herein is not particularly limited, and means that the components of the combination therapy are administered substantially simultaneously, for example, as a mixture or in an immediately subsequent order.
  • administered sequentially is not particularly limited, and means that the components of the combination therapy are not administered simultaneously, but rather between administrations. Means administered one after another or in clusters with a specific time interval.
  • the time interval may be the same or different between the administration of each of the components of the combination therapy and can be selected, for example, in the range of 2 minutes to 96 hours, 1 day to 7 days or 1 week, 2 weeks or 3 weeks.
  • the time interval between administrations can be in the range of several minutes to several hours, for example, in the range of 2 minutes to 72 hours, 30 minutes to 24 hours or 1 to 12 hours. Further examples include time intervals in the range of 24 to 96 hours, 12 to 36 hours, 8 to 24 hours and 6 to 12 hours.
  • disease can mean a pathological condition, particularly cancer, an infectious disease, an inflammatory disease, a degenerative disease, a disease involving apoptosis and graft rejection.
  • treatment refers to or includes the alleviation, inhibition of progression, or prevention of a disease, disorder or condition, or one or more symptoms thereof, and "active ingredient” or “pharmaceutically effective amount” can mean any amount of a composition used in the course of practicing the invention provided herein sufficient to alleviate, inhibit progression, or prevent a disease, disorder or condition, or one or more symptoms thereof.
  • compositions according to one embodiment are used interchangeably and refer to the placement of a composition according to one embodiment into a subject by a method or route that results in at least partial localization of the composition to a desired site according to one embodiment. can mean.
  • the composition according to one embodiment can be administered by any suitable route that delivers the cells or at least a portion of the cellular components to a desired location within a viable organism.
  • the survival period of the cells after administration to the organism can be as short as several hours, for example 24 hours, to several days, or as long as several years.
  • T-cell derived tumors include T-cell prolymphocytic leukemia, T-cell large granular lymphocytic leukemia, chronic lymphoprol i ferat ive disorder of NK cells, aggressive NK leukemia, systemic EBV-positive T-cell 1 lymphoprol i ferat ive disease of chi Idhood, hydroa vaccini forme- 1 ike lymphoma, adult T-cell leukemia/lymphoma, extranodal NK/T-cell 1 lympho (nasal type), Enteropathy-associated T-cel 1 lymphoma, hepatosplenic T-cel 1 lymphoma, subcutaneous panniculitis-like T-cel 1 lymphoma, mycosis fungoides, Se'zary syndrome, primary cutaneous CD30-positive T-cel 1 lymphoproliferative disorders, lymphomatoid papulosis, primary cutaneous anaplastic large cel 1 lymphoma, primary cutaneous gamma/delta T-
  • the administration may be in combination with an additional anticancer agent.
  • additional anticancer agents may include alkylating agents, antimetabolites, spindle inhibitor plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors. Examples of such anticancer agents may include compounds used in targeted therapy and conventional chemotherapy.
  • the antibodies Examples include alemtuzumab, apolizumab, acelizumab, atlizumab, bapineuzumab, bevacizumab, vivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab,
  • the composition according to the present invention may additionally comprise a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier In the case of injections, buffers, preservatives, analgesics, solubilizers, isotonic agents, stabilizers, etc. can be mixed and used, and in the case of local administration, bases, excipients, lubricants, preservatives, etc. can be used.
  • the formulation of the pharmaceutical composition of the present invention can be variously prepared by mixing it with the pharmaceutically acceptable carrier as described above. For example, in the case of injections, it can be prepared in the form of a unit dosage ampoule or a multiple dosage form.
  • the anticancer composition may typically include a surfactant that facilitates movement through a membrane.
  • Such surfactants include those derived from steroids, cationic lipids such as N-[l-(2,3-dioleoyl)propyl-N,N,N-trimethylammonium chloride (DOTMA), or various compounds such as cholesterol hemisuccinate and phosphatidyl glycerol.
  • DOTMA N-[l-(2,3-dioleoyl)propyl-N,N,N-trimethylammonium chloride
  • the composition can be administered in a pharmaceutically effective amount to treat cancer cells or their metastasis, or to inhibit the growth of cancer. It may vary depending on various factors such as cancer type, patient's age, weight, nature and degree of symptoms, type of current treatment, number of treatments, dosage form and route, and can be easily determined by a person skilled in the art.
  • composition according to the present invention can be administered together with the above-mentioned pharmacological or physiological components or administered sequentially, and can also be administered in combination with an additional conventional therapeutic agent, and can be administered sequentially or simultaneously with the conventional therapeutic agent.
  • Such administration may be single or multiple administration. It is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects considering all of the above factors, and this can be easily determined by a person skilled in the art.
  • Another aspect of the present invention provides a method for preventing or treating a T cell-derived tumor, comprising administering to a subject the complex or pharmaceutical composition.
  • Another aspect of the present invention provides a method for producing a chimeric antigen receptor T cell (CAR-T cell) having reduced or eliminated expression of CD3, comprising the steps of (a) down-regulating the expression of CD3 or a T cell receptor (TCR) in a T cell, and (b) introducing a chimeric antigen receptor (CAR) comprising an extracellular domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular domain comprises an antibody or an antigen-binding fragment thereof that specifically binds to a second moiety of an adaptor molecule, wherein steps (a) and (b) are performed in any order.
  • the method for reducing or eliminating the expression of the CD3 or TCR may be performed by a genome editing technique including a CRISPR system, specifically, CRISPR/Cas9, TALEN, zinc finger nuclease, base editing and prime editing, or by a nucleic acid selected from the group consisting of antisense RNA, antagomir RNA, si RNA, shRNA and miRNA.
  • a genome editing technique including a CRISPR system, specifically, CRISPR/Cas9, TALEN, zinc finger nuclease, base editing and prime editing, or by a nucleic acid selected from the group consisting of antisense RNA, antagomir RNA, si RNA, shRNA and miRNA.
  • an adapter molecule comprising an anti-CD3 antigen-binding fragment and a T cell expressing a CD3-chimeric antigen receptor binding thereto exhibit anti-fratricide activity and immunodeficiency toxicity avoidance activity, and thus can be usefully used in a T cell-derived tumor treatment agent.
  • Fig. 1 is a graph showing the results of the first screening of guide genes for human CD3 gene editing.
  • Fig. 2 is a graph showing the results of the second screening of guide genes for human CD3 gene editing.
  • Fig. 3 is a graph showing the analysis of the phenotype of CD3 gene-edited human T cells.
  • Fig. 4 is a graph showing the analysis of the phenotype of CAR T cells introduced with chemical and peptide tags for labeling anti-CD3 adapter antibodies (Cot: cotinine, His: 6XHis).
  • Figure 5 is a graph analyzing the expression rate of 6xHis-CAR in human CAR T cells introduced with His tag.
  • Figure 6 is a schematic diagram showing the structure of anti-CD3 adaptor antibody scFv.
  • Figure 7 is an image showing the result of SDS-PAGE after purification of fusion proteins containing 0KT3-Ck-His scFv and 0KT3-Ck-linker-His.
  • Figure 8 is a schematic diagram showing the structure of anti-CD3 adaptor antibody Fab.
  • Figure 9 is an image showing the result of SDS-PAGE after purification of 1-4-2-Ck-His scFv/l-4-7-Ck-His scFv and 1-4-2-Fab-His/1-4-7-Fab-His fusion proteins.
  • Figure 10 is a graph analyzing the binding ability of 0KT3-Ck-His, 0KT3- Ck- L- His, and UCHT1- Fab- His to the T cell tumor cell line Jurkat.
  • Figure 11 is a graph analyzing the binding ability of 1- 4- 2- Ck- His/1- 4- 7- Ck- His and 1- 4- 2- Fab- Hi s/1- 4- 7- Fab- His to the T cell tumor cell line Jurkat.
  • Figure 12 is a graph analyzing the antitumor activity and tumor killing ability of 0KT3- Ck- His scFv and human anti-6xHis CAR T cells.
  • Figure 13 is a graph analyzing the tumor killing ability of 0KT3-His Fab and human anti-6xHis CAR T cells.
  • Fig. 14 is a graph analyzing the antitumor activity and tumor killing capacity of UCHT1-His Fab and human anti-6xHis CAR T cells.
  • Fig. 15 is a graph analyzing the antitumor activity and tumor killing capacity of 1-4-2-His Fab and 1-4-7-His Fab and human anti-6xHis CAR T cells.
  • Fig. 16 is a graph showing the fratricidal activity of human anti-6xHis CAR T cells.
  • Fig. 17 is a graph analyzing the phenotype of T cells after CD3 gene editing in human anti-6xHis CAR T cells.
  • Fig. 18 is a graph confirming the fratricidal prevention capacity of human conjugated CAR T cells through anti-CD3 adapter antibody.
  • Fig. 14 is a graph analyzing the antitumor activity and tumor killing capacity of UCHT1-His Fab and human anti-6xHis CAR T cells.
  • Fig. 15 is a graph analyzing the anti
  • FIG. 19 is a schematic diagram showing the proliferation conditions of CD3 gene-edited conjugated CAR T cells.
  • Figure 20 is a graph analyzing the phenotype of T cells in the first and second CD3 gene-edited conjugated CAR T cell proliferation stages.
  • Fig. 21 is a graph showing the number of CAR T cells measured at the CD3 gene-edited conjugated CAR T cell proliferation stage.
  • Fig. 22 is a schematic diagram of an anti-tumor activity assay experiment of 0KT3- Ck- Hi s scFv and CD3- human Hi s CAR T cells in a mouse model.
  • FIG. 23 is a graph analyzing the anti-tumor activity and the survival rate of a mouse model of 0KT3- Ck- Hi s scFv and CD3- human Hi s CAR T cells in a mouse model.
  • Fig. 24 is a schematic diagram of an anti-tumor activity assay experiment of UCHT1- Fab- Hi s and CD3 -gene-edited human Hi s CAR T cells in a mouse model.
  • Fig. 25 is a graph analyzing the anti-tumor activity and the survival rate of a mouse model of UCHT1- Fab- Hi s and CD3 -gene-edited human Hi s CAR T cells in a mouse model.
  • Figure 26 is a graph analyzing the immunodeficiency prevention ability of human conjugated CAR T cells.
  • Figure 27 shows the antigen affinity of humanized antibody anti-His.
  • Figure 28 shows the results of confirming CAR expression on the surface of huHis CAR- T cells by flow cytometry.
  • Figure 29 shows the results of verifying the in vitro activity level of conjugated CAR T cells using the produced huHis CAR- T cells and the UCHTl-Fab-His adapter.
  • conjugated CAR-T cells were created to avoid fratricide between CAR-T cells by making the D CAR-T cells themselves not express CD3, and (2) minimize the immunodeficiency side effect in patients caused by CAR-T cells from which CD3 has been removed killing normal T cells that express CD3.
  • a schematic diagram of this concept is as follows.
  • mice were purchased from Orient Bio. NSG mice (NOD. Cg- Prkdcscid 112rgtmlWj 1/SzJ) were purchased from Jackson Laboratory. All mice were housed in a specific pathogen-free (SPF) animal facility at Seoul National University College of Medicine (Seoul, Republic of Korea) and maintained according to the guidelines of the Institutional Animal Care and Use Committee (IACUC).
  • SPF pathogen-free
  • Jurkat human T-cell lymphoma
  • ATCC Manassas, VA USA
  • Phoenix GP was provided by Garry Nolan (Stanford University, USA).
  • GFP-luciferase expressing cell line Jurkat-Luc
  • pLEF-Luc-GFP lentivirus for GFP-Luc expression
  • GFP-positive cells were isolated using a cell sorter (FACS Aria, Becton Dickinson), and then a single cell line was established through limiting dilution.
  • the deficient Jurkat cell line was created by introducing an in-del mutation into the CD3 gene expressed by the cell line using the CRISPR-Cas9 system, then isolating CD3-negative cells using a cell sorter, and then establishing a single cell line through the limiting dilution method.
  • Cot CAR scFv was derived from rabbit-derived anti-cot inine antibody (Cl in Chim Acta 2010; 411:1238-1242), Hi s CAR scFv was derived from mouse-derived anti-Hi s antibody (mutated 3D5 clone; J . Mol . Biol . (2002) 318 , 135-147), and Myc CAR scFv was derived from mouse-derived anti-Myc antibody (9E10 clone; FEES Letters 414 (1997) 33-38).
  • the mouse CAR protein coding region consists of the signal peptide of the mouse immunoglobulin kappa chain, the antibody scFv, and the previously reported CAR backbone region (mouse CD28 extracellular region-transmitter region-intracellular region, mouse CD3 zeta intracellular region; GenBank HM754222.1, Blood 2010: 116(20):4099-4102).
  • CAR cDNAs were produced by requesting DNA synthesis to Integrated DNA technologies (IDT) of Korea, USA.
  • the retroviral vector for producing mouse CAR-T cells for CAR expression was produced by cloning the PuroR gene region downstream of the PGK promoter in the pMSCV-puro (Ckmtech, USA) retroviral vector and replacing it with the produced CAR cDNA.
  • the human CAR protein coding region consists of the signal peptide of human GMCSF, the scFv of anti-His antibody, the extracellular region-transmitter region-intracellular region of human CD28, and the intracellular region of human CD3 zeta.
  • the protein coding region cDNA was produced by requesting DNA synthesis to IDT, USA.
  • the retroviral vector for human CAR- T cell expression was cloning using human CAR cDNA using pMSGV (Addgene Plasmid #64269).
  • the lentiviral vector for human CAR- T cell expression was produced by cloning human CAR cDNA into a partially modified pCDH-EF1 vector (Addgene Plasmid #72266).
  • VSV-G vesicular stomatitis Indiana virus G protein
  • the PG13 cell line for virus production was used.
  • Each retroviral plasmid was transformed into Phoenix Eco cells using Lipofectamine 3000 (Invitrogen). After 48 hours, the culture supernatant containing retrovirus was harvested and added to PG13 cells for transduction. Thereafter, the establishment of retrovirus production cell lines and virus production were performed in the same manner as for retrovirus for mouse cells.
  • each lentivirus plasmid was transfected into 293T cell 1 1 ine (ATCC) together with three types of packaging DNA (pMD.2G, pMDLg/pRRE, pRSV-rev) using Lipofectamine 3000 (Invitrogen). After 24 and 48 hours, the culture supernatant containing secreted lentivirus was harvested and filtered (0.45 ⁇ 1 filter) to remove cell residual particles. After 100-fold concentration using an ultracentrifuge, it was used as a lentivirus concentrate for producing CAR T cells.
  • the CRISPR/Cas9 system was used as a method for CD3 gene editing.
  • the cDNA of the human CD3 gene targeting guide RNA (sgRNAl ⁇ 18) was cloned into the vector (PX458) containing the Cas9 cDNA.
  • the CD3 gene editing was performed by transfecting Jurkat cells, which are CD3-positive cells, with the constructed plasmid through electroporation.
  • CD3 gene editing in human primary T cells white blood cells obtained from normal individuals through leukapheresis were added together with anti-CD3 antibody (0KT3, 10//g/mt, BioXcell) to a 24-well plate coated with anti-CD28 antibody (CD28.2, 2//g/mt, BD Biosciences), and then cultured for 48 hours to activate T cells. Afterwards, a complex of Cas9 protein (Cas9 protein, Thermo) and target guide RNA Ribonucleoprotein (RNP) was produced and gene-edited into T cells using electroporation.
  • Cas9 protein Cas9 protein, Thermo
  • RNP target guide RNA Ribonucleoprotein
  • the gene-edited T cells were cultured in a culture medium without antibiotics but with hIL-2 (200 U/ml) for 24 hours, and then transferred to a culture medium with antibiotics and hIL-2 to culture the T cells.
  • the degree of CD3 protein removal through CD3 gene editing was confirmed 36 hours after gene editing by flow cytometry (FACS-Canto II, BD Biosciences) to determine the degree of CD3 protein expression or T cell receptor expression.
  • sgRNA guide RNA + scaffold RNA
  • Cas9 mRNA were newly produced through an in vitro transcription reaction, and then an experiment was conducted to verify whether CD3 was removed in Jurkat using mRNA electroporation, and the results are shown in Fig. 2.
  • sequences of sgRNA7 and sgRNA15 are shown in the following Table and Table 2, respectively.
  • conjugated CAR-T cells For the production of conjugated CAR-T cells, cotinine, a nicotine metabolite, and peptide epitopes 6XHis tag and myc tag were selected as candidates for the epitope tags to be labeled on the anti-CD3 adapter antibody, and anti-cotinine CAR-T cells (Cot CAR-T cells), anti-6XHis tag CAR-T cells (His CAR-T cells), and anti-myc tag CAR-T cells (Myc CAR-T cells), respectively, were produced.
  • Anti-cotinine CAR-T cells were produced by transducing a retrovirus containing a CAR gene including an anti-cotinine antibody scFv (Korean Patent Publication No. 2018-0031727) into activated mouse T cells.
  • retroviruses carrying a CAR gene including the scFv of a previously reported anti-myc tag antibody c 1 one 9E10, FEES Letters 414 (1997) 33-38
  • a CAR gene including the scFv of an anti-6xHis tag antibody mutated clone 3D5, J. Mol. Biol.
  • mice spleen and lymph node cells obtained from normal mice were added together with a-CD28 antibody (37.51, 2//g/mt, BD Biosciences) to a 24-well plate coated with a-CD3 antibody (145-2C11, 10w/mt, BioXcell), and T cells were activated.
  • a-CD28 antibody 37.51, 2//g/mt, BD Biosciences
  • a-CD3 antibody 145-2C11, 10w/mt, BioXcell
  • T cells were transduced with concentrated retrovirus through spin infection in the presence of 6 ug/ml polybrene (polybrene, Sigma Aldrich) at 24 °C, 2500 rpm, and 90 min. This procedure was repeated once more on the same day.
  • T cells were additionally cultured in the presence of mouse IL-2 (30 U/mt, Gibco) for 48 h. The retrovirus-transduced T cells were washed twice, and then proliferated for 2-3 days in fresh culture medium containing mouse IL-2 (20 U/mt) to be used as CAR T cells.
  • CAR protein on the cell surface was performed using a-rabbit IgG (Cot CAR), Q-mouse IgG (His CAR, Myc CAR), His-biotin (biotin-labeled 6xHis peptide) and fluorescently labeled streptavidin were stained and measured using flow cytometry.
  • Mouse CD3 gene-edited CAR- T cells were subjected to mouse CD3 gene editing using electroporation the day after retroviral transduction. The expression of CAR protein on the surface of each of these cells was confirmed, and this is shown in Fig. 4a.
  • conjugated CAR-T cells To produce conjugated human CAR-T cells, the anti-His CAR gene used in the mouse CAR-T cell experiment was redesigned as a human CAR backbone, and then cloned into a retroviral vector for human CAR T cells to produce them.
  • the produced retrovirus was genetically introduced into human T cells isolated from peripheral blood to produce His CAR-T cells. Specifically, leukocytes isolated from normal blood were added to a 24-well plate coated with anti-CD3 antibody together with anti-CD28 antibody, and then cultured for 48 hours to activate the T cells. The activated T cells were washed twice and then used for retroviral transduction.
  • Retronectin (20w/mt, TaKaRa) overnight at 4°C
  • 2% BSA-DPBS was added to the washed 24-well plate, blocked at 37°C for 30 minutes, and washed.
  • Retrovirus concentrate Imt was added, and centrifuged at 2000xg and 32°C for 2 hours to attach the retrovirus to the bottom of the well.
  • activated T cells 1X106 cells/mt
  • Imt were added to the well, and centrifuged for 10 minutes (lOOOxg, The cells were attached to the retrovirus via the 32°C medium. Subsequently, they were cultured for 48 hours in the presence of human IL-2 (200 U/mt, Pro leukin, Novartis).
  • FIG. 5 is a graph analyzing the expression rate of 6xHis-CAR in human CAR T cells introduced with the His tag.
  • the amino acid sequence by domain of His CAR-T cells is shown in Table 3 below.
  • the 6xHis peptide has been widely used as an epitope tag for protein purification, and therefore a system for mass purification of antibody drugs using it is well-established, so there is an advantage of being able to achieve two purposes at the same time, that is, the role of the adapter epitope tag for the conjugated His CAR T cell and the role of the epitope tag for adapter purification, using one epitope tag.
  • the 6xHis tag was used in the anti-CD3 adapter antibody.
  • 6xHis-tagged adapter antibodies were produced in scFv form using the commercially available anti-human CD3 antibody 0KT3 (Muromonab) sequence or UCHT1 sequence.
  • a construct was produced by connecting the VL region and the VH region of OKT antibody or UCHT1 antibody with a linker ((GGGGS)J, and then connecting the human immunoglobulin kappa constant region (CK) for separation and stability, and directly connecting a 6xHis tag to the C-terminus or connecting a short linker peptide and a 6xHis tag.
  • a schematic diagram of the construct is shown in Fig. 6, and the amino acid sequences of the scFv of 0KT3 are shown in Tables 4 and 5, respectively.
  • Table 4 shows the amino acid sequence of the scFv (0KT3-Ck-His) that does not include a linker after the Ck domain
  • Table 5 shows the amino acid sequence of the scFv (0KT3-Ck-L-His) that includes a linker after the Ck domain.
  • the amino acid sequence of the scFv (UCHTl-Ck-His) of UCHT1 is shown in Table 6 below.
  • a -His adapter antibody was also produced in a Fab form that possesses two 6xHis tags so that it can bind to 6xHis CAR in a bivalent form.
  • a schematic diagram of this is shown in Fig. 8, and the amino acid sequences of the Fabs of 0KT3 and UCHT1 (0KT3-Fab-His, UCHT1-Fab-His), respectively, are shown in Tables 7 and 8 below.
  • anti-human CD3 antibodies were additionally screened for CD3 binding antibodies using an animal immune antibody library. Specifically, human CD3 gamma/epsilon and delta/epsilon extracellular heterodimer proteins were mixed with an adjuvant and immunized in 10 chickens four times in total at two-week intervals.
  • the selected antibodies they were cloned into an animal cell expression vector to produce them in the form of scFv-Ck-6xHis fusion proteins, and then transformed into HEK293F cells. After culturing with shaking for 5 days, the culture medium was harvested and affinity purified using KappaSelect beads, and the expression of the antibodies was confirmed through SDS-PAGE.
  • scFv and Fab types of adapter antibodies were shown, and the amino acid sequences thereof are shown in Tables 9 to 12 below, and in particular, the results of SDS-PAGE analysis of scFv (1-4-2-Ck-His, 1-4-7-Ck-His) of 1-4-2 and 1-4-7 are shown in Figure 9.
  • anti-human CD3 (UCHT1, 1-4-2, 1-4-7) Fab-His antibody and anti-human CD3 (0KT3, UCHT1, 1-4-2, 1-4-7) scFv-His antibody were cloned into the antibody expression vector pCEP4, transduced into 293F cells, and expressed in the culture medium.
  • the expressed antibodies were separated and purified through affinity purification to separate the antibodies present in the culture medium.
  • cell surface binding was verified using an electrophoresis method using SDS-polyacrylamide gel and flow cytometry on a cell line expressing human CD3.
  • CD3-positive cells The binding of CD3-positive cells to the adapter antibody was verified by incubating 1X10 5 cells/lug adapter at 4 °C for 1 hour. After washing to remove free adapter antibodies, the cells were stained with anti-His antibody (J096G46, Biolegend) that captures the adapter antibodies, and then analyzed using flow cytometry.
  • Antitumor activity of anti-CD3 adapter antibody and human conjugated CAR-T cells To verify in vitro whether the complex of the 6xHis-tagged anti-CD3 adapter antibody and human anti-6xHis CAR-T cell (His CAR-T cell) produced in the above Example 2 recognizes and activates CD3-positive T cell tumors, Jurkat leukemia cells were co-cultured with 0KT3 scFv-Ck-6xHis (0KT3-Ck-His), 0KT3 scFv- Ck- 6xHis (0KT3- Ck- L- His), and mouse His CAR-T cells, and the amount of IFN- r secreted by the CAR-T cells was measured by ELISA.
  • Fab-His adapter antibodies were produced using UCHT1 antibody, which is another clone of anti-CD3 antibody, and two antibodies (1-4-2 and 1-4-7) selected through screening, and the in vitro activity and cell killing ability of conjugated CAR T cells were analyzed using the same method. The results are shown in Fig. 14 (UCHT1) and Fig.
  • the conjugated CAR T cells were activated in a concentration-dependent manner only in the presence of anti-CD3 adapter antibody, produced a large amount of IFN- r, and exhibited tumor killing ability.
  • the cancer antigen recognition ability, specific activation, and cell killing ability of the anti-CD3 adapter antibody and CAR T cell complex were confirmed.
  • conjugated CAR-T cells were activated by recognizing tumor cells via the adaptor antibody, tumor cells were pre-incubated with the adaptor antibody for 1 hour, and then unbound free adaptor antibody was removed by washing. After that, the tumor cells with the adaptor attached were used in in vitro experiments together with the conjugated CAR-T cells.
  • Luminometer Luminometer
  • CAR T cells and target cells were mixed at a ratio of 1:5 (CAR- T: 3X104 cells, Jurkat: 1.5X105 cells) and co-cultured in a 96-well plate for 24 hours, and then the culture supernatant was harvested.
  • the amount of IFN- x secreted into the supernatant was measured by ELISA (mouse IFNx, human IFNy ELISA kit, BD Biosciences).
  • the viability analysis was performed using a method that analyzes the ratio of 7-AAD-negative cells (live cells) by utilizing 7-AAD, a fluorescent substance that binds to the surface of dead cells, and the results are shown in Fig. 16. As shown in Fig. 16, it was confirmed that fratricide was induced by confirming that the survival rate of anti-6xHis- CAR T cells was significantly reduced in the presence of anti-CD3 adapter antibodies. Therefore, it was experimentally confirmed that conjugated human CAR T cells require the removal of CD3 protein by CD3 gene editing through anti-CD3 adapter antibodies. Experimental Example 3.
  • DHis CAR T cells, 2 His CAR T cells + anti-CD3 adapter antibody (0KT3-His), ⁇ CD3 deleted His CAR T + anti-CD3 adapter antibody (0 ⁇ 3- ⁇ 8) cells was analyzed using flow cytometry. The results are shown in Fig. 18. As shown in Fig. 17, it was confirmed that human CD3 gene-edited conjugated CAR T cells were successfully produced. As shown in Fig. 18, in the presence of anti-CD3 adapter antibodies, the survival rate of CD3 gene-edited CAR T cells (73.1%) was significantly higher than that of CAR T cells in which CD3 was not deleted (43.3%) (73.1% vs. 43.3% under 24-hour culture conditions).
  • the human CD3 gene-edited conjugated CAR T cells produced through CRISPR/Cas9 transfection and CAR transduction in the above Experimental Example 3 include some CD3-positive cells.
  • a process of purifying CD3-negative cells was required, and negative selection was performed using MACS (magnetic activation-associated cell sorting) using anti-human CD3 magnetic beads.
  • MACS magnetic activation-associated cell sorting
  • CD3 protein gene-editing was performed using electroporation, and then retroviral or lentiviral transduction methods were used to produce CD3 gene-edited conjugated CAR-T cells.
  • CD3-positive cells remaining after gene editing only CD3-negative cells were sorted using magnetic-associated cell sorting (MACS).
  • MCS magnetic-associated cell sorting
  • T cells Five days after CD3 was deleted using CRISPR/Cas9 gene editing technology, CD3 gene-edited T cells were cultured with anti-human CD3 antibodies attached to magnetic microspheres. Afterwards, cells cultured with magnetic microsphere-antibody (hCD3 microbead, Mil tenyi) were passed through a strong magnetic separator, and CD3-positive cells that were not gene-edited were attached to the magnetic microsphere-antibody and attached to the magnetic separator. On the other hand, CD3-negative cells passed through the separator without attaching to the separator and were separated.
  • hCD3 microbead Mil tenyi
  • CD3-negative cells were further cultured in a culture medium containing hIL-2 (200 U/ml).
  • the expression of CAR protein on the cell surface was determined by flow cytometry (FACS-Canto II, BD Biosciences) after staining the expanded CAR T cells with Hi s-biotin and APC-labeled SA (SA-APC, BioLegend) 5 days after retroviral transduction.
  • SA-APC Hi s-biotin and APC-labeled SA
  • CD3-negative human CAR T cells could be effectively separated and purified by subjecting human CD3 gene-edited conjugated CAR T cells to the Ant i-CD3 MACS process, and we also confirmed whether the CD3-negative state was maintained in the T cell population after the secondary cell proliferation period. The results are shown in Figs. 20 and 21.
  • CD3 gene-edited conjugated CAR T cells day 1: approximately 2xlO 7 T cells
  • day 0: IxlO 6 T cells day 0: IxlO 6 T cells
  • CD3 gene-edited human CAR T cell 3xl0 6 cells produced from human T cells were intravenously injected.
  • the first anti-CD3 adapter antibody (0KT3(scFv)- Ck-His) was administered 4 hours before CAR T cell injection at a dose of 20 ug per mouse, and then administered at 2-day intervals for a total of 8 times (Fig. 22).
  • Bioluminescence images were acquired once a week using IVIS 100 (PerkinElmer, Waltham, MA USA) to measure the size of tumor cells. Each mouse was injected intraperitoneally with 2 mg of D-Luci fer in 100 ⁇ l of saline.
  • the size of the tumor cells was measured via IVIS 10 minutes after the intraperitoneal injection.
  • the NSG mice were divided into three groups: 1 a group injected with only the D Jurkat cell line, 2 a group injected with only CD3 gene-edited conjugated CAR T cells without anti-CD3 adapter antibody, and 3 a group injected with anti-CD3 adapter antibody + CD3 gene-edited conjugated CAR T cells, and the experiment was conducted.
  • the survival rate of the mouse model up to 60 days after tumor cell injection was analyzed. The results are shown in Fig. 23. As shown in Fig. 23, compared to when CD3 gene-edited conjugated CAR T cells were administered alone, when anti-CD3 adapter antibody was administered together, a significant decrease in the tumor was observed, and it was confirmed that the survival rate of the mice also increased significantly.
  • a CD3 gene-edited human CAR T cell line produced using human T cells were intravenously injected.
  • the UCHTl-Fab-His adapter antibody was administered as the first anti-CD3 adapter antibody 4 hours before CAR T cell injection at 20 ug per mouse, and was then repeatedly administered 7 times at 2-day intervals. After a 2-week rest period, the anti-CD3 adapter antibody was administered 8 times at 2-day intervals again, for a total of 16 times (Fig. 24).
  • the NSG mice were divided into 1 a group injected with only the Jurkat cell line, 2 a group injected with only CD3 gene-edited conjugated CAR T cells without anti-CD3 adapter antibody, 3 a group injected with only anti-CD3 adapter antibody without CD3 gene-edited conjugated CAR T cells, and ⁇ a group injected with anti-CD3 adapter antibody + CD3 gene-edited conjugated CAR T cells, and the experiment was conducted.
  • the survival rate of the mouse models up to 80 days after tumor cell injection was analyzed. The results are shown in Fig. 25. As shown in Fig.
  • T cells of human peripheral blood mononuclear cells were used as target cells, and after adding anti-human CD3 antibodies and/or CD3 gene-edited human conjugated CAR T cells, the killing of normal T cells was analyzed through flow cytometry, and the results are shown in Fig. 26.
  • PBMCs peripheral blood mononuclear cells
  • FIG. 26 shows that only in the presence of anti-human CD3 adapter antibody, T cells expressing CD3 are rapidly reduced, thereby confirming the killing of normal T cells by CAR T cells, and in the absence of adapter antibody, the number of T cells positively expressing CD3 is maintained regardless of the presence or absence of CD3-conjugated CAR T cells, indicating that normal T cells are not killed.
  • humanization and affinity maturation of the mouse-derived anti-6xHis scFv (mutated 3D5) used in the His CAR were performed.
  • Antibody humanization and affinity maturation For humanization and affinity maturation of the mouse anti-His scFv antibody, first, six CDRs (complementarity determining regions) showing the most appropriate sequence similarity were grafted onto the IGHV1-18 and IGKV2-28 human germline genes. At the same time, several amino acid sites in the framework region (FR) were changed to remove post-translational modifications and perform reverse mutations.
  • CDRs complementarity determining regions
  • the binding kinetics and affinity constants were generated using a Biacore 8K SPR system (Cytiva) equipped with a CM5 sensor chip.
  • Biacore 8K SPR system Cytiva
  • streptavidin was immobilized and then biotin-conjugated 6xHis peptide was captured.
  • the antibody was serially diluted 2-fold with 1x PBS and injected at a flow rate of 30 ⁇ L/min for 3 min followed by a 10-min dissociation step.
  • the surface was regenerated using 10 mM glycine, pH 1.5.
  • the kinetics were analyzed using Biacore 8K Evaluation Software Version 1.0 (Cytiva).
  • Figure 27 shows the antigen affinity of the humanized antibody anti-His, and the final humanized antibody scFv showed an antigen affinity approximately 5.8-fold higher than that of the original scFv, demonstrating successful humanization.
  • a CAR gene similar to the existing His CAR was cloned into a lentiviral vector to produce a lentivirus, which was then transduced into human T cells to produce humanized His CAR- T cells (huHis CAR- T cells).
  • the expression of the CAR on the surface of the huHis CAR- T cells was confirmed by flow cytometry, and the results are shown in Fig. 28.

Landscapes

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

Abstract

The present invention relates to: CD3-targeting switchable CAR-T cells (chimeric antigen receptor T cells) of which a CD3 gene is edited; and use thereof for treating T cell-derived tumors. By using CAR-T cells with reduced or eliminated expression of CD3 and an adapter molecule that regulates the activity of the CAR-T cells in combination, the CAR-T cells exhibit anti-fratricide ability and the ability to avoid immunodeficiency toxicity, and thus can be effectively used in a therapeutic agent for T cell-derived tumors.

Description

【명세서】 【Specification】

【발명의 명칭】 【Title of invention】

CD3 유전자 편집된 CD3 표적 스위처블 CAR-T세포 {CD3 targeting switchable CAR-T cells whose CD3 genes are edited} CD3 gene-edited CD3 targeting switchable CAR-T cells {CD3 targeting switchable CAR-T cells whose CD3 genes are edited}

【기술분야】 본 발명은 CD3 유전자 편집된 CD3 표적 스위처블 CAR-T 세포 (키메라 항원 수용체 T세포) 및 이의 T세포 유래 종양 치료용도에 관한 것으로서 , CD3의 발현이 감소되거나 제거된 CAR-T세포 및 상기 CAR-T세포의 활성을조절하는 어댑터 분자를 병용함으로써 , T세포 유래 종양 치료에 효과적으로사용될 수 있다. 【Technical Field】 The present invention relates to a CD3 gene-edited CD3-targeted switchable CAR-T cell (chimeric antigen receptor T cell) and its use in treating T cell-derived tumors. By using a CAR-T cell with reduced or eliminated expression of CD3 and an adapter molecule that regulates the activity of the CAR-T cell in combination, the CAR-T cell can be effectively used in treating T cell-derived tumors.

【발명기술】 키메라 항원 수용체 (Chimeric antigen receptor : CAR) 유전자 T세포 치료제 (CAR-T 치료제)는 T 세포에 암항원을 인지하는 항체 유전자 (Chimeric Antigen Receptor)를 탑재하여 원하는 표적을 공격하는 세포로 전환시켜 환자에게 주입하는 유전자 이입 T세포 치료제로서, 기존의 암을 표적하는 항체나 면역세포 치료제와 비교하여 획기적인 치료 결과가 미국 등 선진국에서 계속적으로 보고되고 있다. 그러나, CAR-T 치료제의 성공사례는 아직 CD19 양성 B세포 유래 혈액종양으로 국한되어 있어, CD19 외에 다양한 다른 종양 항원 표적을 대상으로 하는 CAR유전자 T세포요법의 개발은혈액종양의 완치율을높이는데 획기적 기여를할것으로 예상된다. 말초성 T세포 림프종 (per ipheral T cel 1 lymphoma)은 T세포 유래의 림프종으로 5년 생존률이 30%정도로예후가매우좋지 않은암종으로알려져 있으나, 적절한 치료법의 개발이 이루어지지 않고 있는 상황이다. 따라서 , T세포 림프종은 새로운 CAR-T세포치료법 개발의 중요한 대상이 될 수 있다. 【Invention Technology】 Chimeric antigen receptor (CAR) gene T-cell therapy (CAR-T therapy) is a gene-transferred T-cell therapy that converts T cells into cells that attack the desired target by loading an antibody gene (Chimeric Antigen Receptor) that recognizes cancer antigens and injects it into the patient. Compared to existing cancer-targeting antibodies or immune cell therapies, groundbreaking treatment results have been continuously reported in advanced countries such as the United States. However, successful cases of CAR-T therapy are still limited to CD19-positive B cell-derived hematological tumors, so the development of CAR gene T-cell therapy targeting various tumor antigen targets in addition to CD19 is expected to make a groundbreaking contribution to increasing the cure rate of hematological tumors. Peripheral T cell 1 lymphoma is a T cell-derived lymphoma and is known to be a cancer with a very poor prognosis with a 5-year survival rate of approximately 30%, but an appropriate treatment method has not been developed. Therefore, T-cell lymphoma may be an important target for the development of novel CAR-T cell therapies.

T세포 림프종의 CAR표적 항원으로는 T세포 림프종에서 광범위하게 발현되는 CD3가가장 이상적인 표적이지만, 정상 T세포에서도발현된다는단점을가지고 있다. 따라서, CD3 표적 CAR유전자를 이입한 T세포를 제조하는 경우, CAR-T세포 자신을 공격함으로써 세포의 자살 (fratricide)을유도하므로 CAR-T세포를제조할수없다는 기술적 장벽이 존재한다. 또한, CD3 표적 CAR-T세포의 또 하나의 기술적 장벽은 이 세포가 환자 체내에 이입되었을 때, T세포 림프종을 살상할 뿐 아니라 환자의 정상 T세포도 살상함으로써 환자의 면역결핍 부작용을 유발할수 있다는 점이다. 이에 , 본 발명에서는 접합형 (conjunctive) CD3표적 CAR-T세포를 제작하여 , 종양살상시에만 CD3 표적 기능을수행하고, 살상기능이 끝난후에는 CD3에 결합하지 않도록 함으로써 상기 두 가지 기술적 어려움을 극복하여 말초성 T세포 림프종에 대한 CAR-T 세포 치료제에 관한 발명을 완성하게 되었다. The ideal target antigen for CARs for T-cell lymphoma is CD3, which is widely expressed in T-cell lymphoma. However, it has the disadvantage of being expressed in normal T cells as well. Therefore, when producing T cells with CD3-targeting CAR genes, there is a technical barrier that CAR-T cells cannot be produced because they induce cellular suicide (fratricide) by attacking the CAR-T cells themselves. In addition, another technical barrier to CD3-targeting CAR-T cells is that when these cells are introduced into a patient's body, they may kill not only T-cell lymphoma but also normal T cells of the patient, causing immunodeficiency side effects in the patient. Therefore, in the present invention, a conjunctive CD3-targeting CAR-T cell is produced, which performs CD3-targeting function only during tumor killing and does not bind to CD3 after the killing function is finished. By overcoming the above two technical difficulties, the invention regarding CAR-T cell therapy for peripheral T-cell lymphoma was completed.

【선행기술문헌】 【Prior art literature】

【특허문헌】 【Patent Document】

(특허문헌 0001) 한국특허공개공보 제 2018- 0031727호 (공개일 : 2018.03.28)(Patent Document 0001) Korean Patent Publication No. 2018-0031727 (Publication Date: 2018.03.28)

【발명의 상세한 설명】 【Detailed description of the invention】

【기술적 과제】 본 발명은 (a) 표적세포 상의 CD3에 결합하는 제 1 모이어티 ; 및 (b) 키메라 항원 수용체 (Chimer i c ant i gen receptor )의 세포외 도메인에 특이적으로 결합하는 제 2 모이어티를 포함하는 , 키메라 항원 수용체 T 세포 (CAR- T 세포)의 활성을 조절하는 어댑터 분자를 제공하고자 한다. 본 발명은 상기 어댑터 분자의 키메라 항원 수용체 T 세포의 활성화를 유도하는 용도를 제공하고자 한다. 본 발명은 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포) 로서 , 상기 CAR-T 세포는 ( i ) 상기 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편을 포함하는 세포외 도메인 , ( i i ) 막횡단 도메인 , 및 ( i i i ) 세포내 신호전달 도메인을 포함하는 키메라 항원 수용체를 발현하는 것인 CAR-T 세포를 제공하고자 한다. 본 발명은 상기 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 복합체를 제공하고자 한다. 본 발명은 상기 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포) 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 T세포 유래 종양을 예방 또는 치료하기 위한 키트를 제공하고자 한다. 본 발명은 상기 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포) 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 , T세포 유래 종양을 예방 또는 치료하기 위한 약학 조성물을 제공하고자 한다. 본 발명은 상기 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포를 포함하고 , 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자와 병용 투여되는 , T세포 유래 종양을 예방 또는 치료하기 위한 약학 조성물을 제공하고자 한다. 본 발명은상기 복합체 또는 약학조성물을 개체에 투여하는단계를 포함하는 T세포 유래 종양을 예방또는 치료하는 방법을 제공하고자 한다. 본 발명은 (a) T 세포에서 CD3 또는 T 세포 수용체 (TCR)의 발현을 하향 조절하는 단계, 및 (b) 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편을 포함하는 세포외 도메인, 막횡단 도메인, 및 세포내 신호전달 도메인을 포함하는 키메릭 항원 수용체 (CAR)를 T 세포에 도입하는 단계를 포함하며, 상기 (a) 및 (b) 단계는 순서와 상관없이 수행되는, CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포)를 제조하는 방법을 제공하고자 한다. 【Technical Problem】 The present invention provides an adapter molecule that regulates the activity of a chimeric antigen receptor T cell (CAR-T cell), comprising (a) a first moiety that binds to CD3 on a target cell; and (b) a second moiety that specifically binds to an extracellular domain of a chimeric antigen receptor. The present invention also provides a use of the adapter molecule for inducing activation of a chimeric antigen receptor T cell. The present invention also provides a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to the second moiety of the adapter molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain. The present invention seeks to provide a complex comprising a chimeric antigen receptor T cell with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell. The present invention seeks to provide a kit for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell. The present invention seeks to provide a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) with reduced or eliminated expression of CD3 and an adapter molecule regulating the activity of the CAR-T cell. The present invention seeks to provide a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell with reduced or eliminated expression of CD3 and co-administered with an adapter molecule regulating the activity of the CAR-T cell. The present invention provides a method for preventing or treating a T cell-derived tumor, comprising a step of administering the complex or pharmaceutical composition to a subject. The present invention provides a method for producing a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or eliminated, comprising (a) a step of down-regulating the expression of CD3 or a T cell receptor (TCR) in a T cell, and (b) a step of introducing a chimeric antigen receptor (CAR) comprising an extracellular domain, a transmembrane domain, and an intracellular signaling domain, wherein the steps (a) and (b) are performed regardless of the order.

【기술적 해결방법】 본 발명의 일 양상은 (a)표적세포상의 CD3에 결합하는제 1모이어티; 및 (b) 키메라 항원 수용체 (Chimeric antigen receptor)의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를포함하는, 키메라항원수용체 T세포 (CAR- T세포)의 활성을 조절하는 어댑터 분자를 제공한다. 본 발명의 다른 양상은 상기 어댑터 분자의 키메라 항원 수용체 T 세포의 활성화를 유도하는 용도를제공한다. 본 명세서에서 "어댑터 분자 (adaptor molecule)"는상기 키메라항원수용체를 이용한 T 세포 치료제, 즉 CAR- T라고 불리는 세포치료제에 있어서 , CAR의 세포외 도메인과 결합하고, 동시에, 표적화하는 세포, 예를 들어 표적화하는 종양세포와 결합하는 분자 (molecule)를의미한다. 어댑터 분자는 T세포매개 종양세포또는 암에 대해 표적화하는특이적 결합능을가지므로, 어댑터분자를교체함으로써 CAR- T세포가 공격하는 종양표적을 바꿀 수 있으며, CAR- T세포의 과도한 활성화로 인해 부작용 발생시 어댑터 분자의 투여 중지 혹은 투여용량의 감소를 통해 CAR 세포의 독성을 완화할 수 있게 한다 (Cao et al . , Angew Chem Int Ed Engl . 2016 June 20; 55(26) : 7520-7524.). 본 명세서에서 , 용어 “항체 (antibody)” 는특정 항원에 특이적으로결합하는 항체로서 , 완전한항체 형태뿐만아니라항체 분자의 항원 결합단편 (antigen binding fragment)을포함한다. 완전한항체는 2개의 전체 길이의 경쇄 및 2개의 전체 길이의 중쇄를 가지는구조이며 각각의 경쇄는중쇄와다이설파이드 결합으로연결되어 있다. 중쇄 불변 영역은 감마 (X), 뮤 ( U ), 알파 U), 델타 ( 6 ) 및 엡실론 (心 타입을 가지고 서브클래스로 감마 1(X1), 감마 2(x2), 감마 3(x3), 감마 4(x4), 알파 l( al) 및 알파 2( a2)를 가진다. 경쇄의 불변영역은 카파 ( K ) 및 람다 (入) 타입을 가진다 (Cellular and Molecular Immunology, Wonsiewicz , M. J . , Ed. , Chapter 45, pp. 41—50, W. B. Saunders Co. Phi ladelphia, PA(1991) : Nisonof f , A. , Introduction to Molecular Immunology, 2nd Ed . , Chapter 4,pp. 45—65, sinauer Associates , Inc. , Sunder 1 and , MA (1984)) . 본 명세서에서, 용어 “항원 결합 단편 (antigen binding fragment)” 은 항원 결합 기능을 보유하고 있는 단편을 의미하며, Fab, F(ab'), 및 Fv, scFv, 또는 sdAB ( s i ng 1 e-doma i n antibody) 등을 포함한다. 항체 단편 중 Fab( fragment antigen binding)는 경쇄 및 중쇄의 가변영역과 경쇄의 불변 영역 및 중쇄의 첫 번째 불변 영역 (CH1)을 가지는 구조로 1개의 항원 결합 부위를 가진다. Fab '는 중쇄 CH1 도메인의 C-말단에 하나이상의 시스테인잔기를포함하는힌지 영역 (hinge region)을 가진다는 점에서 Fab와 차이가 있다. Fv는 중쇄 가변부위 및 경쇄 가변부위만을 가지고 있는 최소의 항체조각으로서, Fv 단편을 생성하는 재조합 기술은 PCT 국제 공개특허출원 W0 88/10649, W0 88/106630, W0 88/07085, W0 88/07086 및 W0 88/09344에 개시되어 있다. 본 명세서에서, 용어 “중쇄” 는항원에 특이성을부여하기 위한충분한 가변 영역 서열을 갖는 아미노산서열을 포함하는 항체의 가변 영역 도메인 VH 및 3개의 불변 영역 도메인 CHI, CH2 및 CH3를 포함하는 전체길이 중쇄 및 이의 단편을 모두 의미한다. 또한 본 명세서에서 용어 “경쇄” 는 항원에 특이성을 부여하기 위한 충분한 가변영역 서열을갖는아미노산 서열을 포함하는항체의 가변 영역 도메인 VL 및 불변 영역 도메인 CL을포함하는 전체길이 경쇄 및 이의 단편을 모두 의미한다. 본 명세서에서, 용어 “CDR( complementarity determining region: 상보성 결정부위 )” 은 면역글로블린 중쇄 및 경쇄의 고가변 영역 (hypervar iable region)의 아미노산서열을 의미한다 (Rabat et al . , Sequences of Proteins of Immunological Interest , 4th Ed. , U.S. Department of Health and Human Services , National Institutes of Health (1987)). 중쇄 (CDRH1, CDRH2 및 CDRH3) 및 경쇄 (CDRL1, CDRL2 및 CDRL3)에는각각 3개의 CDRs이 포함되어 있다. CDR은항체가항원또는에피토프에 결합하는 데 있어서 주요한접촉 잔기를 제공한다. 일 실시예에 있어서 , 상기 제 1 모이어티는 CD3에 특이적으로 결합능을 갖는 단백질을 의미할수 있다. 보다구체적으로, 제 1 모이어티는 표적세포, 예를 들면 T 세포 유래 종양세포의 표면에 발현하는 CD3에 특이적으로결합하는단백질을포함한다. 이 때, CD3에 특이적으로 결합능을 갖는 단백질은 항체, 항원 결합 단편, 애피바디 (aff ibody) , 다이아바디 (diabody) 및 앱타머 (aptamer )로 이루어진 군으로부터 선택되는 어느하나인 것일 수 있다. 또한, 상기 CD3에 특이적으로 결합하는 단백질은 T 세포 매개 종양세포의 표면에 있는 CD3분자에 결합하는단백질로서 , 상기 단백질은항체, 구체적으로단일 클론 항체, 다클론 항체 또는 재조합 항체 중 어느 하나일 수 있다. 또한, 상기 항체는 전장 형태의 항체 및 항체의 단편일 수 있다. 이때, 항체의 단편은 CD3와 결합 능력이 있는항- CD3항체의 일부분을포함할수 있다. 상기 항체의 단편은 Fab, Fab ' , F(ab' )2, Fv, scFv 또는 싱글도메인 항체 (single- domain antibody: sdAB)인 것일 수 있다. 상기 제 1모이어티는 CD3에 특이적으로 결합능을 갖는공지의 항체 또는 항원 단편 결합일 수 있다. 보다 구체적으로, 공지의 항- CD3 항체인 0KT3, UCHT1, Tep 1 i zumab , Otel ixizumab, Vi s i 1 i zumab , 및 Foralumab로부터 선택된 어느 하나의 중쇄 가변 영역 및/또는 경쇄 가변 영역의 상보성 결정 영역 (complementarity determining region, CDR) , 구체적으로 CDR1, CDR2, 또는 CDR3을 포함하는 것일 수 있다. 상기 공지의 항- CD3는 예시에 불과하고 이에 제한되지 않으며 , 상기 항체 또는 이의 결합 단편은 공지의 항- CD3 항체 각각의 CDR1, CDR2, 또는 CDR3의 조합을 포함하는 것일 수 있다. 상기 제 1 모이어티는서열번호 23 내지 25의 아미노산서열로 각각 표시되는 CDR1, CDR2, 및 CDR3를 포함하는 경쇄 가변 영역, 또는 서열번호 26 내지 28의 아미노산 서열로 각각 표시되는 CDR1, CDR2, 및 CDR3를 포함하는 중쇄 가변 영역을 포함할 수 있다. 또한, 이는 서열번호 29의 아미노산 서열을 포함하는 경쇄 가변 영역 및/또는서열번호 30의 아미노산서열을 포함하는중쇄 가변 영역을 포함할수 있다. 본 발명의 일실시예에서, 상기 서열번호 29의 아미노산서열을 포함하는 경쇄 가변 영역과서열번호 30의 아미노산서열을포함하는 중쇄 가변 영역 사이에 링커를 포함하는 것일 수 있다. 상기 링커는 서열번호 1의 아미노산 서열 ((G4Sh 링커)을 가질 수 있으나, 링커의 종류를 이에 한정하는 것은 아니다. 상기 제 1 모이어티는서열번호 32 내지 34의 아미노산서열로 각각 표시되는 CDR1, CDR2, 및 CDR3를 포함하는 경쇄 가변 영역, 또는 서열번호 35 내지 37의 아미노산 서열로 각각표시되는 CDR1, CDR2, 및 CDR3를포함하는것일수있다. 또한, 이는 서열번호 38의 아미노산 서열을 포함하는 경쇄 가변 영역 및/또는 서열번호 39의 아미노산서열을 포함하는중쇄 가변 영역을 포함할수 있다. 상기 제 1 모이어티는서열번호 67 내지 69의 아미노산서열로 각각 표시되는 CDR1, CDR2, 및 CDR3를 포함하는 경쇄 가변 영역, 또는 서열번호 70 내지 72의 아미노산 서열로 각각 표시되는 CDR1, CDR2, 및 CDR3를 포함하는 중쇄 가변 영역을 포함할 수 있다. 또한, 이는 서열번호 73의 아미노산 서열을 포함하는 경쇄 가변 영역 및/또는서열번호 75의 아미노산서열을 포함하는중쇄 가변 영역을 포함할수 있다. 본 발명의 일실시예에서, 상기 서열번호 73의 아미노산서열을 포함하는 경쇄 가변 영역과서열번호 75의 아미노산서열을포함하는 중쇄 가변 영역 사이에 링커를 포함하는 것일수 있다. 상기 링커는서열번호 74의 아미노산서열을가질수 있으나, 링커의 종류를 이에 한정하는 것은 아니다. 본발명의 일실시예에서, 상기 제 1모이어티는서열번호 13, 22, 31, 및 40의 아미노산 서열을 갖는 scFv 단편, 또는 서열번호 41 및 42의 아미노산 서열을 포함하는 Fab 단편일 수 있다. 또한, 상기 항 CD3 항체 또는 이의 결합 단편은 서열번호 13, 22, 31, 40, 41, 및 42의 아미노산 서열, 이들의 일부, 또는 이들과 적어도 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%또는 99% 이상서열 동일성을 갖는 아미노산 서열을 포함할수 있다. 일 실시예에 있어서 , 상기 제 2 모이어티는 펩타이드 태그 또는 합텐 태그인 것일 수 있다. 상기 태그는 His 태그, Myc 태그, 코티닌 태그, FITC 태그, Biotin 태그, Leucin zipper 태그, Flag 태그, Xpress 태그, Avi 태그, 칼모듈린 결합 펩타이드 (CBP) 태그, 폴리글루타메이트 태그, HA 태그, Strep 태그, Softag 1, Softag 3, 및 V5 태그로 이루어진 군으로부터 선택되는 것일 수 있다. 본 발명의 일 실시예에 있어서 , 상기 제 1 모이어티는 항 CD3 항체 또는 항원 결합 단편이거나/이고 상기 제 2모이어티는 His 태그인 것일 수 있다. 본 발명의 일 실시예에 있어서 , 상기 제 1 및 제 2모이어티를포함하는 키메라 항원 수용체 T세포의 활성을조절하는 어댑터 분자는서열번호 13, 22, 31, 및 40의 아미노산 서열을 갖는 scFv 단편, 또는 서열번호 41 및 42의 아미노산 서열을 포함하는 Fab 단편에 6XHis(HHHHHH)를 더 추가한 것일 수 있다. 또한, 상기 어댑터 분자는 서열번호 42및 44가결합된 형태의 Fab, 서열번호 46및 48이 결합된 형태의 Fab, 서열번호 50 및 52이 결합된 형태의 Fab, 또는 서열번호 54 및 56이 결합된 형태의 Fab을 포함하는 것일 수 있다. 하지만, 이에 제한되는 것은 아니다. 일 구체예에 있어서, 상기 경쇄 가변 영역 및 중쇄 가변 영역은 링커를 통해 연결되어 있을수 있다. 예를들어 , 상기 링커는, 1내지 400개, 1내지 200개, 또는 2 내지 200개의 임의의 아미노산으로 이루어진 폴리펩티드일 수 있다. 상기 펩티드 링커는 Gly, Asn 및 Ser 잔기를 포함할 수 있으며, Thr 및 Ala과 같은 중성 아미노산들도 포함될 수 있다. 펩티드 링커에 적합한 아미노산 서열은 당업계에 공지되어 있다. 또한 기능적 일부분 사이의 적절한 분리를 달성하기 위하여 또는 필수적인 내부-일부분 (inter- moiety)의 상호작용을 유지하기 위한 링커의 최적화를 고려하여 카피 수 "n"을 조절할 수 있다. 해당 기술분야에서 다른 가요성 링커들이 알려져 있는데, 예를 들어 수용성을 향상시키기 위하여 극성 아미노산 잔기를 추가하는 것뿐만 아니라 유연성을 유지하기 위하여 T 및 A와 같은 아미노산 잔기를 추가한 G및 S링커가 있을수 있다. 따라서 일 구체예에 있어서 , 상기 링커는 G, S, 및/또는 T 잔기를 포함하는 유연성 링커일 수 있다. 상기 링커는 (GpSs)n 및 (SpGs)n으로부터 선택되는 일반식을가질 수 있고, 이 경우, 독립적으로, 日는 1내지 10의 정수이고, s = 0 내지 10의 0또는 정수이고, p + s는 20 이하의 정수이고, 및 n은 1 내지 20의 정수이다. 더욱 구체적으로 링커의 예는 (GGGGS)n상기 n은 1 내지 20, 또는 1 내지 10의 정수이다. 본 발명의 일 실시예에 있어서 , CD3과 특이적으로 결합하는 항원 결합단편으로 scFv를제조할시, 경쇄 가변 영역과중쇄 가변 영역은 (G4S)3링커 (서열번호 1)를사용하였다. 본 발명의 다른 양상은 상기 어댑터 분자의 키메라 항원 수용체 T 세포의 활성화를 유도하는 용도를제공한다. 【Technical Solution】 One aspect of the present invention provides an adapter molecule that regulates the activity of a chimeric antigen receptor T cell (CAR-T cell), comprising (a) a first moiety that binds to CD3 on a target cell; and (b) a second moiety that specifically binds to an extracellular domain of a chimeric antigen receptor. Another aspect of the present invention provides a use of the adapter molecule to induce activation of a chimeric antigen receptor T cell. As used herein, the term "adaptor molecule" means a molecule that binds to the extracellular domain of a CAR and, at the same time, binds to a target cell, for example, a target tumor cell, in a T cell therapy using the chimeric antigen receptor, i.e., a cell therapy agent called CAR-T. Since the adapter molecule has a specific binding ability to target T cell-mediated tumor cells or cancer, the tumor target attacked by the CAR-T cell can be changed by replacing the adapter molecule, and when side effects occur due to excessive activation of the CAR-T cell, the toxicity of the CAR cell can be alleviated by stopping the administration of the adapter molecule or reducing the administration dose (Cao et al., Angew Chem Int Ed Engl. 2016 June 20; 55(26) : 7520-7524.). As used herein, the term “antibody” refers to an antibody that specifically binds to a specific antigen, and includes not only a complete antibody form but also an antigen binding fragment of the antibody molecule. A complete antibody has a structure having two full-length light chains and two full-length heavy chains, and each light chain is connected to a heavy chain by a disulfide bond. The heavy chain constant region has gamma (X), mu (U), alpha (U), delta (6), and epsilon (心) types and has subclasses gamma 1 (X1), gamma 2 (x2), gamma 3 (x3), gamma 4 (x4), alpha l (al), and alpha 2 (a2). The light chain constant region has kappa (K) and lambda (入) types. (Cellular and Molecular Immunology, Wonsiewicz, M. J., Ed., Chapter 45, pp. 41—50, WB Saunders Co. Philadelphia, PA(1991) : Nisonof f, A., Introduction to Molecular Immunology, 2nd Ed., Chapter 4,pp. 45—65, Sinauer Associates, Inc., Sunder 1 and , MA (1984)). As used herein, the term “antigen binding fragment” means a fragment having antigen binding function, and includes Fab, F(ab'), and Fv, scFv, or sdAB (si ng 1 e-doma in antibody). Among antibody fragments, Fab (fragment antigen binding) has a structure having variable regions of the light and heavy chains, constant region of the light chain, and first constant region (CH1) of the heavy chain, and has one antigen binding site. Fab' differs from Fab in that it has a hinge region containing one or more cysteine residues at the C-terminus of the heavy chain CH1 domain. Fv is the minimum antibody fragment having only heavy chain variable region and light chain variable region, and recombinant technology for producing Fv fragments is disclosed in PCT International Patent Publication Nos. W0 88/10649, W0 88/106630, W0 88/07085, W0 88/07086, and W0 88/09344. As used herein, the term “heavy chain” refers to a full-length heavy chain and fragments thereof comprising an antibody variable domain VH and three constant domains CHI, CH2 and CH3, which comprise an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen. In addition, the term “light chain” as used herein refers to a full-length light chain and fragments thereof comprising an antibody variable domain VL and constant domain CL, which comprise an amino acid sequence having sufficient variable region sequence to confer specificity to an antigen. As used herein, the term “complementarity determining region (CDR)” refers to an amino acid sequence of a hypervariable region of an immunoglobulin heavy chain or light chain (Rabat et al., Sequences of Proteins of Immunological Interest, 4th Ed., US Department of Health and Human Services, National Institutes of Health (1987)). The heavy chain (CDRH1, CDRH2 and CDRH3) and the light chain (CDRL1, CDRL2 and CDRL3) each contain three CDRs. The CDRs provide key contact residues for binding of the antibody to an antigen or epitope. In one embodiment, the first moiety may mean a protein having a specific binding ability to CD3. More specifically, the first moiety includes a protein that specifically binds to CD3 expressed on the surface of a target cell, for example, a T cell-derived tumor cell. In this case, the protein having a specific binding ability to CD3 may be any one selected from the group consisting of an antibody, an antigen-binding fragment, an affibody, a diabody and an aptamer. In addition, the protein that specifically binds to CD3 is a protein that binds to a CD3 molecule on the surface of a T cell-mediated tumor cell, and the protein may be an antibody, specifically, any one of a monoclonal antibody, a polyclonal antibody, or a recombinant antibody. In addition, the antibody may be a full-length antibody or an antibody fragment. At this time, the antibody fragment may include a part of an anti-CD3 antibody having the ability to bind to CD3. The antibody fragment may be Fab, Fab', F(ab')2, Fv, scFv, or a single-domain antibody (sdAB). The first moiety may be a known antibody or antigen fragment binding having the ability to specifically bind to CD3. More specifically, it may include a complementarity determining region (CDR), specifically, CDR1, CDR2, or CDR3, of a heavy chain variable region and/or a light chain variable region selected from the known anti-CD3 antibodies 0KT3, UCHT1, Tep 1 i zumab, Otel ixizumab, Vi si 1 i zumab, and Foralumab. The known anti-CD3 is only an example and is not limited thereto, and the antibody or binding fragment thereof may include a combination of CDR1, CDR2, or CDR3 of each of the known anti-CD3 antibodies. The first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 23 to 25, respectively, or a heavy chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 26 to 28, respectively. Additionally, it may include a light chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and/or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 30. In one embodiment of the present invention, a linker may be included between the light chain variable region comprising the amino acid sequence of SEQ ID NO: 29 and the heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 30. The linker may have an amino acid sequence of SEQ ID NO: 1 ((G4Sh linker), but the type of the linker is not limited thereto. The first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 32 to 34, respectively, or CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 35 to 37, respectively. In addition, it may include a light chain variable region comprising an amino acid sequence of SEQ ID NO: 38 and/or a heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 39. The first moiety may include a light chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 67 to 69, respectively, or a heavy chain variable region comprising CDR1, CDR2, and CDR3 represented by amino acid sequences of SEQ ID NOs: 70 to 72, respectively. In addition, it ... A light chain variable comprising the amino acid sequence of 73 A heavy chain variable region comprising an amino acid sequence of SEQ ID NO: 75 may be included. In one embodiment of the present invention, a linker may be included between the light chain variable region comprising the amino acid sequence of SEQ ID NO: 73 and the heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 75. The linker may have the amino acid sequence of SEQ ID NO: 74, but the type of the linker is not limited thereto. In one embodiment of the present invention, the first moiety may be a scFv fragment having the amino acid sequences of SEQ ID NOs: 13, 22, 31, and 40, or a Fab fragment comprising the amino acid sequences of SEQ ID NOs: 41 and 42. Additionally, the anti-CD3 antibody or binding fragment thereof can comprise an amino acid sequence of SEQ ID NOS: 13, 22, 31, 40, 41, and 42, a portion thereof, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% sequence identity thereto. In one embodiment, the second moiety can be a peptide tag or a hapten tag. The tag can be selected from the group consisting of a His tag, a Myc tag, a cotinine tag, a FITC tag, a Biotin tag, a Leucin zipper tag, a Flag tag, an Xpress tag, an Avi tag, a calmodulin binding peptide (CBP) tag, a polyglutamate tag, an HA tag, a Strep tag, a Softag 1, a Softag 3, and a V5 tag. In one embodiment of the present invention, the first moiety may be an anti-CD3 antibody or an antigen-binding fragment and/or the second moiety may be a His tag. In one embodiment of the present invention, the adapter molecule regulating the activity of a chimeric antigen receptor T cell comprising the first and second moieties may be a scFv fragment having amino acid sequences of SEQ ID NOs: 13, 22, 31, and 40, or a Fab fragment comprising amino acid sequences of SEQ ID NOs: 41 and 42, wherein 6XHis (HHHHHH) is further added. In addition, the adapter molecule may include a Fab in a form in which SEQ ID NOs: 42 and 44 are linked, a Fab in a form in which SEQ ID NOs: 46 and 48 are linked, a Fab in a form in which SEQ ID NOs: 50 and 52 are linked, or a Fab in a form in which SEQ ID NOs: 54 and 56 are linked. However, the present invention is not limited thereto. In one embodiment, the light chain variable region and the heavy chain variable region may be connected via a linker. For example, the linker may be a polypeptide consisting of any amino acid of 1 to 400, 1 to 200, or 2 to 200. The peptide linker may include Gly, Asn, and Ser residues, and may also include neutral amino acids such as Thr and Ala. Suitable amino acid sequences for the peptide linker are known in the art. Optimization of the linker may also be performed to achieve appropriate separation between functional moieties or to maintain essential inter-moiety interactions. The copy number " n " can be controlled by considering. Other flexible linkers are known in the art, for example, G and S linkers which add amino acid residues such as T and A to maintain flexibility as well as adding polar amino acid residues to improve solubility. Thus, in one embodiment, the linker can be a flexible linker comprising G, S, and/or T residues. The linker can have a general formula selected from (GpSs) n and (SpGs) n , wherein independently, 日 is an integer from 1 to 10, s = 0 or an integer from 0 to 10, p + s is an integer less than or equal to 20, and n is an integer from 1 to 20. A more specific example of a linker is (GGGGS) n wherein n is an integer from 1 to 20, or from 1 to 10. In one embodiment of the present invention, when producing scFv as an antigen-binding fragment that specifically binds to CD3, the light chain variable region and the heavy chain variable region used a (G4S)3 linker (SEQ ID NO: 1). Another aspect of the present invention provides the use of the adapter molecule to induce activation of a chimeric antigen receptor T cell.

CAR T 세포 치료요법에 있어서 , 개체의 생체 내 투입된 CAR T세포가 종양을 선택적으로 살상하는데 사용된 후, 종양이 유효하게 감소한 후, 남아있는 잔여 CAR T 세포의 정상세포에 대한 살상의 부작용이 나타난다. 이를 CAR T의 면역결핍독성 또는 정상 T세포 독성이라 한다. 상기 어댑터 분자는 키메라 항원 수용체 T세포의 활성화를 조절하여 CAR T 세포의 면역결핍독성 또는 정상 T 세포 독성 회피능의 효과를 보였다. 본 발명의 일 실시예에 있어서, 본 발명의 CAR T 세포는 기존의 종양을 직접적으로 표적화하는 기존의 CAR T와 달리, CAR T 세포 자체는 어댑터 분자와 결합하고, 어댑터 분자로하여금표적화 하는 종양세포와 결합하는 것인 바, 어댑터 분자의 부재하에서 CAR T 세포를 주입하게 되어도, 표적 세포 또는 종양에 대해 독성을 나타나지 않았다. 즉, CAR T세포는 어댑터 분자의 존재 하에서만 활성화를 유도하고, 이에 표적 세포또는 종양에 대해 독성을 나타낸다. 따라서, 상기 어댑터 분자는 CAR T 세포의 활성화를 유도하거나, 비활성화를 유도할 수 있다. 이런 이유에서 어댑터 분자를 스위처블 (switchable) 분자라고도 한다. 이에, 상기 어댑터 분자의 키메라 항원 수용체 T 세포의 활성화를 유도하는 용도는 CAR T세포치료 요법 후, 잔여하는 CAR T의 정상 세포에 세포 독성 및 이에 의한 정상 T세포살상 부작용을 완화하는 면역결핍 방지능의 용도를 포함한다. 본 발명의 또 다른 양상은 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포)로서, 상기 CAR-T 세포는 (i) 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합단편을 포함하는 세포외 도메인, (ii) 막횡단 도메인, 및 (iii) 세포내 신호전달 도메인을 포함하는 키메라 항원 수용체를 발현하는 것인 CAR-T세포를 제공한다. 상기 "제 1 모이어티", "제 2 모이어티" 또는 "어댑터 분자"에 대해서는 전술한 바와 같다. 본 명세서에서 용어 ''세포외 도메인 (extracel lular domain)"은 세포 외부로 돌출되어 리간드 등과 결합하는 도메인을 의미한다. 본 명세서에서 용어 ''세포내 도메인 (intracellular domain)"은세포의 세포막 안쪽, 즉 세포질에 위치하게 되는 부분으로서, 세포외 도메인과 리간드가 결합되어 전달되는 신호를 세포내로전달하는 도메인을 의미한다. 일 구체예에 있어서 , 상기 키메릭 항원 수용체 (이하, 'CAR'라고도 한다. )란 T-세포 활성화 단백질 (CD3- zeta chain, CD28, 41BBL, 0x40, I COS, high-affinity receptor for IgE (FCERI) 및 기타 T-세포 활성화 단백질)의 세포막 혹은 세포내 신호전달부위를 암 항원 특이적 항체의 항원결합부위 (single chain Fv fragment)와 융합시킨 단백질을 통칭한다. 구체적으로, 1세대 CAR는 암세포에서 특이적으로 발현하는 항원 인식 부위를 포함하는 세포외 도메인, 막통과 도메인 및 세포내 신호전달 도메인을 포함하고, 신호전달 도메인으로서 CD3《만을 이용하는 것일 수 있다. 2세대 CAR는 면역세포에 대한 반응성 향상을위하여 공동자극도메인 (CD28또는 CD137/4- 1BB)과 CD3《를결합한 구조를 가지며 , 1세대 CAR에 비교하여 체내에 잔존하는 CAR 포함 면역세포의 수가 증가하여 나타날수있다. 3세대 CAR에서는두가지 이상의 공동자극도메인을이용여 , 생체 내 CAR를 포함하는 면역세포의 확장 및 지속성 달성을 위해 공동자극 도메인을 4-1BB, CD28 또는 0X40 등과 결합시킨 구조를 갖는 것일 수 있다. 4세대 CAR에서는 IL-12 또는 IL- 15와 같은 사이토카인을 암호화하는 추가 유전자를 포함하여, 사이토카인의 CAR 기반 면역단백질이 추가로 발현될 수 있도록 하고, 5세대 CAR는 면역세포 강화를 위해 인터루킨 리셉터 체인, 예를들어 , IL- 2RP를추가로포함하는 것일 수 있다. 일 구체예에 있어서 , 상기 키메릭 항원 수용체는 1세대 CAR내지 5세대 CAR의 구조를 가지는 것일 수 있다. 일 구체예에 있어서 , 상기 키메라항원수용체의 세포외 연결부는힌지 (hinge) 도메인을 추가로 포함하는 것일 수 있다. 일 구체예에 있어서, 상기 힌지 도메인은 올리고 펩티드 또는 폴리펩티드로 이루어지고, 1 내지 100개의 아미노산 잔기, 구체적으로는 10 내지 70 아미노산 잔기를 포함하는 것을 특징으로 할 수 있으며 , 이에 제한되는 것은 아니다. 일 구체예에 있어서, 상기 키메라 항원 수용체의 세포내 신호전달 도메인 (intracellular signal ing domain)은 면역세포의 세포막 안쪽, 즉 세포질에 위치하게 되는 부분으로서 , 세포외 도메인에 포함된 항원 결합도메인이 표적 항원에 결합하였을 때 , 세포 내에 신호를 전달하여 면역세포의 면역반응을 활성화시키는 부위를 의미할 수 있다. 일 구체예에 있어서, 상기 세포내 신호전달 도메인은 CD3 제타 (《), CD3 감마 (Y), CD3 델타 (6), CD3 엡실론 (E) , FcR 감마, FcR 베타, CD5, CD22, CD79a, CD79b 및 CD66d로구성된 군에서 선택된 하나 이상의 세포내 신호전달도메인인 것일 수 있으나, 이에 한정되는 것은 아니며 , 일 실시예에서 , 상기 키메라 항원 수용체의 세포내 신호전달 도메인은 CD3 제타 (《)이다. 일 구체예에 있어서 , 상기 세포내 신호전달도메인은추가적으로공동자극 (costimulatory) 도메인을 포함하는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다. 본 발명에 따른 공동자극 (co- stimulatory) 도메인은 CD2, CD7, CD27, CD28, CD30, CD40, 4-lBB(CD137) , 0X40(CD134) , I COS, LFA-1 , GITR, MyD88, DAP1, PD-1, LIGHT, NKG2C, B7-H3 및 CD83과 특이적으로 결합하는 리간드로구성된 군에서 선택된 하나 이상의 공동자극 도메인을 포함할 수 있으나, 이에 제한되는 것은 아니다. 일 구체예에 있어서 , 상기 키메라항원 수용체는하나 이상의 세포내 신호전달 도메인과, 하나 이상의 공동자극 도메인을 포함하는 것을 특징으로 할 수 있다. 일 구체예에 있어서, 상기 공동자극 도메인은 4- 1BB인 것일 수 있다. 상기 키메라 항원 수용체 T 세포의 CD3의 발현의 감소 또는 제거는 CAR 유전자를 도입하기 전 T 세포 또는 CAR 유전자를 도입한 CAR-T 세포의 CD3, 구체적으로는 내인성 CD3을 제거하거나, TCR을 제거하는 것에 의해 이루어지는 것일 수 있다. 즉, 상기 CD3의 발현의 감소 또는 제거는 직접적으로 CD3의 유전자를 제거하거나, TCR을제거함으로써 간접적으로 CD3의 발현을 감소, 억제 또는제거하는 것일 수 있다. 상기 CD3 또는 TCR의 발현을 감소 또는 제거하는 것은 CRISPR 시스템, 구체적으로, CRISPR/Cas9, TALEN , Zinc f i nger nuclease, base-editing 및 pr ime- editing을 포함하는 유전자 편집 기법 (genome editing)에 의해 수행되거나, 또는 안티센스 (ant isense) RNA, 안타고미르 (antagomir ) RNA, s i RNA , shRNA 및 miRNA로 이루어진 군에서 선택되는 핵산에 의해 수행되는 것일 수 있다. 상기 CAR T 세포는 상기 어댑터 분자의 제 2 모이어티와 결합하는 것일 수 있다. 보다구체적으로, CAR T세포는 상기 어댑터 분자의 제 2모이어티와 결합하여 표적화 세포 또는 종양 세포에 대하여 세포 독성을 보일 수 있다. 즉, 상기 CAR T 세포, 보다 구체적으로 CAR T 세포의 세포외 도메인은 상기 어댑터 분자의 제 2 모이어티와 결합능을 갖는항체 또는 이의 항원 결합단편을포함하는 것일 수 있다. 상기 제 2 모이어티는 His 태그일 수 있다. 따라서, 상기 CAR T 세포, 보다 구체적으로 CAR T 세포의 세포외 도메인은 His 태그에 특이적으로 결합능이 있는 단백질, 구체적으로 항체 또는 항원 결합단편을 포함하는 것일 수 있다. 본 발명의 일 실시예에서는 상기 His 태그에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편은 서열번호 63의 아미노산 서열, 이들의 일부, 또는 이들과 적어도 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%또는 99%이상동일성을갖는아미노산서열을포함하는 것일 수 있다. 본출원에서 "특정 서열번호의 유전자서열/아미노산서열을 포함하는", 또는 "특정 서열번호의 유전자 서열/아미노산 서열을 가지는"이라고 기재되어 있다 하더라도, 해당 서열번호의 유전자 서열/아미노산 서열로 구성된 것과 동일 혹은 상응하는 기능을가지는 경우라면 , 일부서열이 결실 , 변형 , 치환또는부가된유전자 서열/아미노산 서열을 갖는 것도 본 출원에서 사용될 수 있음은 자명하다. 또한 본 출원에서 유전자서열과 염기 서열은혼용될 수 있다. 예를 들어 , 상기 CAR T 세포와 동일 혹은 상응하는 기능을 가지는 경우라면 해당 서열번호의 서열 내부나 말단에 무의미한 서열이 부가되거나 혹은 해당 서열번호의 서열 내부나 말단의 일부 서열이 결실된 것도 본원의 범위 내에 속하는 것이 자명하다. 상동성 (homology) 및 동일성 (identity)은두개의 주어진 염기 서열과관련된 정도를 의미하며 백분율로 표시될 수 있다. 용어 상동성 및 동일성은 종종 상호교환적으로 이용될 수 있다. 임의의 두 서열이 상동성 또는 동일성을 갖는지 여부는 예를 들어 , Pearson et al (1988) [Proc. Natl. Acad. Sci . USA 85]: 2444에서오} 같은 디폴트 파라미터를 이용하여 "FASTA" 프로그램과 같은 공지의 컴퓨터 알고리즘을 이용하여 결정될 수 있다. 또는, EMBOSS 패키지의 니들만 프로그램 (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al . , 2000, Trends Genet . 16: 276— 277)(버전 5.0.0 또는 이후 버전)에서 수행되는 바와 같은, 니들만-운치 (Needleman- Wunsch) 알고리즘 (Needleman and Wunsch, 1970, J . Mol . Biol . 48: 443- 453)이 사용되어 결정될 수 있다 (GCG프로그램 패키지 (Devereux, J . , et al , Nucleic Acids Research 12: 387 (1984)), BLASTP, BLASTN, FASTA (Atschul, [S.] [F. ,] [ET AL, J MOLEC BIOL 215]: 403 (1990) : Guide to Huge Computers , Martin J . Bishop, [ ED . , ] Academic Press, San Diego, 1994, 및 [CARILLO ETA/.] (1988) SIAM J Applied Math 48: 1073을 포함한다). 예를 들어, 미국 국립생물공학 정보 데이터베이스 센터의 BLAST, 또는 ClustalW를 이용하여 서열의 상동성 또는 동일성을 결정할수 있다. 본 발명의 또다른 양상은상기 CD3의 발현이 감소되거나제거된 키메라항원 수용체 T 세포 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 복합체를 제공한다. 본 발명의 또다른양상은 상기 CD3의 발현이 감소되거나제거된 키메라항원 수용체 T 세포 (CAR- T 세포) 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 T세포 유래 종양을 예방또는 치료하기 위한 키트를 제공한다. 상기 "CAR T 세포", "제 1 모이어티", "제 2 모이어티" 또는 "어댑터 분자"에 대해서는 전술한바와 같다. 본 발명의 또다른 양상은상기 CD3의 발현이 감소되거나제거된 키메라항원 수용체 T 세포 (CAR- T 세포) 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는 조성물을 제공한다. 상기 조성물은 약학조성물일 수 있다. 상기 조성물은 T세포유래 종양의 예방또는 치료용 약학조성물일 수 있다. 본 발명의 또다른 양상은상기 CD3의 발현이 감소되거나제거된 키메라항원 수용체 T세포를 포함하고, 상기 CAR-T세포의 활성을 조절하는 어댑터 분자와 병용 투여되는, T세포 유래 종양을 예방또는 치료하기 위한 약학조성물을 제공한다. 상기 "CAR T 세포", "제 1 모이어티", "제 2 모이어티" 또는 "어댑터 분자"에 대해서는 전술한 바와 같다. 본 명세서에서 용어 ''병용요법 '' 또는 ''병용 치료 (combined treatment )" 또는 "병용하여 ( in combinat ion) "는 적어도 2개의 별개의 치료제들을 사용한 임의 형태의 동시 또는 병행 치료를 지칭한다. 병용 요법의 성분들은 동시에 , 순차적으로, 역순으로 또는 임의의 순서로 투여될 수 있다. 성분들은 상이한 복용량으로 또는 상이한 투여 빈도로 또는 상이한 경로를 통해 적절한 방식으로 투여될 수 있다. 본 명세서에서 용어 "투여 "란 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미하며 , "개체"란 대사질환 또는 대사조절제의 부작용을 보유할 수 있는 인간을 포함한 쥐 , 생쥐 , 가축 등의 모든 생물을 의미한다. 구체적인 예로, 인간을 포함한 포유동물일 수 있다. 본 명세서에서 용어 "동시에 투여되는"은 특별히 제한되지 않으며 , 병용 요법의 성분들이 예를 들면 혼합물로서 또는 즉시 이어지는 순서로 실질적으로 동시에 투여되는 것을 의미한다. 본 명세서에서 용어 "순차적으로 투여되는"은 특별히 제한되지 않으며 , 병용 요법의 성분들이 동시에 투여되지 않고 , 투여 사이에 특정한 시간 간격을 두고 하나씩 차례로 또는 무리지어 투여됨을 의미한다. 시간 간격은 병용 요법의 성분들의 각각의 투여 사이에서 동일하거나 상이할 수 있으며 , 예를 들면 , 2분 내지 96시간, 1일 내지 7일 또는 1주 , 2주 또는 3주의 범위에서 선택될 수 있다. 일반적으로, 투여 사이의 시간 간격은 수 분 내지 수 시간, 예를 들면 2분 내지 72시간, 30분 내지 24시간, 또는 1 내지 12시간 범위일 수 있다. 추가의 예는 24 내지 96시간, 12 내지 36시간, 8 내지 24시간, 및 6 내지 12시간 범위의 시간 간격을 포함한다. 본 명세서에서 있어서 용어 "질환'은 하나의 병리적 상태 , 특히 암, 감염성 질환, 염증성 질환, 퇴행성 질환, 세포사멸 관련 질환 및 이식편 거부를 의미할 수 있다. 본 명세서에서 용어 "치료"는 질환, 장애 또는 병태 , 또는 그의 하나 이상의 증상의 경감, 진행 억제 또는 예방을 지칭하거나, 그를 포함하며 , "유효성분" 또는 "약제학적 유효량"은 질환, 장애 또는 병태 , 또는 그의 하나 이상의 증상의 경감, 진행 억제 또는 예방에 충분한 본원에서 제공되는 발명을 실시하는 과정에서 이용되는 조성물의 임의의 양을 의미할 수 있다. 본 명세서에서 용어 , "투여하는 , " "도입하는" 및 "이식하는"은 상호교환적으로 사용되고 일 구체예에 따른 조성물의 원하는 부위로의 적어도 부분적 국소화를 초래하는 방법 또는 경로에 의한 개체내로의 일 구체예에 따른 조성물의 배치를 의미할 수 있다. 일 구체예에 따른 조성물의 세포 또는 세포 성분의 적어도 일부를 생존하는 개체 내에서 원하는위치로전달하는 임의의 적절한경로에 의해 투여될수 있다. 개체 투여 후 세포의 생존 기간은 짧으면 수 시간, 예를 들면 24시간 내지 수일 내지 길면 수년일 수 있다. 상기 T세포 유래 종양은 T세포 전림프구성 백혈병 (T- cell prolymphocyt ic leukemia) , T세포 거대과립 림프구성 백혈병 (T- cell large granular lymphocytic leukemia) , NK세포의 만성 림프증식성 질환 (chronic lymphoprol i ferat ive disorder of NK cells), 공격성 NK백혈병 (aggressive NK leukemia) , 소아전신 EBV양성 T세포 림프증식성 질환 (systemic EBV-positive T-cel 1 lymphoprol i ferat ive disease of chi Idhood) , 하이드로아 백신 유사 림프종 (hydroa vaccini forme- 1 ike lymphoma) , 성인 T세포 백혈병/림프종 (adult T-cell leukemi a/ lymphoma) , 림프절 외 NK/T세포 림프종 (비강 형 ) (extranodal NK/T-cel 1 lympho (nasal type)) , 장병증 관련 T세포 림프종 (enteropathy- associated T-cel 1 lymphoma) , 간비장 T세포 림프종 (hepatosplenic T-cel 1 lymphoma) , 피하지방중염 유사 T세포 림프종 (subcutaneous panniculitis-like T-cel 1 lymphoma) , 균상 식육종 (mycosis fungoides) , 세자리 증후군 (Se'zary syndrome) , 원발성 피부 CD30 양성 T-세포 림프증식성 질환 (primary cutaneous CD30- posit ive T-cel 1 lymphoprol i ferat ive disorders) , 림프종모양구진증 ( lymphomatoid papulosis) , 원발성 피부 역형성 대세포 림프종 (primary cutaneous anaplastic large cel 1 lymphoma) , 원발성 피부감마/델타 T세포 림프종 (primary cutaneous gamma/delta T-cel 1 lymphoma) , 원발성 피부 CDS 양성 공격성 표피친화성 세포독성 T세포 림프종 (primary cutaneous CD8- positive aggressive epidermotropic cytotoxic T-cel 1 lymphoma) , 원발성 피부 CD4양성 소/중 T세포 림프종 (primary cutaneous CD4— positive smal 1/medium T-cel 1 lymphoma) , 말초 T세포 림프종 NOS (달리 명시되지 않음) (per ipheral T-cel 1 lymphoma N0S(not otherwise specified)) , 혈관면역모구 T세포 림프종 (angio immunob last ic T-cel 1 lymphoma) , ALK 양성 역형성 대세포 림프종 (ALK positive anaplastic large cel 1 lymphoma) 및 ALK음성 역형성 대세포 림프종 (ALK negat ive anaplastic large cel 1 lymphoma)로구성된 군으로부터 선택되는 것일 수 있다. 상기 투여는 추가적인 항암제와 병용 투여되는 것일 수 있다. 추가적인 항암제의 예시는 알킬화제 (alkylating agent), 안티메타볼라이트, 방추체 저해제 식물 알칼로이드, 세포 장애성/항종양 항생 물질, 토포이소머라아제 저해약, 항체, 광 증감제 및 키나아제 저해약이 포함될 수 있다. 상기 항암제의 예시는 타겟팅 요법과 종래의 화학 요법에 사용되는 화합물을 포함할 수 있다. 또한, 상기 항체의 예시는 알렘투주맙, 아폴리주맙, 아셀리주맙, 아틀리주맙, 바피뉴주맙, 베바시주맙, 비바투주맙 메르탄신 , 칸투주맙 메르탄신 , 세델리주맙, 세르톨리주맙페골 , 시드푸시투주맙 , 시드투주맙 , 다클리주맙 , 에쿨리주맙 , 에팔리주맙 , 에프라투주맙 , 에를리주맙, 펠비주맙, 폰톨리주맙, 겜투주맙 오조가마이신 , 이노투주맙 오조가마이신 , 이필리무맙, 라베투주맙, 린투주맙, 마투주맙, 메폴리주맙, 모타비주맙, 모토비주맙, 나탈리주맙, 니모투주맙, 놀로비주맙, 누마비주맙, 오크렐리주맙, 오말리주맙, 팔리비주맙, 파스콜리주맙, 펙푸시투주맙, 펙투주맙, 퍼투주맙, 펙셀리주맙, 랄리비주맙, 라니비주맙, 레슬리비주맙, 레슬리주맙, 레사이비주맙, 로벨리주맙, 루플리주맙, 시브로투주맙, 시플리주맙, 손투주맙, 타카투주맙 테트락세탄 , 타도시주맙 , 탈리주맙 , 테피바주맙 , 토실리주맙 , 토랄리주맙 , 트라스투주맙 , 투코투주맙 셀몰류킨 , 투쿠시투주맙 , 우마비주맙 , 우르톡사주맙 및 비실리주맙이 포함될 수 있다. 본 발명에 따른조성물은 약학적으로 허용되는 담체를추가로포함할 수 있다. 주사제의 경우에는 완충제, 보존제, 무통화제, 가용화제, 등장제, 안정화제 등을 혼합하여 사용할 수 있으며, 국소 투여용의 경우에는 기제, 부형제, 윤활제, 보존제 등을 사용할 수 있다. 본 발명의 약제학적 조성물의 제형은 상술한 바와 같은 약제학적으로 허용되는 담체와 혼합하여 다양하게 제조될 수 있다. 예를 들어, 주사제의 경우에는 단위 투약 앰플 또는 다수회 투약 형태로 제조할 수 있다. 또한, 상기 항암 조성물은 전형적으로 막을 통과한 이동을 용이하게 하는 계면활성제를 포함할 수 있다. 이러한 계면활성제는 스테로이드에서 유도된 것이거나 N-[l-(2,3 - 디올레오일)프로필- N,N,N-트리메틸암모늄클로라이드 (DOTMA) 등의 양이온성 지질 , 또는 콜레스테롤 헤미숙시네이트, 포스파티딜 글리세롤등의 각종화합물 등이 있다. 일 구체예에 있어서, 상기 조성물은 암세포 또는 그들의 전이를 치료하기 위하여, 또는 암의 성장을 억제하기 위하여 약학적으로 효과적인 양으로 투여될 수 있다. 암종류, 환자의 연령, 체중, 증상의 특성 및 정도, 현재 치료법의 종류, 치료 회수, 투여 형태 및 경로등다양한요인에 따라달라질 수 있으며, 통상의 기술자에 의해 용이하게 결정될 수 있다. 본 발명에 따른 조성물은 상기한 약리학적 또는 생리학적 성분을 함께 투여하거나순차적으로투여할 수 있으며 , 또한추가의 종래의 치료제와 병용하여 투여될 수 있고종래의 치료제와는순차적 또는동시에 투여될 수 있다. 이러한 투여는 단일 또는 다중 투여일 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로최대 효과를 얻을수 있는 양을투여하는것이 중요하며 , 이는 통상의 기술자에 의해 용이하게 결정될 수 있다. 본 발명의 또 다른 양상은 상기 복합체 또는 약학조성물을 개체에 투여하는 단계를 포함하는 T세포 유래 종양을 예방또는 치료하는 방법을 제공한다. 본 발명의 또 다른 양상은 (a) T 세포에서 CD3 또는 T 세포 수용체 (TCR)의 발현을 하향 조절하는 단계, 및 (b) 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합단편을포함하는세포외 도메인, 막횡단도메인, 및 세포내 신호전달도메인을 포함하는키메릭 항원수용체 (CAR)를 T세포에 도입하는 단계를 포함하며, 상기 (a) 및 (b) 단계는 순서와 상관없이 수행되는, CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포)를 제조하는 방법을 제공한다. 상기 CD3 또는 TCR의 발현을 감소 또는 제거하는 방법은 CRISPR 시스템, 구체적으로, CRISPR/Cas9, TALEN, 아연집게 단백질 (Zinc finger nuclease), 염기 편집 (base- editing) 및 프라임 편집 (prime- editing)을 포함하는 유전자 편집 기법 (genome editing)에 의해 수행되거나, 또는 안티센스 (ant isense) RNA, 안타고미르 (antagomir ) RNA, s i RNA , shRNA 및 miRNA로 이루어진 군에서 선택되는 핵산에 의해 수행되는 것일 수 있다. In CAR T cell therapy, after CAR T cells injected into a subject's body are used to selectively kill a tumor, and after the tumor is effectively reduced, the remaining residual CAR T cells show a side effect of killing normal cells. This is called CAR T immunodeficiency toxicity or normal T cell toxicity. The adapter molecule showed the effect of CAR T cell immunodeficiency toxicity or normal T cell toxicity evasion ability by regulating the activation of chimeric antigen receptor T cells. In one embodiment of the present invention, unlike existing CAR Ts that directly target existing tumors, the CAR T cell itself binds to the adapter molecule and binds to the tumor cell targeted by the adapter molecule, so that even when the CAR T cell was injected in the absence of the adapter molecule, it did not show toxicity to the target cell or tumor. That is, CAR T cells induce activation only in the presence of the adapter molecule, and thus show toxicity to the target cell or tumor. Therefore, the adapter molecule can induce activation or inactivation of CAR T cells. For this reason, the adapter molecule is also called a switchable molecule. Accordingly, the use of the adapter molecule to induce activation of chimeric antigen receptor T cells includes the use of preventing immunodeficiency by alleviating cytotoxicity of residual CAR T cells to normal cells and the resulting side effect of killing normal T cells after CAR T cell therapy. Another aspect of the present invention provides a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to a second moiety of an adaptor molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain. The "first moiety", the "second moiety" or the "adaptor molecule" are as described above. The term "extracellular domain" as used herein means a domain that protrudes outside a cell and binds to a ligand or the like. The term "intracellular domain" as used herein means a domain that is located inside the cell membrane of a cell, i.e., in the cytoplasm, and transmits a signal transmitted by binding between the extracellular domain and the ligand into the cell. In one specific example, the chimeric antigen receptor (hereinafter also referred to as 'CAR') refers to a protein that fuses a cell membrane or intracellular signaling site of a T-cell activation protein (CD3-zeta chain, CD28, 41BBL, 0x40, I COS, high-affinity receptor for IgE (FCERI) and other T-cell activation proteins) with an antigen-binding site (single chain Fv fragment) of a cancer antigen-specific antibody. Specifically, the first-generation CAR includes an extracellular domain including an antigen recognition site specifically expressed in cancer cells, a transmembrane domain and an intracellular signaling domain, and may use only CD3 as the signaling domain. The second-generation CAR has a structure that combines a co-stimulatory domain (CD28 or CD137/4-1BB) and CD3《 to enhance responsiveness to immune cells, and may increase the number of immune cells containing CAR remaining in the body compared to the first-generation CAR. The third-generation CAR may have a structure that combines a co-stimulatory domain with 4-1BB, CD28 or 0X40, etc., using two or more co-stimulatory domains to achieve expansion and persistence of immune cells containing CAR in the body. The fourth-generation CAR may include an additional gene encoding a cytokine, such as IL-12 or IL-15, to enable additional expression of a CAR-based immune protein of the cytokine, and the fifth-generation CAR may additionally include an interleukin receptor chain, for example, IL-2RP, to strengthen immune cells. In one specific example, the chimeric antigen receptor may have the structure of a first-generation CAR to a fifth-generation CAR. In one specific example, the extracellular connecting portion of the chimeric antigen receptor may further include a hinge domain. In one specific example, the hinge domain may be characterized by being composed of an oligopeptide or a polypeptide and including 1 to 100 amino acid residues, specifically, 10 to 70 amino acid residues, but is not limited thereto. In one specific example, the intracellular signaling domain of the chimeric antigen receptor may be a portion located inside the cell membrane of an immune cell, i.e., in the cytoplasm, and may refer to a portion that transmits a signal within the cell when the antigen binding domain included in the extracellular domain binds to a target antigen and activates an immune response of the immune cell. In one specific embodiment, the intracellular signaling domain may be, but is not limited to, one or more intracellular signaling domains selected from the group consisting of CD3 zeta (《), CD3 gamma (Y), CD3 delta (6), CD3 epsilon (E), FcR gamma, FcR beta, CD5, CD22, CD79a, CD79b and CD66d, and in one embodiment, the intracellular signaling domain of the chimeric antigen receptor is CD3 zeta (《). In one specific embodiment, the intracellular signaling domain may be characterized by further comprising, but is not limited to, a costimulatory domain. The co-stimulatory domain according to the present invention may include one or more co-stimulatory domains selected from the group consisting of ligands that specifically bind to CD2, CD7, CD27, CD28, CD30, CD40, 4-lBB (CD137), 0X40 (CD134), I COS, LFA-1, GITR, MyD88, DAP1, PD-1, LIGHT, NKG2C, B7-H3 and CD83, but is not limited thereto. In one specific embodiment, the chimeric antigen receptor may be characterized by comprising one or more intracellular signaling domains and one or more co-stimulatory domains. In one specific embodiment, the co-stimulatory domain may be 4-lBB. The reduction or elimination of CD3 expression of the chimeric antigen receptor T cells may be achieved by removing CD3, specifically endogenous CD3, of the T cells or CAR-T cells introduced with the CAR gene before introducing the CAR gene, or by removing the TCR. In other words, the reduction or elimination of CD3 expression may be achieved by directly removing the CD3 gene or indirectly reducing, suppressing, or eliminating CD3 expression by removing the TCR. Reducing or eliminating the expression of the CD3 or TCR may be performed by a genome editing technique including a CRISPR system, specifically, CRISPR/Cas9, TALEN, Zinc fi nger nuclease, base-editing and prime-editing, or by a nucleic acid selected from the group consisting of antisense RNA, antagomir RNA, si RNA, shRNA and miRNA. The CAR T cell may be one that binds to the second moiety of the adaptor molecule. More specifically, the CAR T cell may exhibit cytotoxicity against a target cell or a tumor cell by binding to the second moiety of the adaptor molecule. That is, the extracellular domain of the CAR T cell, more specifically, the CAR T cell may include an antibody or an antigen-binding fragment thereof having binding ability to the second moiety of the adaptor molecule. The second moiety may be a His tag. Therefore, the CAR T cell, more specifically, the extracellular domain of the CAR T cell may include a protein having a specific binding ability to His tag, specifically, an antibody or an antigen-binding fragment thereof. In one embodiment of the present invention, the antibody or antigen-binding fragment thereof that specifically binds to the His tag may include an amino acid sequence of SEQ ID NO: 63, a part thereof, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98% or 99% identity therewith. Even if the present application describes "comprising a gene sequence/amino acid sequence of a specific sequence number" or "having a gene sequence/amino acid sequence of a specific sequence number", it is obvious that a gene sequence/amino acid sequence having a part of the sequence deleted, modified, substituted or added may also be used in the present application if it has the same or corresponding function as that composed of the gene sequence/amino acid sequence of the corresponding sequence number. In addition, in the present application, the gene sequence and the base sequence may be used interchangeably. For example, if it has the same or corresponding function as the CAR T cell, it is obvious that it is within the scope of the present application that a meaningless sequence is added to the sequence or the end of the sequence number, or a part of the sequence or the end of the sequence number is deleted. Homology and identity refer to the degree of relationship between two given base sequences and can be expressed as a percentage. The terms homology and identity are often used interchangeably. Whether any two sequences have homology or identity can be determined by, for example, using the default parameters such as Pearson et al (1988) [Proc. Natl. Acad. Sci. USA 85]: 2444 It can be determined using known computer algorithms such as the "FASTA" program. Alternatively, the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443-453) as implemented in the Needleman program of the EMBOSS package (EMBOSS: The European Molecular Biology Open Software Suite, Rice et al. , 2000, Trends Genet . 16: 276— 277) (version 5.0.0 or later) can be determined using the GCG program package (Devereux, J. , et al. , Nucleic Acids Research 12: 387 (1984)), BLASTP, BLASTN, FASTA (Atschul, [S.] [F. ,] [ET AL, J MOLEC BIOL 215]: 403 (1990): Guide to Huge Computers , Martin J . Bishop, [ ED . , ] Academic Press, San Diego, 1994, and [CARILLO ETA/.] (1988) SIAM J Applied Math 48: 1073). For example, sequence homology or identity can be determined using BLAST of the National Center for Biotechnology Information Database, or ClustalW. Another aspect of the present invention provides a complex comprising a chimeric antigen receptor T cell in which the expression of CD3 is reduced or deleted and an adapter molecule that regulates the activity of the CAR-T cell. Another aspect of the present invention provides a kit for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or deleted and an adapter molecule that regulates the activity of the CAR-T cell. The "CAR T cell", the "first moiety", the "second moiety" or the "adapter molecule" are as described above. Another aspect of the present invention provides a composition comprising a chimeric antigen receptor T cell (CAR-T cell) in which the expression of CD3 is reduced or eliminated and an adapter molecule that regulates the activity of the CAR-T cell. The composition may be a pharmaceutical composition. The composition may be a pharmaceutical composition for preventing or treating a T cell-derived tumor. Another aspect of the present invention provides a pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell in which the expression of CD3 is reduced or eliminated and is administered in combination with an adapter molecule that regulates the activity of the CAR-T cell. The "CAR T cell", the "first moiety", the "second moiety" or the "adapter molecule" As mentioned above. As used herein, the term "combination therapy" or "combined treatment" or "in combination" refers to any form of simultaneous or concurrent treatment using at least two separate therapeutic agents. The components of the combination therapy may be administered simultaneously, sequentially, in reverse order, or in any order. The components may be administered in different dosages, at different administration frequencies, or via different routes in an appropriate manner. As used herein, the term "administration" means introducing a given substance into a subject in an appropriate manner, and the "subject" means any living organism, including humans, such as rats, mice, and livestock, that may have a metabolic disease or adverse effects of a metabolic regulator. As a specific example, it may be a mammal, including humans. The term "administered simultaneously" as used herein is not particularly limited, and means that the components of the combination therapy are administered substantially simultaneously, for example, as a mixture or in an immediately subsequent order. The term "administered sequentially" as used herein is not particularly limited, and means that the components of the combination therapy are not administered simultaneously, but rather between administrations. Means administered one after another or in clusters with a specific time interval. The time interval may be the same or different between the administration of each of the components of the combination therapy and can be selected, for example, in the range of 2 minutes to 96 hours, 1 day to 7 days or 1 week, 2 weeks or 3 weeks. Typically, the time interval between administrations can be in the range of several minutes to several hours, for example, in the range of 2 minutes to 72 hours, 30 minutes to 24 hours or 1 to 12 hours. Further examples include time intervals in the range of 24 to 96 hours, 12 to 36 hours, 8 to 24 hours and 6 to 12 hours. The term "disease" as used herein can mean a pathological condition, particularly cancer, an infectious disease, an inflammatory disease, a degenerative disease, a disease involving apoptosis and graft rejection. The term "treatment" as used herein refers to or includes the alleviation, inhibition of progression, or prevention of a disease, disorder or condition, or one or more symptoms thereof, and "active ingredient" or "pharmaceutically effective amount" can mean any amount of a composition used in the course of practicing the invention provided herein sufficient to alleviate, inhibit progression, or prevent a disease, disorder or condition, or one or more symptoms thereof. The terms "administering,""introducing," and "implanting" as used herein are used interchangeably and refer to the placement of a composition according to one embodiment into a subject by a method or route that results in at least partial localization of the composition to a desired site according to one embodiment. can mean. The composition according to one embodiment can be administered by any suitable route that delivers the cells or at least a portion of the cellular components to a desired location within a viable organism. The survival period of the cells after administration to the organism can be as short as several hours, for example 24 hours, to several days, or as long as several years. The above T-cell derived tumors include T-cell prolymphocytic leukemia, T-cell large granular lymphocytic leukemia, chronic lymphoprol i ferat ive disorder of NK cells, aggressive NK leukemia, systemic EBV-positive T-cell 1 lymphoprol i ferat ive disease of chi Idhood, hydroa vaccini forme- 1 ike lymphoma, adult T-cell leukemia/lymphoma, extranodal NK/T-cell 1 lympho (nasal type), Enteropathy-associated T-cel 1 lymphoma, hepatosplenic T-cel 1 lymphoma, subcutaneous panniculitis-like T-cel 1 lymphoma, mycosis fungoides, Se'zary syndrome, primary cutaneous CD30-positive T-cel 1 lymphoproliferative disorders, lymphomatoid papulosis, primary cutaneous anaplastic large cel 1 lymphoma, primary cutaneous gamma/delta T-cel 1 lymphoma, primary cutaneous CDS-positive aggressive epidermotropic cytotoxic It may be selected from the group consisting of primary cutaneous CD8-positive aggressive epidermotropic cytotoxic T-cel 1 lymphoma, primary cutaneous CD4-positive small 1/medium T-cel 1 lymphoma, peripheral T-cel 1 lymphoma NOS (not otherwise specified), angioimmunoblastic T-cel 1 lymphoma, ALK positive anaplastic large cel 1 lymphoma, and ALK negative anaplastic large cel 1 lymphoma. The administration may be in combination with an additional anticancer agent. Additional examples of anticancer agents may include alkylating agents, antimetabolites, spindle inhibitor plant alkaloids, cytotoxic/antitumor antibiotics, topoisomerase inhibitors, antibodies, photosensitizers, and kinase inhibitors. Examples of such anticancer agents may include compounds used in targeted therapy and conventional chemotherapy. In addition, the antibodies Examples include alemtuzumab, apolizumab, acelizumab, atlizumab, bapineuzumab, bevacizumab, vivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, Pascolizumab, pekfucituzumab, pectuzumab, pertuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resaivizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadoxizumab, talizumab, tefibazumab, tocilizumab, toralizumab, trastuzumab, tucotuzumab selmoleukin, tucusituzumab, umavizumab, urtoxazumab, and visilizumab may be included. The composition according to the present invention may additionally comprise a pharmaceutically acceptable carrier. In the case of injections, buffers, preservatives, analgesics, solubilizers, isotonic agents, stabilizers, etc. can be mixed and used, and in the case of local administration, bases, excipients, lubricants, preservatives, etc. can be used. The formulation of the pharmaceutical composition of the present invention can be variously prepared by mixing it with the pharmaceutically acceptable carrier as described above. For example, in the case of injections, it can be prepared in the form of a unit dosage ampoule or a multiple dosage form. In addition, the anticancer composition may typically include a surfactant that facilitates movement through a membrane. Such surfactants include those derived from steroids, cationic lipids such as N-[l-(2,3-dioleoyl)propyl-N,N,N-trimethylammonium chloride (DOTMA), or various compounds such as cholesterol hemisuccinate and phosphatidyl glycerol. In one specific example, the composition can be administered in a pharmaceutically effective amount to treat cancer cells or their metastasis, or to inhibit the growth of cancer. It may vary depending on various factors such as cancer type, patient's age, weight, nature and degree of symptoms, type of current treatment, number of treatments, dosage form and route, and can be easily determined by a person skilled in the art. The composition according to the present invention can be administered together with the above-mentioned pharmacological or physiological components or administered sequentially, and can also be administered in combination with an additional conventional therapeutic agent, and can be administered sequentially or simultaneously with the conventional therapeutic agent. Such administration may be single or multiple administration. It is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects considering all of the above factors, and this can be easily determined by a person skilled in the art. Another aspect of the present invention provides a method for preventing or treating a T cell-derived tumor, comprising administering to a subject the complex or pharmaceutical composition. Another aspect of the present invention provides a method for producing a chimeric antigen receptor T cell (CAR-T cell) having reduced or eliminated expression of CD3, comprising the steps of (a) down-regulating the expression of CD3 or a T cell receptor (TCR) in a T cell, and (b) introducing a chimeric antigen receptor (CAR) comprising an extracellular domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular domain comprises an antibody or an antigen-binding fragment thereof that specifically binds to a second moiety of an adaptor molecule, wherein steps (a) and (b) are performed in any order. The method for reducing or eliminating the expression of the CD3 or TCR may be performed by a genome editing technique including a CRISPR system, specifically, CRISPR/Cas9, TALEN, zinc finger nuclease, base editing and prime editing, or by a nucleic acid selected from the group consisting of antisense RNA, antagomir RNA, si RNA, shRNA and miRNA.

【발명의 효과】 본 발명의 일 양상에 따른 항- CD3 항원결합단편을 포함하는 어댑터 분자 및 이와 결합하는 CD3-키메라항원 수용체를 발현하는 T세포에 의하면, 동족살상방지능 및 면역결핍독성 회피능을 나타내어, T세포 유래 종양 치료제에 유용하게 사용될 수 있다. 【Effect of the invention】 According to one aspect of the present invention, an adapter molecule comprising an anti-CD3 antigen-binding fragment and a T cell expressing a CD3-chimeric antigen receptor binding thereto exhibit anti-fratricide activity and immunodeficiency toxicity avoidance activity, and thus can be usefully used in a T cell-derived tumor treatment agent.

【도면의 간단한설명】 도 1은 인간 CD3 유전자편집용 가이드오때를 1차스크리닝한 결과를 나타낸 그래프이다. 도 2는 인간 CD3 유전자편집용 가이드오때를 2차스크리닝한 결과를 나타낸 그래프이다. 도 3은 CD3 유전자편집한 인간 T세포에서의 표현형을분석한그래프이다. 도 4는 항 CD3 어댑터 항체를 표지할 케미컬 및 펩타이드 태그 도입한 CAR T 세포의 표현형을분석한 그래프이다 (Cot: cotinine, His: 6XHis). 도 5는 His 태그를 도입한 인간 CAR T세포에서의 6xHis-CAR 발현율을 분석한 그래프이다. 도 6은 항 CD3 어댑터 항체 scFv의 구조의 나타낸 모식도이다. 도 7은 0KT3- Ck- His scFv 및 0KT3- Ck-링커- His 유內의 융합단백질을 정제후 SDS- PAGE한 결과를 나타낸 이미지이다. 도 8은 항 CD3 어댑터 항체 Fab의 구조의 나타낸 모식도이다. 도 9는 1-4-2-Ck-His scFv/l-4-7-Ck-His scFv 및 1- 4- 2- Fab- Hi s/1- 4- 7- Fab- His 융합단백질을 정제후 SDS-PAGE 결과를 나타낸 이미지이다. 도 10은 0KT3-Ck-His, 0KT3- Ck- L- His 및 UCHT1- Fab- His의 T세포 종양 세포주 Jurkat와의 결합능을분석한 그래프이다. 도 11은 1- 4- 2- Ck- His/1- 4- 7- Ck- His및 1-4- 2- Fab- Hi s/1- 4- 7- Fab- His의 T세포 종양 세포주 Jurkat와의 결합능을분석한그래프이다. 도 12는 0KT3- Ck- His scFv와 인간 항 6xHis CAR T세포의 항종양 활성 및 종양살상능을 분석한 그래프이다. 도 13은 0KT3-His Fab와 인간 항 6xHis CAR T세포의 종양살상능을 분석한 그래프이다. 도 14는 UCHTl-His Fab와 인간 항 6xHis CAR T세포의 항종양 활성 및 종양살상능을 분석한 그래프이다. 도 15는 1-4- 2- His Fab 및 1-4- 7- His Fab와 인간항 6xHis CAR T세포의 항종양 활성 및 종양살상능을분석한그래프이다. 도 16은 인간 항 6xHis CAR T세포의 동족살상능을 나타낸 그래프이다. 도 17은 인간항 6xHis CAR T세포에서의 CD3유전자편집 후 T세포의 표현형을 분석한 그래프이다. 도 18은항 CD3어댑터 항체를통해 인간접합형 CAR T세포동족살상방지능을 확인한 그래프이다. 도 19는 CD3 유전자편집 접합형 CAR T세포 증식조건을 나타낸 모식도이다. 도 20은 1차및 2차 CD3유전자편집 접합형 CAR T세포증식 단계에서 T세포의 표현형을 분석한 그래프이다. 도 21은 CD3 유전자 편집 접합형 CAR T세포 증식 단계에서 측정된 CAR T 세포의 수를 나타낸 그래프이다. 도 22는 마우스 모델에서 0KT3- Ck- Hi s scFv와 CD3- 인간 Hi s CAR T세포의 항종양능 분석 실험의 모식도이다. 도 23은 마우스 모델에서 0KT3- Ck- Hi s scFv와 CD3- 인간 Hi s CAR T세포의 항종양 활성 및 마우스 모델의 생존률을 분석한 그래프이다. 도 24는 마우스 모델에서 UCHT1- Fab- Hi s와 CD3 -유전자편집 인간 Hi s CAR T세포의 항종양능 분석 실험의 모식도이다. 도 25는 마우스 모델에서 UCHT1- Fab- Hi s와 CD3 -유전자편집 인간 Hi s CAR T세포의 항종양 활성 및 마우스 모델의 생존률을 분석한 그래프이다. 도 26은 인간 접합형 CAR T세포의 면역결핍 방지능을 분석한 그래프이다. 도 27은 인간화 항체 항 Hi s 유內의 항원 친화도를 나타낸 것이다. 도 28은 huHi s CAR- T세포 표면의 CAR발현을 유세포분석으로 확인한 결과이다. 도 29는 제작된 huHi s CAR- T세포와 UCHTl-Fab-Hi s 어댑터를 사용하여 접합형 CAR T세포의 in vitro 활성정도를 검증한 결과이다 . 【Brief description of the drawings】 Fig. 1 is a graph showing the results of the first screening of guide genes for human CD3 gene editing. Fig. 2 is a graph showing the results of the second screening of guide genes for human CD3 gene editing. Fig. 3 is a graph showing the analysis of the phenotype of CD3 gene-edited human T cells. Fig. 4 is a graph showing the analysis of the phenotype of CAR T cells introduced with chemical and peptide tags for labeling anti-CD3 adapter antibodies (Cot: cotinine, His: 6XHis). Figure 5 is a graph analyzing the expression rate of 6xHis-CAR in human CAR T cells introduced with His tag. Figure 6 is a schematic diagram showing the structure of anti-CD3 adaptor antibody scFv. Figure 7 is an image showing the result of SDS-PAGE after purification of fusion proteins containing 0KT3-Ck-His scFv and 0KT3-Ck-linker-His. Figure 8 is a schematic diagram showing the structure of anti-CD3 adaptor antibody Fab. Figure 9 is an image showing the result of SDS-PAGE after purification of 1-4-2-Ck-His scFv/l-4-7-Ck-His scFv and 1-4-2-Fab-His/1-4-7-Fab-His fusion proteins. Figure 10 is a graph analyzing the binding ability of 0KT3-Ck-His, 0KT3- Ck- L- His, and UCHT1- Fab- His to the T cell tumor cell line Jurkat. Figure 11 is a graph analyzing the binding ability of 1- 4- 2- Ck- His/1- 4- 7- Ck- His and 1- 4- 2- Fab- Hi s/1- 4- 7- Fab- His to the T cell tumor cell line Jurkat. Figure 12 is a graph analyzing the antitumor activity and tumor killing ability of 0KT3- Ck- His scFv and human anti-6xHis CAR T cells. Figure 13 is a graph analyzing the tumor killing ability of 0KT3-His Fab and human anti-6xHis CAR T cells. Fig. 14 is a graph analyzing the antitumor activity and tumor killing capacity of UCHT1-His Fab and human anti-6xHis CAR T cells. Fig. 15 is a graph analyzing the antitumor activity and tumor killing capacity of 1-4-2-His Fab and 1-4-7-His Fab and human anti-6xHis CAR T cells. Fig. 16 is a graph showing the fratricidal activity of human anti-6xHis CAR T cells. Fig. 17 is a graph analyzing the phenotype of T cells after CD3 gene editing in human anti-6xHis CAR T cells. Fig. 18 is a graph confirming the fratricidal prevention capacity of human conjugated CAR T cells through anti-CD3 adapter antibody. Fig. 19 is a schematic diagram showing the proliferation conditions of CD3 gene-edited conjugated CAR T cells. Figure 20 is a graph analyzing the phenotype of T cells in the first and second CD3 gene-edited conjugated CAR T cell proliferation stages. Fig. 21 is a graph showing the number of CAR T cells measured at the CD3 gene-edited conjugated CAR T cell proliferation stage. Fig. 22 is a schematic diagram of an anti-tumor activity assay experiment of 0KT3- Ck- Hi s scFv and CD3- human Hi s CAR T cells in a mouse model. Fig. 23 is a graph analyzing the anti-tumor activity and the survival rate of a mouse model of 0KT3- Ck- Hi s scFv and CD3- human Hi s CAR T cells in a mouse model. Fig. 24 is a schematic diagram of an anti-tumor activity assay experiment of UCHT1- Fab- Hi s and CD3 -gene-edited human Hi s CAR T cells in a mouse model. Fig. 25 is a graph analyzing the anti-tumor activity and the survival rate of a mouse model of UCHT1- Fab- Hi s and CD3 -gene-edited human Hi s CAR T cells in a mouse model. Figure 26 is a graph analyzing the immunodeficiency prevention ability of human conjugated CAR T cells. Figure 27 shows the antigen affinity of humanized antibody anti-His. Figure 28 shows the results of confirming CAR expression on the surface of huHis CAR- T cells by flow cytometry. Figure 29 shows the results of verifying the in vitro activity level of conjugated CAR T cells using the produced huHis CAR- T cells and the UCHTl-Fab-His adapter.

【발명을 실시하기 위한 형태】 이하, 본 발명의 이해를 돕기 위하여 바람직한 실시 예를 제시한다. 그러나 하기의 실시 예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐 , 하기 실시 예에 의해 본 발명의 내용이 한정되는 것은 아니다. 실시 예들은 다양한 변환을 가할 수 있는 바, 실시 예들은 이하에서 개시되는 실시 예들에 한정되는 것이 아니라 다양한 형태로 구현될 수 있다. 실시예 1 . 인간 접합형 CD3 편집 CAR-T 세포의 제작 【Form for carrying out the invention】 Hereinafter, preferred embodiments are presented to help understanding of the present invention. However, the following embodiments are provided only to more easily understand the present invention, and the content of the present invention is not limited by the following embodiments. The embodiments can be modified in various ways, and the embodiments are not limited to the embodiments disclosed below, but can be implemented in various forms. Example 1. Production of human conjugated CD3 edited CAR-T cells

CAR-T 세포가 종양 표면항원 CD3를 인식하여 선택적으로 종양을 살상하기 위해 , (D CAR-T 세포 자체는 CD3를 발현하지 않게 함으로써 CAR-T 세포간 동족살상을 회피하고 , ② 이렇게 CD3가 제거된 CAR-T 세포가 CD3를 발현하는 정상 T세포를 살상함으로써 야기되는 환자의 면역결핍부작용을 최소화하도록 접합형 ( conj unct ive) CAR-T 세포를 제작하고자 하였다. 이 개념의 모식도는 아래와 같다. In order for CAR-T cells to recognize the tumor surface antigen CD3 and selectively kill tumors, (1) conjugated CAR-T cells were created to avoid fratricide between CAR-T cells by making the D CAR-T cells themselves not express CD3, and (2) minimize the immunodeficiency side effect in patients caused by CAR-T cells from which CD3 has been removed killing normal T cells that express CD3. A schematic diagram of this concept is as follows.

Figure imgf000020_0001
Figure imgf000020_0001

[접합형 CD3표적 CAR-T세포] 실험 재료 [Conjugated CD3-targeted CAR-T cells] Experimental materials

(1) 마우스 (1) Mouse

C57BL/6CB6) 마우스는 오리엔트 바이오에서 구매하였다. NSG 마우스 (NOD. Cg- Prkdcscid 112rgtmlWj 1/SzJ)는 Jackson Laboratory로부터 구매하였다. 모든마우스는 서울대학교 의과대학 (대한민국, 서울시)의 특정 무병원체 (SPF) 동물 시설에 수용되었고 기관 동물 관리 및 사용 위원회 (IACUC)의 지침에 따라유지되었다. C57BL/6CB6) mice were purchased from Orient Bio. NSG mice (NOD. Cg- Prkdcscid 112rgtmlWj 1/SzJ) were purchased from Jackson Laboratory. All mice were housed in a specific pathogen-free (SPF) animal facility at Seoul National University College of Medicine (Seoul, Republic of Korea) and maintained according to the guidelines of the Institutional Animal Care and Use Committee (IACUC).

(2) 세포주 (2) Cell line

Jurkat (사람 T세포림프종)은 ATCC(ATCC, Manassas , VA USA)에서 구매하였다. Phoenix GP오} Phoenix Eco 서]포주는 Garry Nolan (Stanford University, 미국)으로부터 제공받았다. GFP-루시퍼라제 발현 세포주 (Jurkat- Luc)는 GFP-Luc 발현용 렌티바이러스 (pLEF- Luc- GFP)를 Jurkat 세포에 형질도입 (transduct ion)한 후 세포 분류기 (cell sorter , FACS Aria, Becton Dickinson)를 이용하여 GFP양성 세포를 분리 후, 제한적 희석법 (limiting dilution)을통해 단일세포주를확립하였다. CD3가 결핍된 Jurkat 세포주는 CRISPR-Cas9 시스템을 이용하여 세포주가 발현하는 CD3 유전자에 in-del 돌연변이를 도입 후 , 세포 분류기를 이용하여 CD3 음성세포를 분리하고 , 이어서 제한적 희석법을 통해 단일 세포주를 확립하였다. Jurkat (human T-cell lymphoma) was purchased from ATCC (Manassas, VA USA). Phoenix GP was provided by Garry Nolan (Stanford University, USA). GFP-luciferase expressing cell line (Jurkat-Luc) was transduced into Jurkat cells with lentivirus for GFP-Luc expression (pLEF-Luc-GFP), and GFP-positive cells were isolated using a cell sorter (FACS Aria, Becton Dickinson), and then a single cell line was established through limiting dilution. CD3 The deficient Jurkat cell line was created by introducing an in-del mutation into the CD3 gene expressed by the cell line using the CRISPR-Cas9 system, then isolating CD3-negative cells using a cell sorter, and then establishing a single cell line through the limiting dilution method.

1-1. CAR발현 레트로바이러스 및 렌티바이러스 제작 접합형 CAR- T세포에서 사용된 scFv의 경우 , Cot CAR scFv는 토끼 유래의 항 cot inine 항체 (Cl in Chim Acta 2010; 411: 1238- 1242)에서 기원하였으며 , Hi s CAR의 scFv는 마우스 유래의 항 Hi s 항체 (mutated 3D5 clone; J . Mol . Biol . (2002) 318 , 135-147)에서 기원하였으며 , Myc CAR의 scFv는 마우스 유래의 항 Myc 항체 (9E10 clone; FEES Letters 414 (1997) 33- 38)에서 기원하였다. 마우스 CAR 단백질 코딩부위 (protein coding region)는 마우스 면역글로불린 kappa chain의 신호펩티드 (signal pept ide) , 항체의 scFv, 기존 보고된 CAR backbone부위 (마우스 CD28 세포외부위-세포막투과부위-세포내부위 , 마우스 CD3제타의 세포내부위 ; GenBank HM754222.1 , Blood 2010 : 116(20): 4099- 4102)로 구성됨 . CAR cDNA들은 한국 미국 IDT ( Integrated DNA technologies)사에 DNA합성을 의뢰하여 제작하였다. CAR 발현용 마우스 CAR-T 세포 제작용 레트로바이러스 벡터는 pMSCV- puro(Ckmtech , 미국) 레트로바이러스 벡터에서 PGK프로모터 하위의 PuroR유전자 부위를 상기 제작된 CAR cDNA로 치환하여 클로닝하여 제작하였다. 인간 CAR 단백질 코딩부위는 인간 GMCSF의 신호펩티드 (signal pept ide) , 항 Hi s 항체의 scFv, 인간 CD28 세포외부위- 세포막투과부위-세포내부위 , 인간 CD3제타의 세포내부위로 구성됨 . 단백질 코딩부위 cDNA는 미국 IDT사에 DNA합성을 의뢰하여 제작하였다. 인간 CAR- T세포 발현용 레트로바이러스 벡터는 pMSGV(Addgene Plasmid #64269)를 이용하여 인간 CAR cDNA를 사용하여 cloning을 진행하였으며 . 인간 CAR- T세포 발현용 렌티바이러스 벡터는 일부 변형된 pCDH- EF1벡터 (Addgene Plasmid #72266)에 인간 CAR cDNA를 클로닝하여 제작하였다. 1-1. Production of CAR-expressing retrovirus and lentivirus In the case of scFv used in conjugated CAR- T cells, Cot CAR scFv was derived from rabbit-derived anti-cot inine antibody (Cl in Chim Acta 2010; 411:1238-1242), Hi s CAR scFv was derived from mouse-derived anti-Hi s antibody (mutated 3D5 clone; J . Mol . Biol . (2002) 318 , 135-147), and Myc CAR scFv was derived from mouse-derived anti-Myc antibody (9E10 clone; FEES Letters 414 (1997) 33-38). The mouse CAR protein coding region consists of the signal peptide of the mouse immunoglobulin kappa chain, the antibody scFv, and the previously reported CAR backbone region (mouse CD28 extracellular region-transmitter region-intracellular region, mouse CD3 zeta intracellular region; GenBank HM754222.1, Blood 2010: 116(20):4099-4102). CAR cDNAs were produced by requesting DNA synthesis to Integrated DNA technologies (IDT) of Korea, USA. The retroviral vector for producing mouse CAR-T cells for CAR expression was produced by cloning the PuroR gene region downstream of the PGK promoter in the pMSCV-puro (Ckmtech, USA) retroviral vector and replacing it with the produced CAR cDNA. The human CAR protein coding region consists of the signal peptide of human GMCSF, the scFv of anti-His antibody, the extracellular region-transmitter region-intracellular region of human CD28, and the intracellular region of human CD3 zeta. The protein coding region cDNA was produced by requesting DNA synthesis to IDT, USA. The retroviral vector for human CAR- T cell expression was cloning using human CAR cDNA using pMSGV (Addgene Plasmid #64269). The lentiviral vector for human CAR- T cell expression was produced by cloning human CAR cDNA into a partially modified pCDH-EF1 vector (Addgene Plasmid #72266).

1-2. CAR 발현용 레트로바이러스와 렌티바이러스의 생산 마우스 T세포에 형질도입을 위한 ecotrophi c 레트로바이러스를 얻기 위해서 , CAR construct는 바이러스 외피 단백질로서 수포성 구내염 인디애나 바이러스 G단백질 (VSV- G)를 인코딩하는 cDNA를 포함하는 pMD2.G 플라스미드와 함께 Phoenix GP 세포주로 리포펙타민 3000( Invi trogen)을 이용하여 형질전환 ( transfect ion)하였다. 48시간 후 , VSV-G 위형 레트로바이러스 (pseudotyped retrovi rus)가 존재하는 상증액을 수확하여 Phenix Eco 세포주에 형질도입 (transduction)하였다. 3〜 5일 후, Cot CAR의 경우 항토끼 IgG( Jackson ImmunoResearch, West Grove, PA USA) , His CAR & Myc CAR의 경우 항마우스 I gG( Jackson ImmunoResearch, West Grove, PA USA)로 염색한 후 CAR 고발현 Phoenix Eco 세포를 FACS Ar ia(Becton Dickinson)로 분리 (sorting)하여 레트로바이러스 생산 세포주를 확립하였다. 이 세포주로부터 생산된 레트로바이러스 배양 상층액을 원심분리필터장치 (Amicon Ultra-100 kDa cutoff, Millipore, USA)를사용하여 5〜 10배 농축하여 사용하였다. 사람 T세포에 형질도입을 위한 양쪽지향성 (amphotropic) 레트로바이러스를 얻기 위해서, 바이러스 생산용 PG13 세포주를 사용하였다. 각 레트로바이러스 플라스미드를 리포펙타민 3000(Lipofectamin 3000)(Invitrogen)을 이용하여 Phoenix Eco 세포주에 형질전환을 수행하였다. 48시간 후, 레트로바이러스가 분비된 배양 상층액을 수확하여 PG13 세포주에 가하여 형질도입 (transduct ion)을 진행하였다. 이후 레트로바이러스 생산 세포주 확립과 바이러스 생산은 마우스 세포를 위한 레트로바이러스의 경우와동일한 방식으로 진행하였다. 사람 T세포에 형질도입을위한 렌티바이러스를 얻기 위해서 , 각 렌티바이러스 플라스미드를 리포펙타민 3000 (Invitrogen)을 이용하여 packaging DNA 3종 (pMD.2G, pMDLg/ pRRE , pRSV- rev)과 함께 293T cel 1 1 ine (ATCC)에 형질전환 (transfect ion)한 후, 24, 48시간동안 분비된 lentivirus가 포함된 배양상층액을 수확 및 필터 (0.45仰 1 필터)를 이용한 세포 잔존입자를 제거하고, 초고속원심분리기를 사용하여 100배 농축한 후 CAR T세포 제작을 위한 렌티바이러스 농축액으로사용하였다 1-2. Production of retrovirus and lentivirus for CAR expression To obtain ecotrophic retrovirus for transduction of mouse T cells, the CAR construct was transfected into Phoenix GP cells using Lipofectamine 3000 (Invitrogen) together with the pMD2.G plasmid containing cDNA encoding vesicular stomatitis Indiana virus G protein (VSV-G) as a viral envelope protein. After 48 hours, VSV-G pseudotyped retrovirus was present. The supernatant was harvested and transduced into the Phenix Eco cell line. After 3–5 days, for Cot CAR, the cells were stained with anti-rabbit IgG (Jackson ImmunoResearch, West Grove, PA USA) and for His CAR & Myc CAR, anti-mouse I gG (Jackson ImmunoResearch, West Grove, PA USA). Then, CAR-high CAR-expressing Phoenix Eco cells were separated by FACS Ar ia (Becton Dickinson) to establish a retrovirus-producing cell line. The retrovirus culture supernatant produced from this cell line was concentrated 5–10 times using a centrifugal filter device (Amicon Ultra-100 kDa cutoff, Millipore, USA) and used. To obtain amphotropic retrovirus for transduction of human T cells, the PG13 cell line for virus production was used. Each retroviral plasmid was transformed into Phoenix Eco cells using Lipofectamine 3000 (Invitrogen). After 48 hours, the culture supernatant containing retrovirus was harvested and added to PG13 cells for transduction. Thereafter, the establishment of retrovirus production cell lines and virus production were performed in the same manner as for retrovirus for mouse cells. To obtain lentivirus for transduction of human T cells, each lentivirus plasmid was transfected into 293T cell 1 1 ine (ATCC) together with three types of packaging DNA (pMD.2G, pMDLg/pRRE, pRSV-rev) using Lipofectamine 3000 (Invitrogen). After 24 and 48 hours, the culture supernatant containing secreted lentivirus was harvested and filtered (0.45 × 1 filter) to remove cell residual particles. After 100-fold concentration using an ultracentrifuge, it was used as a lentivirus concentrate for producing CAR T cells.

1.3 CRISPR/Cas9시스템을 이용한 인간 CD3편집 T세포제작 1.3 Production of human CD3-edited T cells using the CRISPR/Cas9 system

CD3 유전자편집을 위한 방법으로 CRISPR/Cas9 시스템을 사용하였다. 이 시스템을 효율적으로 사용하기 위하여 Cas9 cDNA가 탑재된 벡터 (PX458)에 인간 CD3 유전자 표적 가이드 RNA (sgRNAl〜 18)의 cDNA를 클로닝하였다. CD3 양성세포인 Jurkat세포에 제작된 플라스미드를 전기천공법 (electroporat ion)을 통해 형질전환 (transfect ion)하여 CD3 유전자편집을 진행하였다. 인간 일차 T세포에서의 CD3 유전자편집을 위해서는, 정상인으로부터 백혈구성분채혈 (leukapheresis)을통해 얻어진 백혈구를항 CD3항체 (0KT3 , 10//g/mt, BioXcell)가 코팅된 24웰 플레이트에 항 CD28항체 (CD28.2, 2//g/mt, BD Biosciences)와 함께 가한후 48시간동안 배양하여 T세포를 활성화시킴. 이후, Cas9 단백질 (Cas9 protein, Thermo)과 표적 가이드 RNA의 복합체인 리보핵단백질 (Ribonulceoprotein, RNP)를 제작하여 전기천공법을 통한 형질전환 방법으로 T세포에 유전자편집을진행하였다. 유전자편집 T세포는항생제가없고 hlL- 2(200U/ml)가 존재하는 배양액에서 24시간 배양 후, 다시 항생제와 hIL- 2가 있는 배양액으로 옮겨 T세포 배양을 진행하였다. CD3 유전자편집을 통한 CD3 단백질 제거 정도는 유전자편집 36시간 이후 시점부터 CD3 단백질 발현 정도혹은 T세포수용체의 발현정도를 유세포분석 (flow cytometry) (FACS-Canto II , BD Biosciences)을 통해 확인하였다. The CRISPR/Cas9 system was used as a method for CD3 gene editing. In order to use this system efficiently, the cDNA of the human CD3 gene targeting guide RNA (sgRNAl~ 18) was cloned into the vector (PX458) containing the Cas9 cDNA. The CD3 gene editing was performed by transfecting Jurkat cells, which are CD3-positive cells, with the constructed plasmid through electroporation. For CD3 gene editing in human primary T cells, white blood cells obtained from normal individuals through leukapheresis were added together with anti-CD3 antibody (0KT3, 10//g/mt, BioXcell) to a 24-well plate coated with anti-CD28 antibody (CD28.2, 2//g/mt, BD Biosciences), and then cultured for 48 hours to activate T cells. Afterwards, a complex of Cas9 protein (Cas9 protein, Thermo) and target guide RNA Ribonucleoprotein (RNP) was produced and gene-edited into T cells using electroporation. The gene-edited T cells were cultured in a culture medium without antibiotics but with hIL-2 (200 U/ml) for 24 hours, and then transferred to a culture medium with antibiotics and hIL-2 to culture the T cells. The degree of CD3 protein removal through CD3 gene editing was confirmed 36 hours after gene editing by flow cytometry (FACS-Canto II, BD Biosciences) to determine the degree of CD3 protein expression or T cell receptor expression.

CRISPR-Cas9 시스템을 이용한 인간 CD3 제거를 위하여 인간 CD3 ORF 표적부위에 대한 가이드 RNA서열 18종을 디자인한 후, 인간의 급성 T 세포 백혈병 세포주인 Jurkat에서 가이드 오때와 Cas9 뉴클레아제를 동시에 발현하는 플라스미드 日시요를 제작하고, 전기천공법을 이용하여 유전자를 도입하며, CD3 제거 여부를 검증하는 1차스크리닝을 진행하였고, 그 결과를 도 1에 나타내었다. To remove human CD3 using the CRISPR-Cas9 system, 18 guide RNA sequences for the human CD3 ORF target region were designed. Then, a plasmid that simultaneously expresses the guide ORF and Cas9 nuclease in the human acute T cell leukemia cell line Jurkat was constructed, and the gene was introduced using electroporation. A primary screening was performed to verify whether CD3 was removed. The results are shown in Fig. 1.

1차스크리닝에서 효율이 좋은가이드 RNA서열 4종을 선정하여 T세포유전자 전달효율이 좋은 mRNA 형질전환 (transfect ion)을 사용하여 2차 스크리닝을 진행하였다. 이를 위해 sgRNA (가이드 RNA + 스캐폴드 RNA)와 Cas9 mRNA를 in vitro 전人} (transcription) 반응을 통해 새롭게 제작한 후, mRNA 전기천공법을 이용하여 Jurkat에 CD3제거 여부를검증하는실험을 진행하였고, 그결과를 도 2에 나타내었다. 또한, 하기 표 및 표 2에 각각 sgRNA7 및 sgRNA15의 서열을 나타내었다. In the first screening, four guide RNA sequences with good efficiency were selected, and a second screening was conducted using mRNA transfection with good T cell gene transfer efficiency. To this end, sgRNA (guide RNA + scaffold RNA) and Cas9 mRNA were newly produced through an in vitro transcription reaction, and then an experiment was conducted to verify whether CD3 was removed in Jurkat using mRNA electroporation, and the results are shown in Fig. 2. In addition, the sequences of sgRNA7 and sgRNA15 are shown in the following Table and Table 2, respectively.

【표 11

Figure imgf000023_0001
【Table 11
Figure imgf000023_0001

【표 2]

Figure imgf000023_0002
도 1 및 도 2에 나타낸바와 같이 , 플라스미드 DNA transfection에 비해 높은 제거 (deletion) 효율을 얻을 수 있었으며 , 특히 2종의 sgRNA(sgRNA7 와 sgRNA15)의 경우 80% 이상의 높은 제거 효율이 관찰되었다. 이어서 제거 효율이 좋은 sgRNA7에 대하여 보다 간편하면서도 유전자편집효율이 좋은 것으로 알려진 RNP(r ibonucleoprotein) 전기천공법 방식을 통해 인간 T세포에서의 CD3제거를 시도하였으며 , 그 결과를 도 3에 나타내었다. 도 3에 나타낸 바와 같이, 재조합 Cas9단백질 및 sgRNA7의 복합체를 in
Figure imgf000024_0001
RNP형태로제작하여 활성화된 인간 T세포에 전기천공법을통해 CD3제거를 70% 이상의 효율로 획득하였다. 이상과 같이 , CRISPR/Cas9시스템을이용한인간 T세포에서의 CD3유전자편집 방법을 효과적으로 확립하였다. 【Table 2]
Figure imgf000023_0002
As shown in Figures 1 and 2, a high deletion efficiency was obtained compared to plasmid DNA transfection, and in particular, a high deletion efficiency of over 80% was observed for two types of sgRNA (sgRNA7 and sgRNA15). Next, CD3 deletion in human T cells was attempted using the RNP (ribonucleoprotein) electroporation method, which is known to be simpler and more efficient in gene editing, for sgRNA7 with good deletion efficiency, and the results are shown in Fig. 3. As shown in Fig. 3, the complex of recombinant Cas9 protein and sgRNA7 was in
Figure imgf000024_0001
CD3 deletion was achieved with an efficiency of over 70% in activated human T cells by electroporation produced in the form of RNP. As described above, a method for CD3 gene editing in human T cells using the CRISPR/Cas9 system was effectively established.

1.4여러 접합형 CAR-T세포에서의 CD3유전자편집 가능성 탐색 접합형 CAR-T세포제작을위해 항 CD3 어댑터 항체에 표지될 에피토프 태그의 후보로 니코틴 대사체인 코티닌, 펩타이드 에피토프인 6XHis 태그와 myc 태그를 선정하여, 각각 항-코티닌 CAR-T 세포 (Cot CAR-T 세포), 항- 6XHis 태그 CAR-T 세포 (His CAR-T세포), 및 항- myc 태그 CAR-T세포 (Myc CAR-T세포)를 제조하였다. 활성화된 마우스 T세포에 항-코티닌 항체 scFv (한국특허공개공보 제 2018- 0031727호)를 포함하는 CAR유전자를 탑재한 레트로바이러스를 형질 도입 (transduct ion)하여 항-코티닌 CAR-T세포를제작하였다. 동일한방법으로, 각각 기존에 보고된 항- myc 태그 항체의 scFv ( c 1 one 9E10, FEES Letters 414 (1997) 33 - 38)를 포함하는 CAR유전자 및 항- 6xHis 태그 항체의 scFv (mutated clone 3D5, J . Mol. Biol. (2002) 318, 135-147)를 포함하는 CAR유전자를 탑재한 레트로바이러스를 제작하고, 이들 레트로바이러스를 활성화된 마우스 T세포에 형질 도입 (transduct ion)하여 Myc CAR-T세포 및 His CAR-T세포를 제작하였다. 구체적으로, 마우스 CAR T세포의 경우 정상마우스로부터 얻은 비장과 림프절 세포를 a-CD3 항체 (145- 2C11, 10w/mt, BioXcell)가 코팅된 24웰 플레이트에 a- CD28 항체 (37.51, 2//g/mt, BD Biosciences)와 함께 가한후 T세포를 활성화하였다. 24시간 후, T세포를 24 °C , 2500rpm, 90분동안 6ug/ml의 폴리브렌 (polybrene, Sigma Aldrich) 존재하에 스핀 감염 (spin infection)을 통하여 농축된 레트로바이러스로 형질도입을 진행하였다. 이 절차는 같은 날 한번 더 반복하였다. 스핀 감염 후 T세포는 마우스 IL- 2 (30U/mt, Gibco) 존재 하에서 48시간동안 추가로 배양하였다. 이렇게 레트로바이러스 형질도입된 T세포를 2회 세척한후, 마우스 IL-2 (20U/mt)가 포함된 신선한 배양액을 가하여 2- 3일간 증식시켜 CAR T세포로 사용하였다. 세포 표면의 CAR 단백질의 발현은 a-토끼 IgG(Cot CAR), Q-마우스 IgG(His CAR, Myc CAR), His-비오틴 (비오틴이 표지된 6xHis 펩타이드)과 형광이 표지된 스트렙타비딘 (streptavidin)으로 염색한 후, 유세포측정법을 사용하여 측정하였다. 마우스 CD3 유전자편집 CAR- T세포는 레트로바이러스 형질도입 다음날 전기청공법을 이용하여 마우스 CD3 유전자편집을 진행하였다. 이들 각각의 세포 표면에서 CAR 단백질의 발현여부를 확인하였고, 이를 도 4a에 나타내었다. 또한, 케미컬 (코티닌) 및 펩타이드 (6XHis 태그 또는 myc 태그)에 대한 CAR-T 세포에 대해 CD3가 제거된 CAR-T 세포의 제작이 가능한지를 확인하기 위하여 , CAR- 레트로바이러스를 통한 형질도입과 CD3 deleting sgRNA/Cas9 mRNA 형질전환 (transfection)을 연속하여 진행하였고, CAR 발현과 CD3 제거가 동시에 유도된 CAR-T세포의 제작하였고, 이를 도 4b에 나타내었다. 도 4a 및 4b에 나타낸 바와 같이, 항-코티닌 CAR-T 세포의 경우 CD4 및 CD8양성세포에서 50-70% 정도로 관찰되었고, 세개의 CAR-T 세포 중에는 His CAR T세포에서 가장높은 발현율을 확인하였다. 이상의 결과를 통해 , 세 종류의 CAR-T세포 (Cot CAR-T, His CAR-T, Myc CAR- T 세포)에서 모두 CRISPR-Cas9 시스템을 이용한 CD3 유전자 편집이 가능함을 확인하였다. 1.4 Exploration of the possibility of CD3 gene editing in various conjugated CAR-T cells For the production of conjugated CAR-T cells, cotinine, a nicotine metabolite, and peptide epitopes 6XHis tag and myc tag were selected as candidates for the epitope tags to be labeled on the anti-CD3 adapter antibody, and anti-cotinine CAR-T cells (Cot CAR-T cells), anti-6XHis tag CAR-T cells (His CAR-T cells), and anti-myc tag CAR-T cells (Myc CAR-T cells), respectively, were produced. Anti-cotinine CAR-T cells were produced by transducing a retrovirus containing a CAR gene including an anti-cotinine antibody scFv (Korean Patent Publication No. 2018-0031727) into activated mouse T cells. Using the same method, retroviruses carrying a CAR gene including the scFv of a previously reported anti-myc tag antibody (c 1 one 9E10, FEES Letters 414 (1997) 33-38) and a CAR gene including the scFv of an anti-6xHis tag antibody (mutated clone 3D5, J. Mol. Biol. (2002) 318, 135-147) were constructed, and these retroviruses were transduced into activated mouse T cells to construct Myc CAR-T cells and His CAR-T cells. Specifically, for mouse CAR T cells, spleen and lymph node cells obtained from normal mice were added together with a-CD28 antibody (37.51, 2//g/mt, BD Biosciences) to a 24-well plate coated with a-CD3 antibody (145-2C11, 10w/mt, BioXcell), and T cells were activated. After 24 h, T cells were transduced with concentrated retrovirus through spin infection in the presence of 6 ug/ml polybrene (polybrene, Sigma Aldrich) at 24 °C, 2500 rpm, and 90 min. This procedure was repeated once more on the same day. After spin infection, T cells were additionally cultured in the presence of mouse IL-2 (30 U/mt, Gibco) for 48 h. The retrovirus-transduced T cells were washed twice, and then proliferated for 2-3 days in fresh culture medium containing mouse IL-2 (20 U/mt) to be used as CAR T cells. The expression of CAR protein on the cell surface was performed using a-rabbit IgG (Cot CAR), Q-mouse IgG (His CAR, Myc CAR), His-biotin (biotin-labeled 6xHis peptide) and fluorescently labeled streptavidin were stained and measured using flow cytometry. Mouse CD3 gene-edited CAR- T cells were subjected to mouse CD3 gene editing using electroporation the day after retroviral transduction. The expression of CAR protein on the surface of each of these cells was confirmed, and this is shown in Fig. 4a. In addition, in order to confirm whether CD3-deleted CAR-T cells were possible for CAR-T cells for chemicals (cotinine) and peptides (6XHis tag or myc tag), transduction via CAR- retrovirus and CD3 deleting sgRNA/Cas9 mRNA transfection were performed sequentially, and CAR-T cells in which CAR expression and CD3 deletion were simultaneously induced were produced, and this is shown in Fig. 4b. As shown in Figures 4a and 4b, in the case of anti-cotinine CAR-T cells, it was observed at about 50-70% in CD4 and CD8 positive cells, and the highest expression rate was confirmed in His CAR T cells among the three CAR-T cells. Through the above results, it was confirmed that CD3 gene editing using the CRISPR-Cas9 system was possible in all three types of CAR-T cells (Cot CAR-T, His CAR-T, and Myc CAR- T cells).

1.5접합형 CAR-T세포의 제작 접합형 인간 CAR-T세포 제작을 위해 상기 마우스 CAR-T세포 실험에 사용한 항 His CAR유전자를인간 CAR백본 (backbone)으로다시 디자인한후, 인간 CAR T세포용 레트로바이러스 벡터에 클로닝하여 제작하였다. 제작된 레트로바이러스를 말초혈액으로부터 분리한인간 T세포에 유전자이입하여 His CAR-T세포를제작하였다 구체적으로, 정상인 혈액으로부터 분리한 백혈구를항 CD3항체가코팅된 24웰 플레이트에 항 CD28항체와함께 가한후 48시간동안배양하여 T세포를활성화시켰다. 활성화된 T세포를 2회 세척한 후, 레트로바이러스 형질도입 (transduct ion)에 사용하였다. 레트로넥틴 (Retronectin)(20w/mt, TaKaRa)을 4°C에서 밤새 코팅한 후 세척한 24웰 플레이트에 2% BSA- DPBS를 가하여 37°C에서 30분간 blocking및 세척한 후, 레트로바이러스 농축액 Imt을 가하고 2000xg, 32°C에서 2시간동안 원심분리하여 레트로바이러스를 well밑면에 부착시켰다. Virus농축액을제거하고 well을세척 후, 활성화된 T세포 (1X106 cells/mt) Imt를 well에 가하고 10분간의 원심분리 (lOOOxg, 32°C)를 통해 세포를 레트로바이러스에 부착하였다. 이어서 인간 IL-2 (200U/mt, Pro leukin, Novartis) 존재 하에서 48시간동안배양하였다. 렌티바이러스형질도입의 경우, 48시간동안활성화된 인간 T세포배양액에 렌티바이러스를가한후 48시간동안 함께 배양하였다. 이렇게바이러스에 의한형질도입된 T세포를 2회 세척하여 배양액에 남은 바이러스를 제거하고, 3일씩 2번 (3일 +3일) 인간 IL-2 (200U/mt)가 포함된 배양액을 가하여 CAR T세포를 제작하였다. 도 5는 His 태그를 도입한 인간 CAR T세포에서의 6xHis-CAR 발현율을 분석한 그래프이다. 또한, 하기 표 3에 His CAR-T세포의 도메인별 아미노산서열을 나타내었다.1.5 Production of conjugated CAR-T cells To produce conjugated human CAR-T cells, the anti-His CAR gene used in the mouse CAR-T cell experiment was redesigned as a human CAR backbone, and then cloned into a retroviral vector for human CAR T cells to produce them. The produced retrovirus was genetically introduced into human T cells isolated from peripheral blood to produce His CAR-T cells. Specifically, leukocytes isolated from normal blood were added to a 24-well plate coated with anti-CD3 antibody together with anti-CD28 antibody, and then cultured for 48 hours to activate the T cells. The activated T cells were washed twice and then used for retroviral transduction. After coating with Retronectin (20w/mt, TaKaRa) overnight at 4°C, 2% BSA-DPBS was added to the washed 24-well plate, blocked at 37°C for 30 minutes, and washed. Retrovirus concentrate Imt was added, and centrifuged at 2000xg and 32°C for 2 hours to attach the retrovirus to the bottom of the well. After removing the virus concentrate and washing the well, activated T cells ( 1X106 cells/mt) Imt were added to the well, and centrifuged for 10 minutes (lOOOxg, The cells were attached to the retrovirus via the 32°C medium. Subsequently, they were cultured for 48 hours in the presence of human IL-2 (200 U/mt, Pro leukin, Novartis). In the case of lentivirus transduction, lentivirus was added to the human T cell culture medium activated for 48 hours, and then cultured together for 48 hours. The T cells transduced with the virus in this way were washed twice to remove the virus remaining in the culture medium, and the culture medium containing human IL-2 (200 U/mt) was added twice every 3 days (day 3 + day 3) to produce CAR T cells. Figure 5 is a graph analyzing the expression rate of 6xHis-CAR in human CAR T cells introduced with the His tag. In addition, the amino acid sequence by domain of His CAR-T cells is shown in Table 3 below.

【표 3]

Figure imgf000026_0001
도 5에 나타낸 바와같이, 유세포분석을통하여 CAR발현율을분석한결과, 약 40%정도의 유전자 전달 효율으로 인간 항 6xHis CAR T세포가 성공적으로 제작됨을 확인하였다. 실시예 2. 항 CD3 어댑터 항체의 제조 【Table 3]
Figure imgf000026_0001
As shown in Fig. 5, the CAR expression rate was analyzed through flow cytometry, and it was confirmed that human anti-6xHis CAR T cells were successfully produced with a gene transfer efficiency of approximately 40%. Example 2. Production of anti-CD3 adapter antibody

2.1 항 CD3 항체를통해 어댑터 항체 제조 - 0KT3, UCHT1 코티닌과 같은 케미컬 태그의 경우, 화학반응을 통해 어댑터에 접합해야 하므로, 일정한 접합효율을 유지하기 어렵고, 하나의 어댑터에 접합하는 케미컬의 분자수 (Drug Antibody Ratio, DAR)를 조절하기 어려운 단점이 있다, 반면, 6xHis 태그나 myc 태그와 같은 펩타이드 에피토프의 경우, 재조합 항체 ORF유전자에 펩타이드를 유전자 재조합을 통해 에피토프 태그 (epitope tagging)방식으로 연결할 수 있으므로, 100% 접합효율과 함께, 하나의 어댑터에 하나의 태그가 결합되는 1:1 DAR를 유지할 수 있다는 장점이 있다. 특히, 6xHis 펩타이드는 단백질 정제용 에피토프 태그로많이 사용되어 왔고, 따라서 이를 이용한항체의약품의 대량정제를 위한 시스템이 잘 갖추어져 있으므로, 한가지 에피토프 태그를 이용하여 접합형 His CAR T세포의 어댑터 에피토프 태그 역할과 어댑터 정제를위한 에피토프 태그 역할의 두가지 목적을동시에 달성할수 있다는장점이 있다. 또한, 상대적으로생체내에서의 면역원성이 낮은 장점이 있다. 이에, 6xHis 태그를 항- CD3 어댑터 항체에 이용하였다. 시판되는 항-인간 CD3항체 0KT3(Muromonab) 서열 또는 UCHT1서열을 이용하여 6xHis- tagged 어댑터 항체를 scFv형태로 제작하였다. OKT항체 또는 UCHT1항체의 VL 부위와 VH 부위를 링커 ((GGGGS)J로 연결한 후 (scFv), 분리정제 및 안정성을 위해 사람 면역글로불린 카파 불변 영역 (CK )을 연결하고 c-말단에 6xHis 태그를 바로 연결하거나, 짧은 링커 펩타이드와 6xHis 태그를 연결하는 작제물 (construct)를 제작하였다. 이의 모식도를 도 6에 나타내었고, 각각 0KT3의 scFv 아미노산 서열을 표 4 및 표 5에 나타내었다. 이 때, 표 4는 Ck도메인 다음에 링커를 포함하지 않은 scFv (0KT3-Ck-His), 표 5는 Ck 도메인 다음에 링커를 포함하는 scFv (0KT3- Ck- L- His) 아미노산서열을 나타낸 것이다. 또한, UCHT1의 scFv (UCHTl-Ck-His) 아미노산 서열을 아래 표 6에 나타내었다. 상기 작제물을 발현하는 플라스미드를 HEK293K 세포주에 형질전환 후, 친화도 크로마토그래프를 통해 정제를 하였고, 이를 도 7에 나타내었다. 2.1 Production of adapter antibodies via anti-CD3 antibodies - In the case of chemical tags such as 0KT3, UCHT1 cotinine, it is difficult to maintain a constant conjugation efficiency because they must be conjugated to the adapter through a chemical reaction, and there are disadvantages in that it is difficult to control the number of molecules of the chemical conjugated to one adapter (Drug Antibody Ratio, DAR). On the other hand, 6xHis In the case of peptide epitopes such as tag or myc tag, since the peptide can be linked to the recombinant antibody ORF gene through genetic recombination in the form of an epitope tag, there is an advantage of maintaining 100% conjugation efficiency and a 1:1 DAR in which one tag is bound to one adapter. In particular, the 6xHis peptide has been widely used as an epitope tag for protein purification, and therefore a system for mass purification of antibody drugs using it is well-established, so there is an advantage of being able to achieve two purposes at the same time, that is, the role of the adapter epitope tag for the conjugated His CAR T cell and the role of the epitope tag for adapter purification, using one epitope tag. In addition, there is an advantage of relatively low immunogenicity in vivo. Therefore, the 6xHis tag was used in the anti-CD3 adapter antibody. 6xHis-tagged adapter antibodies were produced in scFv form using the commercially available anti-human CD3 antibody 0KT3 (Muromonab) sequence or UCHT1 sequence. A construct was produced by connecting the VL region and the VH region of OKT antibody or UCHT1 antibody with a linker ((GGGGS)J, and then connecting the human immunoglobulin kappa constant region (CK) for separation and stability, and directly connecting a 6xHis tag to the C-terminus or connecting a short linker peptide and a 6xHis tag. A schematic diagram of the construct is shown in Fig. 6, and the amino acid sequences of the scFv of 0KT3 are shown in Tables 4 and 5, respectively. At this time, Table 4 shows the amino acid sequence of the scFv (0KT3-Ck-His) that does not include a linker after the Ck domain, and Table 5 shows the amino acid sequence of the scFv (0KT3-Ck-L-His) that includes a linker after the Ck domain. In addition, the amino acid sequence of the scFv (UCHTl-Ck-His) of UCHT1 is shown in Table 6 below. The above constructs After the expression plasmid was transformed into the HEK293K cell line, it was purified through affinity chromatography, and the results are shown in Figure 7.

【표 4]

Figure imgf000027_0001
Figure imgf000028_0001
【Table 4]
Figure imgf000027_0001
Figure imgf000028_0001

【표 5]

Figure imgf000028_0002
【Table 5]
Figure imgf000028_0002

【표 6]

Figure imgf000028_0003
scFv형태뿐 아니라, 어댑터 항체의 CAR결합력을 증가시키는 일환으로, 2개의 6xHis 태그를 보유하여 2가 (bi valent) 형태로 6xHis CAR에 결합할 수 있도록 Fab 형태로도 -His 어댑터 항체를 제작하였고, 이의 모식도를 도 8에 나타내었고, 각각 0KT3 및 UCHT1의 Fab (0KT3- Fab- His , UCHTl-Fab-His) 아미노산서열을 아래 표 7 및 8에 나타내었다. 【Table 6]
Figure imgf000028_0003
In addition to the scFv form, as part of an effort to increase the CAR binding affinity of the adapter antibody, a -His adapter antibody was also produced in a Fab form that possesses two 6xHis tags so that it can bind to 6xHis CAR in a bivalent form. A schematic diagram of this is shown in Fig. 8, and the amino acid sequences of the Fabs of 0KT3 and UCHT1 (0KT3-Fab-His, UCHT1-Fab-His), respectively, are shown in Tables 7 and 8 below.

【표 7]

Figure imgf000029_0001
【Table 7]
Figure imgf000029_0001

【표 8]

Figure imgf000029_0002
Figure imgf000030_0001
【Table 8]
Figure imgf000029_0002
Figure imgf000030_0001

2.2신규 항 CD3 항체를통해 어댑터 항체 제조 - 1-4-2, 1-4-7 항-인간 CD3항체는 상기 0KT3 또는 UCHT1 항체뿐 아니라, 동물 면역 항체 라이브러리를 이용하여 CD3 결합 항체를 추가로 스크리닝하였다. 구체적으로, 사람 CD3 감마/입실론과 델타/입실론 세포외 해테로다이머단백질을 adjuvant와 혼합하여 2주 간격으로 총 4회 각각 닭 10마리에 면역한 후, 닭의 비장, 골수, 활액낭을 수거하여 이로부터 total RNA분리, cDNA 합성 variable 경쇄 &중쇄 유전자 PCR을 수행하고, 파지 디스플레이용 벡터인 PComb3XSS에 클로닝 함으로써 닭 면역 항체 라이브러리를 제작하였다. 이후, bio- panning을 수행하여 사람 CD3 단백질에 특이적으로 결합하는 항체 클론을 스크리닝하였다. 양성 항체 클론을 scFv형태로 분리정제한 후, CD3(+) Jurkat cell에 결합하는 항체 2종 (1-4- 2, 1-4- 7클론)을 최종 선별하였다. 선별한 항체에 대하여, scFv-Ck-6xHis 융합단백질 형태로 생산하기 위하여 동물 세포 발현용 벡터에 클로닝을 진행하고 HEK293F 세포주에 형질 전환하였다. 5일간 진탕 배양 후 배양액을 회수하여 KappaSelect bead를 이용한 친화도 정제하였고, 항체의 발현을 SDS- PAGE를 통해 확인하였다. 즉, 신규 스크리닝 항체 1-4-2 및 1-4- 7에 대하여도, scFv 및 Fab 형태의 어댑터 항체를 나타내었고, 하기 표 9 내지 12에 이의 아미노산 서열을 내타내었고, 특히 1-4-2, 및 1-4- 7의 scFv (1-4-2-Ck-His, 1- 4- 7- Ck- His)의 SDS-PAGE분석 결과를 도 9에 나타내었다.2.2 Production of adapter antibodies using novel anti-CD3 antibodies - 1-4-2, 1-4-7 In addition to the above 0KT3 or UCHT1 antibodies, anti-human CD3 antibodies were additionally screened for CD3 binding antibodies using an animal immune antibody library. Specifically, human CD3 gamma/epsilon and delta/epsilon extracellular heterodimer proteins were mixed with an adjuvant and immunized in 10 chickens four times in total at two-week intervals. Then, the spleen, bone marrow, and synovial bursa of the chickens were collected, total RNA was isolated, cDNA was synthesized, variable light chain & heavy chain genes were PCR-processed, and cloned into P Comb3XSS, a phage display vector, to produce a chicken immune antibody library. Thereafter, bio-panning was performed to screen for antibody clones that specifically bind to human CD3 protein. After purifying the positive antibody clones in the form of scFv, two antibodies (clones 1-4-2 and 1-4-7) that bind to CD3(+) Jurkat cells were finally selected. For the selected antibodies, they were cloned into an animal cell expression vector to produce them in the form of scFv-Ck-6xHis fusion proteins, and then transformed into HEK293F cells. After culturing with shaking for 5 days, the culture medium was harvested and affinity purified using KappaSelect beads, and the expression of the antibodies was confirmed through SDS-PAGE. That is, for the novel screening antibodies 1-4-2 and 1-4-7, scFv and Fab types of adapter antibodies were shown, and the amino acid sequences thereof are shown in Tables 9 to 12 below, and in particular, the results of SDS-PAGE analysis of scFv (1-4-2-Ck-His, 1-4-7-Ck-His) of 1-4-2 and 1-4-7 are shown in Figure 9.

【표 이

Figure imgf000030_0002
【Table 2
Figure imgf000030_0002

Figure imgf000031_0001
Figure imgf000031_0001

【표 10]

Figure imgf000031_0002
【Table 10]
Figure imgf000031_0002

【표 11]

Figure imgf000031_0003
Figure imgf000032_0001
【Table 11]
Figure imgf000031_0003
Figure imgf000032_0001

【표 12]

Figure imgf000032_0002
구체적으로, 항 인간 CD3(UCHT1, 1-4-2, 1-4-7) Fab- His항체 및 항 인간 CD3(0KT3, UCHT1 , 1-4-2, 1-4-7) scFv- His항체는 항체 발현 벡터인 pCEP4에 유전자를 클로닝하고 293F세포에 형질도입하여 배양액을 통해 발현하였다. 발현된 항체는 친화도 정제를 통해 배양액 내에 존재하는 항체를 분리 정제하였다. 정제된 항체의 경우 SDS-폴리아크릴아마이드 겔 (SDS-polyacrylamide gel)을 이용한전기영동 방법과 인간 CD3를 발현하는 세포주에 유세포측정법을 통하여 세포표면 결합을 검증하였다. CD3 양성 세포와 어댑터 항체의 결합 검증은 lX105cells/lug 어댑터를 4 °C에 1시간 배양하였다. 이후 자유 어댑터 항체를 제거하기 위해 세척 후, 어댑터 항체를 잡는 항 His항체 (J096G46, Biolegend)로 염색한 뒤 유세포측정법을 이용하여 분석하였다. 이상과 같이 , 발현 및 정제된 항 인간 CD3 항체가 T세포종양표면에 발현하는 CD3단백질과 결합하는지를 분석하기 위하여, 제조한 0KT3-His scFv (0KT3- Ck- His , 0KT3-Ck-L-His), UCHT1- Fab- His에 대해서는 Jurkat T세포에 결합여부를 유세포분석 (flow cytometry)한 결과를 도 10에 나타내었다. 동일한 방법으로 1-4 - 2의 scFv 및 Fab, 1-4- 7의 scFv 및 Fab 항체단백질에 대해서도 유세포분석 (flow cytometry)을수행하였고, 그 결과를 도 11에 나타내었다. 도 10에 나타낸 바와 같이 , 항 CD3 항체로 제조한 scFv, 및 Fab가 Jurkat 세포주에 특이적으로 결합하는 것을 확인하였다. 도 11에 나타낸 바와 같이, CD3음성 세포주 (CD3- deleted Jurkat)에는 결합하지 않고, Jurkat 세포주에만특이적으로 결합하는 것을 확인하였다. 실험예 1. 항 CD3 어댑터 항체와 인간 접합형 CAR-T 세포의 항종양 활성
Figure imgf000033_0001
상기 실시예 2에서 제작한 6xHis- tagged 항 CD3 어댑터 항체와 인간 항- 6xHis CAR-T 세포 (His CAR-T 세포) 복합체가 CD3양성 T세포 종양을 인지하여 활성화되는지를 in vitro에서 검증하기 위해, Jurkat leukemia세포와 0KT3 scFv-Ck- 6xHis(0KT3-Ck-His), 0KT3 scFv- Ck- 6xHis(0KT3- Ck- L- His)및 마우스 His CAR-T세포를 공배양한 후, CAR-T세포가분비하는 IFN- r의 양을 ELISA로측정하였다. 그결과, 두 가지 어댑터 존재하에서 T세포의 IFN- r생성이 증가함에 따라 CAR- T세포의 기능이 활성화됨을 확인하였다. 또한 어댑터 용량을 5배 증가함에 따라 그 효과가 더 증가하는 용량의존도를 보였다. 그 결과를 도 12a에 나타내었다. 이어서 , 두어댑터 중좀 더 활성이 좋은 0KT3 scFv- Ck- 6xHis(0KT3- Ck- His)를 Jurkat leukemia세포 및 인간 His CAR-T세포를공배양한후, CAR-T세포가분비하는 IFN- r의 양을 ELISA로 측정하였다. 또한, Jurkat 종양세포에 대한 세포살상능 (Cytotoxicity assay)을 확인하기 위한 실험을 진행하였고, 그 결과를 도 12b에 나타내었다. 동일한 방법으로, scFv어댑터 항체를마우스접합형 CAR T세포모델에서 검증된 Fab-His형태로 변환시켜 Jurkat 종양세포에 대한세포살상능실험을반복하였고, 그 결과를 도 13에 나타내었다. 또한, 항- CD3항체의 다른 클론인 UCHT1항체 및 스크리닝을 통해 선별한 두 종의 항체 (1-4- 2, 및 1-4- 7)를 이용하여 Fab-His 어댑터 항체를 제작하고, 이를 이용하여 접합형 CAR T세포의 in vitro 활성 및 세포살상능을 동일한 방법으로 분석하였다. 그 결과를 각각도 14(UCHT1) 및 도 15(1-4-2, 1-4- 7)에 나타내었다. 도 12내지 15에 나타낸바와같이 , 접합형 CAR T세포는항 CD3어댑터 항체의 존재하에서만 농도의존적으로 활성화되어 IFN- r를 다량 생산하고, 종양살상능을 보였다. 이상의 결과를통해 , 항 CD3어댑터 항체와 CAR T세포복합체의 암항원 인지력 , 특이적인 활성화 및 세포살상능력을 확인하였다. 접합형 CAR T세포의 종양살상능, 사이토카인 생성능 및 동족살상 방지기능 츺정 어댑터 항체를통해 종양세포를 인식하여 접합형 CAR- T세포가활성화되는지를 테스트하기 위해, 종양세포를 어댑터 항체와 1시간 동안 미리 배양 후, 붙지 않은 free 어댑터 항체를 세척을 통해 제거하였다. 이후 어댑터가 부착된 종양세포를 접합형 CAR- T세포와 함께 시험관내 실험에 사용하였다. Luciferase 기반종양살상능 측정의 경우, 레트로바이러스 또는 렌티바이러스 형질도입 후 증식된 CAR T세포 (1.2X103〜 7.5 X105 cells/100M/well)를, Jurkat— Luc세포 (3X104cel ls/50M/wel 1 )에 여러 비율로 (CAR- T:Jurkat=0.04〜 25:1)로 가하여 96웰 플레이트에서 밤새 공동배양 (co- culture)한 후, D-Luciferin (600/zg/mt, Pr omega) 50/z£를 가한 후 37°C에서 10분간 배양하여, 그 때까지 생존한 Jurkat- Luc세포에서의 luciferase 효소작용을 유발하였다. 이들 세포의 발광도를 Luminometer (Tecan)를 이용하여 측정하여, CAR T세포를 처리하지 않은 Jurkat- Luc세포의 발광도와 비교하여 종양세포의 생존율을 계산함으로써 CAR T세포의 종양살상능을 계측하였다 . 【Table 12]
Figure imgf000032_0002
Specifically, anti-human CD3 (UCHT1, 1-4-2, 1-4-7) Fab-His antibody and anti-human CD3 (0KT3, UCHT1, 1-4-2, 1-4-7) scFv-His antibody were cloned into the antibody expression vector pCEP4, transduced into 293F cells, and expressed in the culture medium. The expressed antibodies were separated and purified through affinity purification to separate the antibodies present in the culture medium. For the purified antibodies, cell surface binding was verified using an electrophoresis method using SDS-polyacrylamide gel and flow cytometry on a cell line expressing human CD3. The binding of CD3-positive cells to the adapter antibody was verified by incubating 1X10 5 cells/lug adapter at 4 °C for 1 hour. After washing to remove free adapter antibodies, the cells were stained with anti-His antibody (J096G46, Biolegend) that captures the adapter antibodies, and then analyzed using flow cytometry. As described above, in order to analyze whether the expressed and purified anti-human CD3 antibody binds to the CD3 protein expressed on the surface of T cell tumors, the binding of the manufactured 0KT3-His scFv (0KT3-Ck-His, 0KT3-Ck-L-His) and UCHT1-Fab-His to Jurkat T cells was analyzed by flow cytometry, and the results are shown in Fig. 10. In the same way, flow cytometry was performed on the scFv and Fab of 1-4-2, and the scFv and Fab of 1-4-7, and the results are shown in Fig. 11. As shown in Fig. 10, it was confirmed that the scFv and Fab manufactured as anti-CD3 antibodies specifically bound to the Jurkat cell line. As shown in Fig. 11, it was confirmed that it specifically binds only to the Jurkat cell line, but not to the CD3-negative cell line (CD3-deleted Jurkat). Experimental Example 1. Antitumor activity of anti-CD3 adapter antibody and human conjugated CAR-T cells
Figure imgf000033_0001
To verify in vitro whether the complex of the 6xHis-tagged anti-CD3 adapter antibody and human anti-6xHis CAR-T cell (His CAR-T cell) produced in the above Example 2 recognizes and activates CD3-positive T cell tumors, Jurkat leukemia cells were co-cultured with 0KT3 scFv-Ck-6xHis (0KT3-Ck-His), 0KT3 scFv- Ck- 6xHis (0KT3- Ck- L- His), and mouse His CAR-T cells, and the amount of IFN- r secreted by the CAR-T cells was measured by ELISA. As a result, it was confirmed that the function of CAR-T cells was activated as the IFN- r production of T cells increased in the presence of the two adapters. In addition, it showed a dose-dependency in which the effect further increased as the adapter dose was increased fivefold. The results are shown in Fig. 12a. Next, 0KT3 scFv- Ck- 6xHis (0KT3- Ck- His), which has better activity among the two adapters, was co-cultured with Jurkat leukemia cells and human His CAR-T cells, and the amount of IFN- r secreted by CAR-T cells was measured by ELISA. In addition, an experiment was conducted to confirm the cytotoxicity assay for Jurkat tumor cells, and the results are shown in Fig. 12b. Using the same method, the scFv adapter antibody was converted to the Fab-His form verified in the mouse conjugated CAR T cell model, and the cytotoxicity experiment for Jurkat tumor cells was repeated, and the results are shown in Fig. 13. In addition, Fab-His adapter antibodies were produced using UCHT1 antibody, which is another clone of anti-CD3 antibody, and two antibodies (1-4-2 and 1-4-7) selected through screening, and the in vitro activity and cell killing ability of conjugated CAR T cells were analyzed using the same method. The results are shown in Fig. 14 (UCHT1) and Fig. 15 (1-4-2, 1-4-7), respectively. As shown in Figs. 12 to 15, the conjugated CAR T cells were activated in a concentration-dependent manner only in the presence of anti-CD3 adapter antibody, produced a large amount of IFN- r, and exhibited tumor killing ability. Through the above results, the cancer antigen recognition ability, specific activation, and cell killing ability of the anti-CD3 adapter antibody and CAR T cell complex were confirmed. To test whether conjugated CAR-T cells are activated by recognizing tumor cells via the adaptor antibody, tumor cells were pre-incubated with the adaptor antibody for 1 hour, and then unbound free adaptor antibody was removed by washing. After that, the tumor cells with the adaptor attached were used in in vitro experiments together with the conjugated CAR-T cells. For luciferase-based tumor killing activity measurement, CAR T cells (1.2 X 10 3 to 7.5 X 10 5 cells/100 M/well) expanded after retrovirus or lentivirus transduction were added to Jurkat-Luc cells (3 X 10 4 cells/50 M/well 1) at various ratios (CAR- T: Jurkat = 0.04 to 25:1) and co-cultured overnight in a 96-well plate. Then, D-Luciferin (600/zg/mt, Pro omega) 50/z£ was added and incubated at 37°C for 10 minutes to induce luciferase enzyme activity in Jurkat-Luc cells that survived until then. The tumor killing ability of CAR T cells was measured by measuring the luminescence of these cells using a Luminometer (Tecan) and comparing it with the luminescence of Jurkat-Luc cells that were not treated with CAR T cells to calculate the survival rate of tumor cells.

CAR T세포의 사이토카인 생성능을 측정하기 위하여, CAR T세포와 표적세포 (Jurkat세포)를 1:5비율로 (CAR- T:3X104 cells, Jurkat : 1.5X105 cells) 혼합하여 96 웰 플레이트에서 24시간동안공동배양한후, 배양상층액을수확하였다. 상층액으로 분비된 IFN- x의 양을 ELISA법 (마우스 IFNx , 인간 IFNy ELISA kit, BD Biosciences)으로즉정하였다. 실험예 2. 인간접합형 CAR-T세포의 동족살상 (fratricide) 현상분석 접합형 CAR T세포가항 CD3 어댑터 항체와결합하였을 때, 접합형 CAR T세포가 다른 CAR T세포표면의 CD3를 표적으로 삼아 서로 살상하는 동족살상 (fratricide) 현상이 일어나고, 유전자편집에 의한 CD3 제거가 이러한 동족살상현상을 방지할 수 있는지를 in = 에서 검증하였다. 구체적으로, 항 6xHis- CAR T세포 단독으로 (대조군) 또는 6xHis- tagged 항 CD3 어댑터 항체와 인간 항 6xHis- CAR T세포를 함께 (실험군) 배양한 후, 37°C에서 각각 배양후 24시간, 48시간, 72시간 경과한 시점에서 CAR T세포의 생존율을 유세포분석법으로 분석하였다. 생존율분석은 사멸된 세포의 日때에 결합하는 형광물질인 7- AAD를이용하여 7- AAD음성 세포 (live cel Is)의 비율을분석하는방법을 이용하였고, 그 결과를 도 16에 나타내었다. 도 16에 나타낸 바와 같이, 항 CD3 어댑터 항체 존재하에서 항 6xHis- CAR T세포의 생존율이 현저히 떨어짐을 확인함으로써 , 동족살상이 유발됨이 확인되었다. 따라서 , 항 CD3 어댑터 항체를통해 접합형 인간 CAR T세포는 CD3유전자편집에 의한 CD3 단백질의 제거가 필요함이 실험적으로 확인하였다. 실험예 3. 인간 CD3편집 접합형 CAR-T세포의 제작및동족살상방지기능검증 상기 실험예 2에서 확인한 바와 같이 , 동족살상현상이 배제된 CAR T세포를 제작하기 위해 CD3 유전자가 편집되고 동시에 접합형 CAR를 발현하는 인간 T세포를 제작하였다. 구체적으로, 말초혈액으로부터 분리한 인간 T세포에 CD3 deleting sgRNA/Cas9 protein RNP 형질전환 (transfect ion)과 CAR-레트로바이러스 형질도입 (transduction)을 순차적으로 진행한 결과, CAR발현과 CD3 제거가 동시에 유도된 인간 T세포의 제작하였고, 각각 CAR의 발현 및 CD3의 발현 여부를 유세포분석을 통해 확인하였다. CD3 제거와 CAR의 도입하는 과정에서의 유세포분석 결과를 도 17에 나타내었다. 또한, CD3의 제거를 통해 접합형 CAR T세포의 동족살상방지 기능을 검증하기 위해, 7AAD 형광염색을 통한 세포생존율 분석실험을 진행하였다. 구체적으로, 각각 (DHis CAR T세포, ② His CAR T세포 + 항 CD3 어댑터 항체 (0KT3- His) , ③ CD3 deleted His CAR T세포, @CD3 deleted His CAR T + 항 CD3 어댑터 항체(0紅3-田8)세포로 나누어 37°C에서 24시간동안배양한후, 7AAD형광염색을통한세포생존율분석하였다. 또한, (DHis CAR T세포, ② His CAR T세포 + 항 CD3 어댑터 항체 (0KT3- His) , ©CD3 deleted His CAR T + 항 CD3 어댑터 항체(0紅3-田8)세포의 CD3 발현 정도를 유세포 분석을 통해 분석하였다. 그 결과를 도 18에 나타내었다. 도 17에 나타낸 바와 같이, 인간 CD3 유전자 편집 접합형 CAR T세포가 성공적으로 제작되었음을 확인하였다. 도 18에 나타낸 바와 같이, 항 CD3 어댑터 항체의 존재하에서, CD3 유전자 편집된 CAR T세포의 생존율 (73.1%)이 CD3가 제거되지 않은 CAR T세포의 생존율 (43.3%)에 비해 월등히 높음을 확인하였다 (24시간 배양조건에서 73.1% vs 43.3%). 또한, @CD3 deleted His CAR T + 항 CD3 어댑터 항체 (0KT3- His)군에서 CD3발현 또한 현저히 감소되어 있음을 확인하였다. 이상의 결과를통해, CD3유전자편집을 통하여 동족살상현상이 배제된 항 CD3 접합형 CAR T세포가성공적으로제작되었음을 확인하였다. 실험예 4. 항 CD3 어댑터 항체와 인간 접합형 CAR-T세포의 항종양활성 분석 ( in vivo) To measure the cytokine production ability of CAR T cells, CAR T cells and target cells (Jurkat cells) were mixed at a ratio of 1:5 (CAR- T: 3X104 cells, Jurkat: 1.5X105 cells) and co-cultured in a 96-well plate for 24 hours, and then the culture supernatant was harvested. The amount of IFN- x secreted into the supernatant was measured by ELISA (mouse IFNx, human IFNy ELISA kit, BD Biosciences). Experimental Example 2. Analysis of the fratricide phenomenon of human conjugated CAR-T cells When conjugated CAR T cells are combined with an anti-CD3 adapter antibody, the conjugated CAR T cells target CD3 on the surface of other CAR T cells and kill each other, which is a phenomenon called fratricide. We verified in = whether CD3 removal by gene editing can prevent this fratricide phenomenon. Specifically, anti-6xHis- CAR T cells were cultured alone (control group) or together with 6xHis-tagged anti-CD3 adapter antibodies and human anti-6xHis- CAR T cells (experimental group), and the viability of CAR T cells was analyzed by flow cytometry at 24 hours, 48 hours, and 72 hours after culture at 37°C, respectively. The viability analysis was performed using a method that analyzes the ratio of 7-AAD-negative cells (live cells) by utilizing 7-AAD, a fluorescent substance that binds to the surface of dead cells, and the results are shown in Fig. 16. As shown in Fig. 16, it was confirmed that fratricide was induced by confirming that the survival rate of anti-6xHis- CAR T cells was significantly reduced in the presence of anti-CD3 adapter antibodies. Therefore, it was experimentally confirmed that conjugated human CAR T cells require the removal of CD3 protein by CD3 gene editing through anti-CD3 adapter antibodies. Experimental Example 3. Production of human CD3-edited conjugated CAR-T cells and verification of anti-fratricide function As confirmed in the above Experimental Example 2, in order to produce CAR T cells that exclude fratricide, human T cells in which the CD3 gene was edited and which simultaneously express the conjugated CAR were produced. Specifically, CD3 deleting sgRNA/Cas9 protein RNP transfection and CAR retrovirus transduction were sequentially performed on human T cells isolated from peripheral blood, resulting in the production of human T cells in which CAR expression and CD3 deletion were simultaneously induced, and CAR expression and CD3 expression were confirmed, respectively, through flow cytometry. The results of flow cytometry analysis during the process of CD3 deleting and CAR introducing are shown in Fig. 17. In addition, to verify the anti-fratricide function of conjugated CAR T cells through CD3 deleting, a cell viability analysis experiment using 7AAD fluorescence staining was performed. Specifically, each was divided into ① (DHis CAR T cells, ② His CAR T cells + anti-CD3 adapter antibody (0KT3-His), ③ CD3 deleted His CAR T cells, @CD3 deleted His CAR T + anti-CD3 adapter antibody (0紅3-田8) cells and cultured at 37°C for 24 hours, and then cell viability was analyzed using 7AAD fluorescence staining. In addition, the CD3 expression level of (DHis CAR T cells, ② His CAR T cells + anti-CD3 adapter antibody (0KT3-His), ©CD3 deleted His CAR T + anti-CD3 adapter antibody (0紅3-田8) cells was analyzed using flow cytometry. The results are shown in Fig. 18. As shown in Fig. 17, it was confirmed that human CD3 gene-edited conjugated CAR T cells were successfully produced. As shown in Fig. 18, in the presence of anti-CD3 adapter antibodies, the survival rate of CD3 gene-edited CAR T cells (73.1%) was significantly higher than that of CAR T cells in which CD3 was not deleted (43.3%) (73.1% vs. 43.3% under 24-hour culture conditions). In addition, it was confirmed that CD3 expression was also significantly reduced in the @CD3 deleted His CAR T + anti-CD3 adapter antibody (0KT3-His) group. Through the above results, it was confirmed that anti-CD3 conjugated CAR T cells that excluded fratricide were successfully produced through CD3 gene editing. Experimental Example 4. Analysis of anti-tumor activity of anti-CD3 adapter antibodies and human conjugated CAR-T cells (in vivo)

4.1 인간 CAR T세포의 증식조건 확립 4.1 Establishment of proliferation conditions for human CAR T cells

CD3 유전자 편집 접합형 인간 CAR T세포와 항 CD3 어댑터 항체를 이용한 종양치료효과를 생체내 (in r/w)에서 확인하기 위하여 충분한 T세포 수의 확보가 필수적이므로, 인간 CAR T세포의 대량증식 조건의 확립이 필요하다. 1차적으로, CD3 양성 세포를 제거하여 CD 음성 세포의 분리 정제 효율을 높인 후, 실험에 필요한 충분한 수의 CD3 유전자편집 CAR T세포를 확보하기 위해, 분리 정제 후 4일간 hlL- 2(300U/ml)를 처리하여 2차 세포증식을 유도하였다. 이와 같이, 확립된 CD3 유전자 편집 접합형 CAR T세포 증식조건 모식도는 도 19에 나타내었다. 상기 실험예 3에서 CRISPR/Cas9 형질전환 (transfect ion) 및 CAR transduct ion을 통해 제작한 인간 CD3 유전자 편집 접합형 CAR T세포는 일부 CD3 양성인 세포를 포함한다. 동족살상현상을 방지하고 , CAR T의 표적 효율을 높이기 위해 CD3 음성인 세포를 정제하는 과정이 필요하고, 이에 항-인간 CD3 magnetic bead를 이용하여 MACSGnagnet- associated cel 1 sorting)를 통해 negat ive selection을수행하였다. 구체적으로, 인간 CD3 유전자 편집 CAR T세포의 경우, 세포 활성화 2일 후, 전기천공법을 통한 CD3 단백질 유전자편집 후 레트로바이러스 혹은 렌티바이러스 형질도입 방법을사용하여 CD3 유전자편집 접합형 CAR- T세포를 제작하였다. 유전자 편집 이후 남아있는 CD3 양성 세포의 경우 자기 활성화 세포 분류법 (Magent- associated cell sorting, MACS)을사용하여 CD3 음성세포만을분류하였다. T세포의 CD3를 CRISPR/Cas9 유전자편집 기술로 제거 후 5일 뒤 , CD3 유전자 편집 T세포를 마그네틱 미세구술이 붙은 항 인간 CD3 항체와 배양하였다. 이후 마그네틱 미세구술- 항체 (hCD3 mi crobead , Mi l tenyi )와 배양한 세포를 강한 자성을 띄는 마그네틱 분리기에 통과시켜 , 유전자편집이 되지 않은 CD3 양성 세포는 마그네틱 미세구술- 항체가 붙어 있어 마그네틱 분리기에 부착됨 . 반면에 , CD3 음성세포들은 분리기에 붙지 않고 통과되어 따로 분리됨 . 이렇게 분리된 CD3 음성세포를 hIL- 2(200U/ml )포함 배양액에 추가 배양하였다. 세포표면의 CAR단백질의 발현은 , 레트로바이러스 형질도입 5일 뒤부터 증식된 CAR T세포를 Hi s-비오틴 및 APC가 표지된 SA (SA-APC , BioLegend)로 염색한 후 , 유세포측정법 ( f low cytometry) (FACS-Canto I I , BD Biosciences)으로 즉정하였다. 인간 CD3 유전자 편집 접합형 CAR T세포에 Ant i-CD3 MACS 과정을 거침으로써 CD3음성 인간 CAR T세포를 효과적으로 분리 정제할 수 있는지 확인하였고 , 2차 세포증식 기간 이후 T세포군에서 CD3 음성 상태를 유지하는지 여부를 확인하였다. 그 결과를 도 20 및 도 21에 나타내었다. 도 20에 나타낸 바와 같이 , 2차 세포 증식 기간 이후 T세포군은 대부분 CD3 음성 상태 및 CAR발현 정도 역시 유지됨을 확인하였다. 도 21에 나타낸 바와 같이 , 최초 인간 T세포 분리 및 활성화일 (day 0: IxlO6 T세포)이후 총 11일간의 배양 기간 동안 20배 증식된 CD3 유전자 편집 접합형 CAR T세포 (dayll: 약 2xlO7T세포)를 최종적으로 확보하였다. In order to confirm the tumor treatment effect using CD3 gene-edited conjugated human CAR T cells and anti-CD3 adapter antibodies in vivo (in r/w), it is essential to secure a sufficient number of T cells. Therefore, it is necessary to establish conditions for mass proliferation of human CAR T cells. First, CD3-positive cells were removed to increase the separation and purification efficiency of CD-negative cells. Then, in order to secure a sufficient number of CD3 gene-edited CAR T cells required for the experiment, secondary cell proliferation was induced by treating hlL-2 (300 U/ml) for 4 days after separation and purification. A schematic diagram of the established CD3 gene-edited conjugated CAR T cell proliferation conditions is shown in Fig. 19. The human CD3 gene-edited conjugated CAR T cells produced through CRISPR/Cas9 transfection and CAR transduction in the above Experimental Example 3 include some CD3-positive cells. To prevent fratricide and increase the targeting efficiency of CAR T, a process of purifying CD3-negative cells was required, and negative selection was performed using MACS (magnetic activation-associated cell sorting) using anti-human CD3 magnetic beads. Specifically, in the case of human CD3 gene-edited CAR T cells, 2 days after cell activation, CD3 protein gene-editing was performed using electroporation, and then retroviral or lentiviral transduction methods were used to produce CD3 gene-edited conjugated CAR-T cells. In the case of CD3-positive cells remaining after gene editing, only CD3-negative cells were sorted using magnetic-associated cell sorting (MACS). T cells Five days after CD3 was deleted using CRISPR/Cas9 gene editing technology, CD3 gene-edited T cells were cultured with anti-human CD3 antibodies attached to magnetic microspheres. Afterwards, cells cultured with magnetic microsphere-antibody (hCD3 microbead, Mil tenyi) were passed through a strong magnetic separator, and CD3-positive cells that were not gene-edited were attached to the magnetic microsphere-antibody and attached to the magnetic separator. On the other hand, CD3-negative cells passed through the separator without attaching to the separator and were separated. These separated CD3-negative cells were further cultured in a culture medium containing hIL-2 (200 U/ml). The expression of CAR protein on the cell surface was determined by flow cytometry (FACS-Canto II, BD Biosciences) after staining the expanded CAR T cells with Hi s-biotin and APC-labeled SA (SA-APC, BioLegend) 5 days after retroviral transduction. We confirmed that CD3-negative human CAR T cells could be effectively separated and purified by subjecting human CD3 gene-edited conjugated CAR T cells to the Ant i-CD3 MACS process, and we also confirmed whether the CD3-negative state was maintained in the T cell population after the secondary cell proliferation period. The results are shown in Figs. 20 and 21. As shown in Fig. 20, it was confirmed that most of the T cell population maintained the CD3-negative state and the level of CAR expression after the secondary cell proliferation period. As shown in Figure 21, CD3 gene-edited conjugated CAR T cells (day 1: approximately 2xlO 7 T cells) that had been expanded 20-fold over a total culture period of 11 days after the initial human T cell isolation and activation day (day 0: IxlO 6 T cells) were finally obtained.

4.2 인간 CAR T세포의 항종양능 확인 분석 (0KT3- scFV- His) (、in vivo) 항 CD3 어댑터 항체와 CD3 유전자 편집 접합형 CAR T세포를 사용하여 생체내 ( in vivo) T세포 림프종에 대한 항암효능을 검증하기 위해 , 면역결핍 NSG마우스에 luci ferase가 과발현된 인간유래의 T세포 종양인 •九»±31:세포0111厂꼬31:-匕110)를 주입하여 Xenogene i c 마우스 tumor model 실험 모델을 구죽하였다. 구체적으로, NSG마우스에 CD3양성 Jurkat- Luc세포주 ( luci f erase- transfected) 3xl06세포를 정맥 주사를 통해 주입하였다. Jurkat -Luc 주사 3일 후 , 사람 T세포로 제작한 CD3 유전자 편집 인간 CAR T세포 3xl06세포를 정맥 주사하였다. 항 CD3 어댑터 항체로는 0KT3(scFv)- Ck- Hi s를 사용하여 마우스당 20ug씩 CAR T세포 주사 4시간 전 첫 번째 항 CD3 어댑터 항체 (0KT3(scFv)- Ck- Hi s)를 투여하였고 , 이후 2일 간격으로 총 8회 투여하였다 (도 22) . 생체발광 이미지 (Bioluminescence image)는 일주일에 한번씩 IVIS 100 (PerkinElmer , Wal tham, MA USA)을 이용하여 종양세포의 크기를 측정하였다. 각각의 마우스는 식염수 100ul에 녹인 D-Luci fer in 2mg을 복강 주사하였다. 복강주사 10분 후 IVIS를 통하여 종양세포의 크기를 측정하였다. 각각 NSG마우스에 (DJurkat세포주만 주입한 군 , ②이에 더하여 항 CD3 어댑터 항체 없이 CD3 유전자 편집 접합형 CAR T세포만 주입한 군 , ③항 CD3 어댑터 항체 +CD3 유전자 편집 접합형 CAR T세포를 주입한 군으로 나누어 실험을 진행하였고 , 종양세포 주입후 60일까지의 마우스모델의 생존률을 분석하였다. 그 결과를 도 23에 나타내었다. 도 23에 나타낸 바와 같이 , CD3 유전자 편집 접합형 CAR T세포를 단독으로 투여한 경우에 비해 항 CD3 어댑터 항체를 함께 투여하는 경우 , 현저한 종양의 감소를 관찰할 수 있었고 , 마우스의 생존율 역시 크게 증가함을 확인하였다. 4.2 Analysis of anti-tumor activity of human CAR T cells (0KT3- scFV- His) (in vivo) To verify the anti-cancer efficacy against T-cell lymphoma in vivo using anti-CD3 adapter antibody and CD3 gene-edited conjugated CAR T cells, a human-derived T-cell tumor •九»±31: cell0111厂コ31:-匕110) overexpressing luciferase was injected into immunodeficient NSG mice to establish a xenogeneic mouse tumor model. Specifically, CD3-positive Jurkat-Luc cell line (luciferase-transfected) 3xl0 6 cells were intravenously injected into NSG mice. Three days after the Jurkat-Luc injection, CD3 gene-edited human CAR T cell 3xl0 6 cells produced from human T cells were intravenously injected. The first anti-CD3 adapter antibody (0KT3(scFv)- Ck-His) was administered 4 hours before CAR T cell injection at a dose of 20 ug per mouse, and then administered at 2-day intervals for a total of 8 times (Fig. 22). Bioluminescence images were acquired once a week using IVIS 100 (PerkinElmer, Waltham, MA USA) to measure the size of tumor cells. Each mouse was injected intraperitoneally with 2 mg of D-Luci fer in 100 μl of saline. The size of the tumor cells was measured via IVIS 10 minutes after the intraperitoneal injection. The NSG mice were divided into three groups: ① a group injected with only the D Jurkat cell line, ② a group injected with only CD3 gene-edited conjugated CAR T cells without anti-CD3 adapter antibody, and ③ a group injected with anti-CD3 adapter antibody + CD3 gene-edited conjugated CAR T cells, and the experiment was conducted. The survival rate of the mouse model up to 60 days after tumor cell injection was analyzed. The results are shown in Fig. 23. As shown in Fig. 23, compared to when CD3 gene-edited conjugated CAR T cells were administered alone, when anti-CD3 adapter antibody was administered together, a significant decrease in the tumor was observed, and it was confirmed that the survival rate of the mice also increased significantly.

4.3 인간 CAR T세포의 항종양능 확인 분석 (UCHT1- Fab- Hi s) ( in vivo) 상기 실험예 4. 2에서 진행한 0KT3(scFv)- Ck- Hi s어댑터 항체에 추가로, 다른 종류의 항 CD3어댑터 항체인 UCHT1- Fab- Hi s를 이용하여 생체내 항종양능을 증명하기 DNLGO , 면역결핍 NSG마우스에 인간유래의 T세포 종양인 Jurkat- Luc세포를 주입하여 Xenogene i c 마우스 종양 모델 실험 모델을 구죽하였다. 구체적으로, NSG마우스에 CD3양성 Jurkat- Luc세포주 3xl06세포를 정맥 주사하였다. Jurkat 주사 3일 후 , 사람 T세포로 제작한 CD3 유전자 편집 인간 CAR T세포 3xl06세포를 정맥 주사하였다. UCHTl-Fab-Hi s 어댑터 항체는 마우스당 20ug씩 CAR T세포 주사 4시간 전 첫 번째 항 CD3 어댑터 항체를 투여하였고 , 이후 2일 간격으로 7회 반복 투여하였다. 2주간 휴식기를 가진 후 , 다시 2일 간격으로 8회 반복 투여하여 , 항 CD3 어댑터 항체는 총 16회 투여하였다 (도 24) . 각각 NSG마우스에 ① Jurkat세포주만 주입한 군 , ② 이에 더하여 항 CD3 어댑터 항체 없이 CD3 유전자 편집 접합형 CAR T세포만 주입한 군 , ③ CD3 유전자 편집 접합형 CAR T세포 없이 항 CD3 어댑터 항체만 주입한 군 , © 항 CD3 어댑터 항체 +CD3 유전자 편집 접합형 CAR T세포를 주입한 군으로 나누어 실험을 진행하였고 , 종양세포 주입후 80일까지의 마우스모델의 생존률을 분석하였다. 그 결과를 도 25에 나타내었다. 도 25에 나타낸 바와 같이 , CD3 유전자 편집 접합형 CAR T세포를 단독으로 투여한 경우에 비해 항 CD3어댑터 항체를 함께 투여하는 경우 , 0KT3 scFv 어댑터를 사용한 실험 결과와 유사하게 현저한 종양의 감소를 관찰할 수 있었고 , 마우스의 생존율 역시 크게 증가함을 확인하였다. 이상의 결과를 통해서 , 항 CD3항체 어댑터 항체 (0KT3- scFv 및 UCHT1- Fab)를 이용한 생체내 뛰어난 항종양능을 확인함으로써 CD3접합형 CAR T세포의 T세포 종양에 대해 치료제로 제공될 수 있음을 확인하였다. 뿐만 아니라, 다양한 항 CD3항체를 이용하여 CD3접합형 CAR T세포의 어댑터 항체로 이용가능함을 확인하였다. 실험예 5. 인간접합형 CAR T세포의 면역결핍독성 (정상 T세포독성) 회피능 항인간 CD3어댑터항체 및 인간접합형 CAR T세포복합체의 정상 T세포에 대한 독성 회피능을확인하고자, 정상 T세포를표적세포로하여 정상 T세포에 대한살상능을 확인하였다. 이 때, 면역결핍 방지능은 생체내 (九 r/ra)에서 검증할 수 없으므로, 시험관내 (九 vitro) 실험을통해 확인하였다. 구체적으로, 인간 말초혈액 단핵세포 (PBMC)의 T세포를 표적세포로 이용하여 항 인간 CD3항체 및/또는 CD3유전자편집 인간접합형 CAR T세포를넣어준후, 유세포 분석을 통해 정상 T세포살상여부를분석하였고, 그 결과를 도 26에 나타내었다. 도 26에 나타낸 바와 같이, 항 인간 CD3 어댑터항체가 존재하에서만 CD3를 발현하는 T세포가 급격히 줄어들어, 이를 통해 CAR T세포의 정상 T세포 살상이 확인되고, 어댑터항체가 없는 상황에서는, CD3접합형 CAR T세포의 유무와 관계없이 CD3을 양성으로 발현하는 T세포의 수치가 유지되어, 정상 T세포를 살상하지 않음을 알 수 있다. 이를통해, 실제 생체내에서 어댑터항체의 투여 여부에 따라정상 T세포에 대한 독성을 조절하여 , 본 발명의 CD3 접합형 CAR T세포의 면역결핍독성의 부작용을 회피할 수 있음을 간접적으로 확인하였다. 실험예 6. 인간화항체를 이용한접합형 CAR- T세포의 제작 4.3 Analysis of anti-tumor activity of human CAR T cells (UCHT1- Fab- Hi s) (in vivo) In addition to the 0KT3(scFv)- Ck- Hi s adapter antibody performed in Experimental Example 4. 2, another type of anti-CD3 adapter antibody, UCHT1- Fab- Hi s, was used to demonstrate the anti-tumor activity in vivo. A xenogeneic mouse tumor model experimental model was created by injecting Jurkat-Luc cells, a T cell tumor derived from human origin, into DNLGO and immunodeficient NSG mice. Specifically, 3xl0 6 cells, a CD3-positive Jurkat-Luc cell line, were intravenously injected into NSG mice. Three days after the Jurkat injection, 3xl0 6 cells, a CD3 gene-edited human CAR T cell line produced using human T cells, were intravenously injected. The UCHTl-Fab-His adapter antibody was administered as the first anti-CD3 adapter antibody 4 hours before CAR T cell injection at 20 ug per mouse, and was then repeatedly administered 7 times at 2-day intervals. After a 2-week rest period, the anti-CD3 adapter antibody was administered 8 times at 2-day intervals again, for a total of 16 times (Fig. 24). The NSG mice were divided into ① a group injected with only the Jurkat cell line, ② a group injected with only CD3 gene-edited conjugated CAR T cells without anti-CD3 adapter antibody, ③ a group injected with only anti-CD3 adapter antibody without CD3 gene-edited conjugated CAR T cells, and © a group injected with anti-CD3 adapter antibody + CD3 gene-edited conjugated CAR T cells, and the experiment was conducted. The survival rate of the mouse models up to 80 days after tumor cell injection was analyzed. The results are shown in Fig. 25. As shown in Fig. 25, when the CD3 gene-edited conjugated CAR T cells were administered together with the anti-CD3 adapter antibody, a significant decrease in tumors was observed, similar to the results of the experiment using the 0KT3 scFv adapter, and the survival rate of the mice was also confirmed to be significantly increased. Through the above results, the excellent anti-tumor activity in vivo using the anti-CD3 antibody adapter antibody (0KT3-scFv and UCHT1-Fab) was confirmed, thereby demonstrating the efficacy of CD3-conjugated CAR T cells on T-cell tumors. It was confirmed that the anti-human CD3 adapter antibody and human conjugated CAR T cell complex can be provided as a therapeutic agent for the treatment of cancer. In addition, it was confirmed that various anti-CD3 antibodies can be used as adapter antibodies for CD3-conjugated CAR T cells. Experimental Example 5. Evasion of immunodeficiency toxicity (normal T cell toxicity) of human conjugated CAR T cells In order to confirm the evasion of toxicity against normal T cells of the anti-human CD3 adapter antibody and human conjugated CAR T cell complex, the killing ability against normal T cells was confirmed using normal T cells as target cells. At this time, since the immunodeficiency prevention ability could not be verified in vivo (九 r/ra), it was confirmed through an in vitro (九 vitro) experiment. Specifically, T cells of human peripheral blood mononuclear cells (PBMCs) were used as target cells, and after adding anti-human CD3 antibodies and/or CD3 gene-edited human conjugated CAR T cells, the killing of normal T cells was analyzed through flow cytometry, and the results are shown in Fig. 26. As shown in Fig. 26, only in the presence of anti-human CD3 adapter antibody, T cells expressing CD3 are rapidly reduced, thereby confirming the killing of normal T cells by CAR T cells, and in the absence of adapter antibody, the number of T cells positively expressing CD3 is maintained regardless of the presence or absence of CD3-conjugated CAR T cells, indicating that normal T cells are not killed. Through this, it was indirectly confirmed that the side effect of immunodeficiency toxicity of the CD3-conjugated CAR T cells of the present invention can be avoided by controlling the toxicity to normal T cells depending on the administration of adapter antibody in an actual living body. Experimental Example 6. Production of conjugated CAR-T cells using humanized antibodies

CD3유전자 편집 접합형 CAR T세포의 상용화를위하여 상기 His CAR에 사용된 마우스유래 항- 6xHis scFv (mutated 3D5)의 인간화 (humanizat ion) 및 친화도성숙 (affinity maturation)을 진행하였다. 항체 인간화 및 친화성 성숙 쥐 항- His scFv항체의 인간화및 친화성 성숙을위해 먼저, 가장적절한서열 유사성을 보이는 6개의 CDR (상보성 결정 영역)을 IGHV1-18 및 IGKV2-28 인간 생식선 유전자에 접목시켰다. 동시에, 번역 후 변형을 제거하고 역변이를 수행하기 위해 프레임워크 영역 (FR)의 여러 아미노산 부위를 변경하였다. 그 후, 친화성 성숙을 위해 6개의 CDR에서 부위 지향돌연변이를포함하는인간화유內의 하위 라이브러리를 구축하였다. Exp i293 발현 시스템을 사용하여 항체를 생산하고 친화성을 정제하여 항원과의 결합 친화성을 확인하기 위해 ELISA를수행하였다 (WuXi Biologies). 인간화 항체의 SPR결합 친화도측정 For commercialization of CD3 gene-edited conjugated CAR T cells, humanization and affinity maturation of the mouse-derived anti-6xHis scFv (mutated 3D5) used in the His CAR were performed. Antibody humanization and affinity maturation For humanization and affinity maturation of the mouse anti-His scFv antibody, first, six CDRs (complementarity determining regions) showing the most appropriate sequence similarity were grafted onto the IGHV1-18 and IGKV2-28 human germline genes. At the same time, several amino acid sites in the framework region (FR) were changed to remove post-translational modifications and perform reverse mutations. Subsequently, a sub-library of humanized antibodies containing site-directed mutations in the six CDRs was generated for affinity maturation. The antibody was produced using the Exp i293 expression system, affinity purified, and ELISA was performed to confirm the binding affinity to the antigen (WuXi Biologies). SPR binding affinity measurement of humanized antibodies

CM5센서 칩이 장착된 Biacore 8K SPR시스템 (사이터바)을사용하여 결합동력 속도 및 친화도 상수를 생성하였다. 항- His 항체의 포획 표면을 준비하기 위해 스트렙타비딘을 고정시킨 다음 비오틴 접합 6xHis 펩타이드를 포획하였다. 항체를 1배 PBS로 2배 직렬 희석한다음 30uL/min의 유속으로 3분동안주입하여 10분동안 해리 단계를 거쳤다. 표면은 10mM글리신, pH 1.5를사용하여 재생되었다. 동역학은 Biacore 8K평가소프트웨어 버전 l.O(Cytiva)을사용하여 분석하였다. 도 27은 인간화 항체 항 His 유內의 항원 친화도를 나타낸 것으로, 최종 인간화 항체 scFv는 기존 scFv에 비해 약 5.8배 높은 항원 친화도를 보임으로써 성공적인 인간화가증명되었다. 이어서 , 인간화항- 6xHis scFv를이용하여 기존 His CAR와유사한 CAR유전자를 렌티바이러스 벡터에 클로닝하여 렌티바이러스를 생산한후, 인간 T세포에 형질도입 (transduct ion)하여 인간화 His CAR- T세포 (huHis CAR- T세포)를 제작하였다. HuHis CAR- T세포 표면의 CAR발현은 유세포분석으로 확인하였고 그 결과를 도 28에 나타내었다. 제작된 huHis CAR- T세포와 UCHTl-Fab-His 어댑터를 사용하여 접합형 CAR T세포의 in vitro 활성정도를 검증한 결과, 기존의 His CAR-세포와동일한 양상으로 huHis CAR- T세포 또한 어댑터 항체가 있는 상황에서 T세포의 활성화 및 종양에 대한 세포 살상능을 보임이 확인되었다 (도 29). 따라서, 임상적용이 가능한 접합형 인간화 CAR- T세포 제작이 가능함이 확인됨으로써 , 본 발명의 상용화가능성이 증명되었다. The binding kinetics and affinity constants were generated using a Biacore 8K SPR system (Cytiva) equipped with a CM5 sensor chip. To prepare the capture surface of anti-His antibody, streptavidin was immobilized and then biotin-conjugated 6xHis peptide was captured. The antibody was serially diluted 2-fold with 1x PBS and injected at a flow rate of 30 μL/min for 3 min followed by a 10-min dissociation step. The surface was regenerated using 10 mM glycine, pH 1.5. The kinetics were analyzed using Biacore 8K Evaluation Software Version 1.0 (Cytiva). Figure 27 shows the antigen affinity of the humanized antibody anti-His, and the final humanized antibody scFv showed an antigen affinity approximately 5.8-fold higher than that of the original scFv, demonstrating successful humanization. Next, using the humanized anti-6xHis scFv, a CAR gene similar to the existing His CAR was cloned into a lentiviral vector to produce a lentivirus, which was then transduced into human T cells to produce humanized His CAR- T cells (huHis CAR- T cells). The expression of the CAR on the surface of the huHis CAR- T cells was confirmed by flow cytometry, and the results are shown in Fig. 28. The in vitro activity of the conjugated CAR T cells using the produced huHis CAR- T cells and the UCHTl-Fab-His adapter was verified, and it was confirmed that the huHis CAR- T cells also showed T cell activation and tumor cell killing ability in the presence of the adapter antibody, similar to the existing His CAR- cells (Fig. 29). Therefore, by confirming that it is possible to produce a conjugated humanized CAR- T cell that can be applied clinically, the commercialization possibility of the present invention was proven.

【표 13]

Figure imgf000040_0001
Figure imgf000041_0001
이상의 결과를 통해 , 항 CD3 어댑터 항체와 CD3편집 접합형 CAR T세포의 복합투여가 T세포 종양에 대한 강력한 치료효과를 달성할 수 있을 뿐 아니라, 접합형이 CAR T를 구현하여 , 정상 T세포독성에 의한 면역결핍부작용의 회피 개연성을 입증하였다. 전술한 본 발명의 설명은 예시를 위한 것이며 , 본 발명이 속하는 기술분야의 통상의 지식을 가진 자는 본 발명의 기술적 사상이나 필수적인 특징을 변경하지 않고서 다른 구체적인 형태로 쉽게 변형이 가능하다는 것을 이해할 수 있을 것이다. 그러므로 이상에서 기술한 실시 예들은 모든 면에서 예시적인 것이며 한정적이 아닌 것으로 이해해야만 한다. 【Table 13]
Figure imgf000040_0001
Figure imgf000041_0001
The results above demonstrate that combined administration of anti-CD3 adapter antibodies and CD3-edited conjugated CAR T cells can not only achieve a powerful therapeutic effect on T-cell tumors, but also demonstrate the possibility of avoiding immunodeficiency side effects caused by normal T-cell cytotoxicity by implementing conjugated CAR T. The above description of the present invention is for the purpose of illustration, and those skilled in the art to which the present invention pertains will understand that the present invention can be easily modified in other specific forms without changing the technical spirit or essential characteristics of the present invention. Therefore, it should be understood that the embodiments described above are exemplary in all aspects and not limiting.

Claims

【청구범위】 【Scope of Claims】 【청구항 11 【Claim 11 CD3의 발현이 감소되거나제거된 키메라 항원 수용체 T세포 (CAR- T세포) 및 상기 CAR-T 세포의 활성을 조절하는 어댑터 분자를 포함하는, T세포 유래 종양을 예방 또는 치료하기 위한 약학조성물로서 , 상기 어댑터 분자는 (a)표적세포상의 CD3에 결합하는제 1모이어티 (moiety) 및 (b) 키메라 항원 수용체 (Chimeric antigen receptor)의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를 포함하며 , 상기 CAR-T 세포는 (i) 상기 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편을 포함하는 세포외 도메인, (ii) 막횡단 도메인, 및 (iii) 세포내 신호전달도메인을포함하는, 키메라항원수용체 (Chimeric antigen receptor)를 발현하는 것인, 약학조성물. A pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated and an adapter molecule that regulates the activity of the CAR-T cell, wherein the adapter molecule comprises (a) a first moiety that binds to CD3 on a target cell and (b) a second moiety that specifically binds to an extracellular domain of a chimeric antigen receptor, and wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to the second moiety of the adapter molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain. 【청구항 2] 【Claim 2] CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T(CAR T) 세포를 포함하는, T세포 유래 종양을 예방또는 치료하기 위한 약학조성물로서, 상기 약학조성물은 CAR-T세포의 활성을조절할 수 있는어댑터 분자와병용 투여되며 , 상기 어댑터 분자는 (a) 표적세포상의 CD3에 결합하는제 1모이어티 (moiety) 및 (b) 상기 키메라 항원 수용체의 세포외 도메인에 특이적으로 결합하는 제 2 모이어티를 포함하며 , 상기 키메라 항원 수용체는 상기 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합단편을 포함하는 세포외 도메인, 막횡단도메인 및 세포내 신호전달도메인을 것인, 약학조성물. A pharmaceutical composition for preventing or treating a T cell-derived tumor, comprising a chimeric antigen receptor T (CAR T) cell in which CD3 expression is reduced or eliminated, wherein the pharmaceutical composition is administered in combination with an adaptor molecule capable of modulating the activity of the CAR-T cell, wherein the adaptor molecule comprises (a) a first moiety that binds to CD3 on a target cell and (b) a second moiety that specifically binds to an extracellular domain of the chimeric antigen receptor, and wherein the chimeric antigen receptor has an extracellular domain, a transmembrane domain, and an intracellular signaling domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to the second moiety of the adaptor molecule. 【청구항 3] 청구항 1 또는 청구항 2에 있어서, 상기 제 1 모이어티는 항체, 항원 결합 단편 , 애피바디 (aff ibody) , 다이아바디 (diabody) 및 앱타머 (aptamer )로 이루어진 군으로부터 선택되는 것인, 약학조성물. 【Claim 3】 A pharmaceutical composition according to claim 1 or 2, wherein the first moiety is selected from the group consisting of an antibody, an antigen-binding fragment, an affibody, a diabody, and an aptamer. 【청구항 4] 청구항 1 내지 청구항 3 중 어느 한 항에 있어서, 상기 항원 결합 단편은 Fab, Fab ' , F(ab' )2, Fv, scFv 또는 싱글도메인 항체 (single- domain antibody: sdAB)인, 약학조성물. 【Claim 4】 A pharmaceutical composition according to any one of claims 1 to 3, wherein the antigen-binding fragment is Fab, Fab', F(ab')2, Fv, scFv or a single-domain antibody (sdAB). 【청구항 5] 청구항 1 내지 청구항 4 중 어느 한 항에 있어서, 상기 제 2 모이어티는 펩타이드 태그 또는 합텐 태그인, 약학조성물. 【Claim 5】 A pharmaceutical composition according to any one of claims 1 to 4, wherein the second moiety is a peptide tag or a hapten tag. 【청구항 6] 청구항 1내지 청구항 5중 어느한 항에 있어서 , 상기 태그는 His 태그, Myc 태그, 코티닌 태그, FITC태그, 비오틴 태그, Leucin zipper 태그, Flag태그, Xpress 태그, Avi 태그, 칼모듈린 결합 펩타이드 (CBP) 태그, 폴리글루타메이트 태그, HA 태그, Strep 태그, Softag 1, Softag 3, 및 V5 태그로 이루어진 군으로부터 선택되는 것인, 약학조성물. 【Claim 6] A pharmaceutical composition according to any one of claims 1 to 5, wherein the tag is selected from the group consisting of a His tag, a Myc tag, a cotinine tag, a FITC tag, a biotin tag, a Leucin zipper tag, a Flag tag, an Xpress tag, an Avi tag, a calmodulin binding peptide (CBP) tag, a polyglutamate tag, an HA tag, a Strep tag, Softag 1, Softag 3, and a V5 tag. 【청구항 7] 청구항 1내지 청구항 6중 어느한 항에 있어서, 상기 제 1모이어티는항 CD3 항체 또는 이의 항원 결합단편이거나/이고, 상기 제 2모이어티는 His 태그인, 약학 조성물. 【Claim 7】 A pharmaceutical composition according to any one of claims 1 to 6, wherein the first moiety is an anti-CD3 antibody or an antigen-binding fragment thereof, and/or the second moiety is a His tag. 【청구항 8] 청구항 1내지 청구항 7 종 어느한항에 있어서, 상기 제 1모이어티는 0KT3, UCHT1 , 서열번호 23의 아미노산 서열으로 구성된 CDR1, 서열번호 24의 아미노산 서열으로 구성된 CDR2, 및 서열번호 25의 아미노산서열으로구성된 CDR3을포함하는 경쇄 가변 영역 및 서열번호 26의 아미노산 서열으로 구성된 CDR1, 서열번호 27의 아미노산 서열으로 구성된 CDR2, 및 서열번호 28의 아미노산서열으로구성된 CDR3을 포함하는 중쇄 가변 영역을포함하는항체; 서열번호 32의 아미노산서열으로구성된 CDR1, 서열번호 33의 아미노산 서열으로 구성된 CDR2, 및 서열번호 34의 아미노산 서열으로 구성된 CDR3을 포함하는 경쇄 가변 영역 및 서열번호 35의 아미노산 서열으로 구성된 CDR1, 서열번호 36의 아미노산서열으로구성된 CDR2, 및 서열번호 37의 아미노산서열으로구성된 CDR3을 포함하는중쇄 가변 영역 ; 및 서열번호 67의 아미노산 서열으로 구성된 CDR1, 서열번호 68의 아미노산서열으로구성된 CDR2, 및 서열번호 69의 아미노산 서열으로 구성된 CDR3을 포함하는 경쇄 가변 영역 및 서열번호 70의 아미노산 서열으로 구성된 CDR1, 서열번호 기의 아미노산 서열으로 구성된 CDR2, 및 서열번호 72의 아미노산 서열으로 구성된 CDR3을 포함하는 중쇄 가변 영역을 포함하는 항체로 이루어진 군으로부터 선택되고, 상기 제 2 모이어티는 His 태그인, 약학조성물. 【Claim 8】 An antibody according to any one of claims 1 to 7, wherein the first moiety comprises a light chain variable region comprising 0KT3, UCHT1, a CDR1 consisting of the amino acid sequence of SEQ ID NO: 23, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 24, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 25, and a heavy chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 26, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 27, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 28; A pharmaceutical composition selected from the group consisting of an antibody comprising a light chain variable region comprising CDR1 consisting of the amino acid sequence of SEQ ID NO: 32, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 33, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 34 and a heavy chain variable region comprising CDR1 consisting of the amino acid sequence of SEQ ID NO: 35, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 36, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 37; and a light chain variable region comprising CDR1 consisting of the amino acid sequence of SEQ ID NO: 67, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 68, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 69 and a heavy chain variable region comprising CDR1 consisting of the amino acid sequence of SEQ ID NO: 70, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 70, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 72, wherein the second moiety is a His tag. 【청구항이 청구항 1 내지 청구항 8 종 어느 한 항에 있어서 , CAR-T세포의 CD3 발현의 감소나 제거는 CAR-T세포의 CD3 또는 T cell receptor (TCR)를 제거하는 것에 의해 이루어지는 것인, 약학조성물. 【A pharmaceutical composition according to any one of claims 1 to 8, wherein the reduction or elimination of CD3 expression of CAR-T cells is achieved by removing CD3 or T cell receptor (TCR) of CAR-T cells. 【청구항 10】 청구항 1 내지 청구항 9중 어느 한 항에 있어서 , T세포 유래 종양은 T세포 전림프구성 백혈병 (T- cell prolymphocyt ic leukemia) , T세포 거대과립 림프구성 백혈병 (T- cell large granular lymphocytic leukemia) , NK세포의 만성 림프증식성 질환 (chronic lymphoprol i ferat ive disorder of NK cells), 공격성 NK 백혈병 (aggressive NK leukemia) , 소아 전신 EBV 양성 T세포 림프증식성 질환 (systemic EBV-positive T-cel 1 lymphoprol i ferat ive disease of chi Idhood) , 하이드로아 백신 유사 림프종 (hydroa vacc i n i f orme- 1 i ke lymphoma) , 성인 T세포 백혈병/림프종 (adult T-cel 1 leukemia/ lymphoma) , 림프절 외 NK/T세포 림프종 (비강 형 ) (extranodal NK/T-cel 1 lympho (nasal type)) , 장병증 관련 T세포 림프종 (enteropathy- associated T-cel 1 lymphoma) , 간비장 T세포 림프종 (hepatosplenic T-cel 1 lymphoma) , 피하지방중염 유사 T세포 림프종 (subcutaneous panniculitis-like T-cel 1 lymphoma) , 균상 식육종 (mycosis fungoides) , 세자리 증후군 (Se'zary syndrome) , 원발성 피부 CD30 양성 T-세포 림프증식성 질환 (primary cutaneous CD30- posit ive T-cel 1 lymphoprol i ferat ive disorders) , 림프종모양구진증 ( lymphomatoid papulosis) , 원발성 피부 역형성 대세포 림프종 (primary cutaneous anaplastic large cel 1 lymphoma) , 원발성 피부감마/델타 T세포 림프종 (primary cutaneous gamma/delta T-cel 1 lymphoma) , 원발성 피부 CDS 양성 공격성 표피친화성 세포독성 T세포 림프종 (primary cutaneous CD8- positive aggressive epidermotropic cytotoxic T-cel 1 lymphoma) , 원발성 피부 CD4양성 소/중 T세포 림프종 (primary cutaneous CD4— positive smal 1/medium T-cel 1 lymphoma) , 말초 T세포 림프종 NOS (달리 명시되지 않음) (per ipheral T-cel 1 lymphoma N0S(not otherwise specified)) , 혈관면역모구 T세포 림프종 (angio immunob last ic T-cel 1 lymphoma) , ALK 양성 역형성 대세포 림프종 (ALK positive anaplastic large cel 1 lymphoma) 및 ALK음성 역형성 대세포 림프종 (ALK negat ive anaplastic large cel 1 lymphoma)로구성된 군으로부터 선택되는, 약학조성물. 【Claim 10】 The T-cell derived tumor of any one of claims 1 to 9 is T-cell prolymphocytic leukemia, T-cell large granular lymphocytic leukemia, chronic lymphoprol i ferat ive disorder of NK cells, aggressive NK Leukemia (aggressive NK leukemia), systemic EBV-positive T-cell 1 lymphoprol i ferat ive disease of chi dhood, hydroa vacc inif orme- 1 i ke lymphoma, adult T-cell leukemia/lymphoma, extranodal NK/T-cell 1 lympho oma (nasal type), enteropathy-associated T-cell 1 lymphoma, hepatosplenic T-cell 1 lymphoma, subcutaneous panniculitis-like T-cell 1 lymphoma, mycosis fungoides fungoides, Se'zary syndrome, primary cutaneous CD30-positive T-cell lymphoproliferative disorders, lymphomatoid papulosis, primary cutaneous anaplastic large cel 1 lymphoma, primary cutaneous gamma/delta T-cell lymphoma, primary cutaneous CD8-positive aggressive epidermotropic cytotoxic T-cel 1 lymphoma, primary cutaneous CD4-positive small/medium T-cell 1 lymphoma, peripheral T-cell lymphoma NOS (not otherwise specified) A pharmaceutical composition selected from the group consisting of per ipheral T-cel 1 lymphoma N0S (not otherwise specified), angioimmunoblastic T-cel 1 lymphoma, ALK positive anaplastic large cel 1 lymphoma and ALK negative anaplastic large cel 1 lymphoma. 【청구항 11】 【Claim 11】 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 (Chimeric antigen receptor) T세포 (CAR- T세포) 및 상기 CAR-T세포의 활성을조절하는 어댑터 분자를 포함하는 복합체로서 , 상기 어댑터 분자는 (a)표적세포상의 CD3에 결합하는제 1모이어티 (moiety) 및 (b) 키메라 항원 수용체 (Chimeric antigen receptor)의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를 포함하며 , 상기 CAR-T 세포는 (i) 상기 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편을 포함하는 세포외 도메인, (ii) 막횡단 도메인, 및 (iii) 세포내 신호전달 도메인을 포함하는, 키메라 항원 수용체를 발현하는 것인, 복합체. A complex comprising a chimeric antigen receptor (CAR-T cell) with reduced or deleted expression of CD3 and an adapter molecule that regulates the activity of the CAR-T cell, wherein the adapter molecule comprises (a) a first moiety that binds to CD3 on a target cell and (b) a second moiety that specifically binds to an extracellular domain of the chimeric antigen receptor, and wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to the second moiety of the adapter molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain. 【청구항 12】 청구항 1 내지 청구항 10 중 어느 한 항에 따른 약학 조성물 또는 청구항 11의 복합체를 개체에 투여하는 것을 포함하는, T세포 유래 종양을 예방 또는 치료하는 방법 . 【Claim 12】 A method for preventing or treating a T cell-derived tumor, comprising administering to a subject a pharmaceutical composition according to any one of claims 1 to 10 or a complex according to claim 11. 【청구항 13】 【Claim 13】 CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T 세포 (CAR- T 세포) 로서, 상기 CAR-T 세포는 (i) 어댑터 분자의 제 2 모이어티에 특이적으로 결합하는 항체 또는 이의 항원 결합단편을 포함하는 세포외 도메인, (ii) 막횡단도메인, 및 (iii) 세포내 신호전달도메인을 포함하는, 키메라 항원 수용체를 발현하고, 이때 상기 어댑터 분자는 (a) 표적세포 상의 CD3에 결합하는 제 1 모이어티 (moiety) 및 (b) 상기 키메라 항원 수용체의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를 포함하는 것인, CAR-T세포. A chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the CAR-T cell expresses a chimeric antigen receptor comprising (i) an extracellular domain comprising an antibody or an antigen-binding fragment thereof that specifically binds to a second moiety of an adaptor molecule, (ii) a transmembrane domain, and (iii) an intracellular signaling domain, wherein the adaptor molecule comprises (a) a first moiety that binds CD3 on a target cell and (b) a second moiety that specifically binds to the extracellular domain of the chimeric antigen receptor. 【청구항 14】 청구항 13에 있어서, 상기 제 1 모이어티는 항 CD3 항체 또는 이의 항원 결합 단편이거나/이고 상기 제 2모이어티는 His 태그인 것인, CAR-T세포. 【Claim 14】 A CAR-T cell according to claim 13, wherein the first moiety is an anti-CD3 antibody or an antigen-binding fragment thereof and/or the second moiety is a His tag. 【청구항 15】 청구항 13 또는 청구항 14에 있어서, 상기 제 2 모이어티는 His 태그이고, 상기 His 태그에 특이적으로 결합하는 항체 또는 이의 항원 결합 단편은 서열번호 63의 아미노산서열을 갖는 것인, CAR-T세포. 【Claim 15】 A CAR-T cell according to claim 13 or 14, wherein the second moiety is a His tag, and the antibody or antigen-binding fragment thereof that specifically binds to the His tag has an amino acid sequence of SEQ ID NO: 63. 【청구항 16】 청구항 13 내지 청구항 15중 어느 한 항에 있어서 , 서열번호 63 내지 65의 아미노산 서열을 갖는 CAR-T세포. 【Claim 16】 A CAR-T cell having an amino acid sequence of SEQ ID NOs: 63 to 65 according to any one of claims 13 to 15. 【청구항 17】 【Claim 17】 (a) T세포에서 CD3 또는 T세포수용체 (TCR)의 발현을 하향 조절하는 단계 , 및 (a) a step of down-regulating the expression of CD3 or T cell receptor (TCR) in T cells, and (b) 어댑터 분자의 제 2모이어티에 특이적으로결합하는항체 또는 이의 항원 결합 단편을 포함하는 세포외 도메인, 막횡단도메인, 및 세포내 신호전달도메인을 포함하는 키메릭 항원 수용체 (CAR)를 T세포에 도입하는 단계를 포함하며 , 상기 (a) 및 (b) 단계는 순서와 상관없이 수행되는, CD3의 발현이 감소되거나 제거된 키메라 항원 수용체 T세포 (CAR- T세포)를 제조하는 방법으로서 , 이때. 상기 어댑터 분자는 (i) 표적세포 상의 CD3에 결합하는 제 1 모이어티 (moiety) 및 (ii) 상기 키메라 항원 수용체의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를 포함하는 것인, 방법. (b) a step of introducing a chimeric antigen receptor (CAR) comprising an extracellular domain, a transmembrane domain, and an intracellular signaling domain, wherein the second moiety of an adapter molecule comprises an antibody or an antigen-binding fragment thereof that specifically binds to an adapter molecule, wherein steps (a) and (b) are performed in any order, a method for producing a chimeric antigen receptor T cell (CAR-T cell) in which CD3 expression is reduced or eliminated, wherein the adapter molecule comprises (i) a first moiety that binds to CD3 on a target cell and (ii) a second moiety that specifically binds to an extracellular domain of the chimeric antigen receptor. 【청구항 18】 청구항 17에 있어서, 상기 (a) T세포에서 CD3 또는 T 세포 수용체 (TCR)의 발현을 하향 조절하는 단계는 유전자편집 (genome editing) 기법 , shRNA , siRNA 또는 miRNA에 의해 수행되는, 방법 【Claim 18】 In claim 17, the step of down-regulating the expression of CD3 or T cell receptor (TCR) in the (a) T cell is performed by a genome editing technique, shRNA, siRNA or miRNA. 【청구항 1이 청구항 17 또는 청구항 18에 있어서 , 유전자편집 (genome editing)기법은 CRISPR(clustered regular ly interspaced short pal indromic repeats)/Cas 9, TALEN , zinc f inger nuclease, base-editing, prime— editing에 의해 CAR-T세포의 CD3 또는 T cell receptor (TCR)를제거하는 것인, 방법. 【Claim 1】 A method according to claim 17 or 18, wherein the genome editing technique removes CD3 or T cell receptor (TCR) of CAR-T cells by CRISPR (clustered regularly interspaced short palindromic repeats)/Cas 9, TALEN, zinc finger nuclease, base-editing, or prime— editing. 【청구항 20] 청구항 17내지 청구항 19중어느한 항에 있어서 , 서열번호 57혹은서열번호 58의 폴리뉴클레오티드 서열을 포함하는 가이드 오때를 사용하는 CRISPR/Cas9기법에 의해 CAR-T세포의 CD3 또는 T cell receptor (TCR)를 제거하는, 방법. 【Claim 20】 A method for removing CD3 or T cell receptor (TCR) of CAR-T cells by CRISPR/Cas9 technique using a guide gene comprising a polynucleotide sequence of SEQ ID NO: 57 or SEQ ID NO: 58, according to any one of claims 17 to 19. 【청구항 21] 【Claim 21] (a) 표적세포 상의 CD3에 결합하는 제 1 모이어티; 및 (a) a first moiety that binds to CD3 on a target cell; and (b) 키메라 항원 수용체 (Chimeric antigen receptor)의 세포외 도메인에 특이적으로 결합하는 제 2모이어티를 포함하는, 키메라 항원수용체 T세포 (CAR- T세포)의 활성을조절하기 위한어댑터 분자. (b) An adaptor molecule for regulating the activity of a chimeric antigen receptor T cell (CAR-T cell), comprising a second moiety that specifically binds to the extracellular domain of a chimeric antigen receptor. 【청구항 22] 청구항 21에 있어서, 상기 제 1 모이어티는 항체, 항원 결합 단편, 애피바디 (aff ibody) , 다이아바디 (diabody) 및 앱타머 (aptamer )로 이루어진 군으로부터 선택되는, 어댑터 분자. 【Claim 22】 An adapter molecule according to claim 21, wherein the first moiety is selected from the group consisting of an antibody, an antigen-binding fragment, an affibody, a diabody, and an aptamer. 【청구항 23] 청구항 21 또는 청구항 22에 있어서, 상기 항원 결합 단편은 Fab, Fab' , F(ab' )2, Fv, scFv 또는 싱글도메인 항체 (single- domain antibody: sdAB)인 , 어댑터 분자. 【Claim 23】 An adapter molecule according to claim 21 or claim 22, wherein the antigen-binding fragment is Fab, Fab', F(ab')2, Fv, scFv or a single-domain antibody (sdAB). 【청구항 24】 청구항 21 내지 청구항 23 중 어느 한 항에 있어서, 상기 제 2 모이어티는 태그, 폴리펩타이드 또는 합텐인, 어댑터 분자. 【Claim 24】 An adapter molecule according to any one of claims 21 to 23, wherein the second moiety is a tag, a polypeptide or a hapten. 【청구항 25] 청구항 21 내지 청구항 24중 어느 한 항에 있어서 , 상기 태그는 His 태그, Myc 태그, 코티닌 태그, FITC 태그, Biotin 태그, Leucin zipper 태그, Flag 태그, Xpress태그, Avi 태그, 칼모듈린결합펩타이드 (CBP) 태그, 폴리글루타메이트태그, HA 태그, Strep 태그, Softag 1, Softag 3, 및 V5 태그로 이루어진 군으로부터 선택되는 것인, 어댑터 분자. 【Claim 25】 An adapter molecule according to any one of claims 21 to 24, wherein the tag is selected from the group consisting of a His tag, a Myc tag, a cotinine tag, a FITC tag, a Biotin tag, a Leucin zipper tag, a Flag tag, an Xpress tag, an Avi tag, a calmodulin binding peptide (CBP) tag, a polyglutamate tag, an HA tag, a Strep tag, Softag 1, Softag 3, and a V5 tag. 【청구항 26] 청구항 21 내지 청구항 25 중 어느 한 항에 있어서, 상기 제 1 모이어티는 항 CD3항체 또는 이의 항원 결합단편이거나/이고, 상기 제 2모이어티는 His 태그인, 어댑터 분자. 【Claim 26] An adapter molecule according to any one of claims 21 to 25, wherein the first moiety is an anti-CD3 antibody or an antigen-binding fragment thereof, and/or the second moiety is a His tag. 【청구항 27] 청구항 21 내지 청구항 26 중 어느 한 항에 있어서, 상기 제 1 모이어티는 0KT3, UCHT1 , 서열번호 23의 아미노산서열으로구성된 CDR1, 서열번호 24의 아미노산 서열으로 구성된 CDR2, 및 서열번호 25의 아미노산서열으로구성된 CDR3을포함하는 경쇄 가변 영역 및 서열번호 26의 아미노산 서열으로 구성된 CDR1, 서열번호 27의 아미노산 서열으로 구성된 CDR2, 및 서열번호 28의 아미노산서열으로구성된 CDR3을 포함하는 중쇄 가변 영역을포함하는항체; 서열번호 32의 아미노산서열으로구성된 CDR1, 서열번호 33의 아미노산 서열으로 구성된 CDR2, 및 서열번호 34의 아미노산 서열으로 구성된 CDR3을 포함하는 경쇄 가변 영역 및 서열번호 35의 아미노산 서열으로 구성된 CDR1, 서열번호 36의 아미노산서열으로구성된 CDR2, 및 서열번호 37의 아미노산 서열으로 구성된 CDR3을 포함하는 중쇄 가변 영역을 포함하는 항체; 및 서열번호 67의 아미노산서열으로구성된 CDR1, 서열번호 68의 아미노산서열으로 구성된 CDR2, 및 서열번호 69의 아미노산 서열으로 구성된 CDR3을 포함하는 경쇄 가변 영역 및서열번호 70의 아미노산서열으로구성된 CDR1, 서열번호기의 아미노산 서열으로 구성된 CDR2, 및 서열번호 72의 아미노산서열으로구성된 CDR3을포함하는 중쇄 가변 영역을 포함하는 항체로 이루어진 군으로부터 선택되고, 상기 제 2 모이어티는 His 태그인, 어댑터 분자. 【Claim 27】 An antibody according to any one of claims 21 to 26, wherein the first moiety comprises a light chain variable region comprising 0KT3, UCHT1, a CDR1 consisting of the amino acid sequence of SEQ ID NO: 23, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 24, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 25, and a heavy chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 26, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 27, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 28; An adapter molecule selected from the group consisting of an antibody comprising a light chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 32, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 33, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 34, and a heavy chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 35, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 36, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 37; and an antibody comprising a light chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 67, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 68, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 69, and a heavy chain variable region comprising a CDR1 consisting of the amino acid sequence of SEQ ID NO: 70, a CDR2 consisting of the amino acid sequence of SEQ ID NO: 70, and a CDR3 consisting of the amino acid sequence of SEQ ID NO: 72, wherein the second moiety is a His tag.
PCT/IB2024/054913 2023-05-19 2024-05-20 Cd3-targeting switchable car-t cells of which cd3 gene is edited Pending WO2024241208A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020257015183A KR20250088543A (en) 2023-05-19 2024-05-20 CD3 gene-edited CD3-targeted switchable CAR-T cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR20230065321 2023-05-19
KR10-2023-0065321 2023-05-19

Publications (1)

Publication Number Publication Date
WO2024241208A1 true WO2024241208A1 (en) 2024-11-28

Family

ID=93589831

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2024/054913 Pending WO2024241208A1 (en) 2023-05-19 2024-05-20 Cd3-targeting switchable car-t cells of which cd3 gene is edited

Country Status (2)

Country Link
KR (1) KR20250088543A (en)
WO (1) WO2024241208A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019158942A1 (en) * 2018-02-16 2019-08-22 Crescendo Biologics Limited Therapeutic molecules that bind to lag3 and pd1
KR20210016431A (en) * 2018-05-31 2021-02-15 워싱턴 유니버시티 Chimeric antigen receptor T cells for cancer treatment (CAR-T)
KR20220008841A (en) * 2019-05-13 2022-01-21 락바디 테라퓨틱스 리미티드 Activatable bispecific antibody comprising a linker between two binding domains, which is a human immunoglobulin hinge region, or a variant thereof, and uses thereof
KR20220132577A (en) * 2020-01-24 2022-09-30 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 Quantitative control of the activity of engineered cells expressing universal immune receptors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11478555B2 (en) 2015-08-17 2022-10-25 Seoul National University R&Db Foundation Chimeric antigen receptor to which anti-cotinine antibody is linked, and use thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019158942A1 (en) * 2018-02-16 2019-08-22 Crescendo Biologics Limited Therapeutic molecules that bind to lag3 and pd1
KR20210016431A (en) * 2018-05-31 2021-02-15 워싱턴 유니버시티 Chimeric antigen receptor T cells for cancer treatment (CAR-T)
KR20220008841A (en) * 2019-05-13 2022-01-21 락바디 테라퓨틱스 리미티드 Activatable bispecific antibody comprising a linker between two binding domains, which is a human immunoglobulin hinge region, or a variant thereof, and uses thereof
KR20220132577A (en) * 2020-01-24 2022-09-30 더 트러스티스 오브 더 유니버시티 오브 펜실베니아 Quantitative control of the activity of engineered cells expressing universal immune receptors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE GenPept 17 October 2024 (2024-10-17), ANONYMOUS: "Chain L, Anti-his tag antibody 3d5 variable light chain, Peptide linke ", XP093243083, Database accession no. PDB: 1KTR_L. *
FUJIWARA KENTO, KITAURA MASAKI, TSUNEI AYAKA, KUSABUKA HOTAKA, OGAKI ERIKA, OKADA NAOKI: "Structure of the Signal Transduction Domain in Second-Generation CAR Regulates the Input Efficiency of CAR Signals", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, MOLECULAR DIVERSITY PRESERVATION INTERNATIONAL (MDPI), BASEL, CH, vol. 22, no. 5, Basel, CH , pages 2476, XP093243075, ISSN: 1422-0067, DOI: 10.3390/ijms22052476 *

Also Published As

Publication number Publication date
KR20250088543A (en) 2025-06-17

Similar Documents

Publication Publication Date Title
JP7660382B2 (en) Immune cells expressing a universal chimeric antigen receptor for targeting multiple diverse antigens, a method for producing said immune cells, and use of said immune cells for the treatment of cancer, infectious diseases and autoimmune diseases
US12103973B2 (en) Anti-B7-H3 monoclonal antibody and use thereof in cell therapy
EP3715373A1 (en) Novel scfv amino acid sequence, chimeric antigen receptor comprising same and application thereof
TW201900672A (en) Improved antigen binding receptor type
US20140322216A1 (en) Glypican-3-specific antibody and uses thereof
JP2021536266A (en) Chimeric antigen receptor for solid cancer and T cells expressing chimeric antigen receptor
CA3147834A1 (en) Chimeric antigen receptor and immune effector cell expressing chimeric antigen receptor
TR201904484T4 (en) Truncated epidermal growth factor receptor (EGFRt) for transduced T cell selection.
US20230357385A1 (en) Anti-gpc3 antibody, anti-gpc3 chimeric antigen receptor and gpc3/cd3 bispecific antibody
US20250041416A1 (en) P329g antibody targeting bcma, combination of same with chimeric antigen receptor cell, and use thereof
WO2023072307A1 (en) Antigen binding fragment targeting cd70, single-chain antibody and chimeric antigen receptor, and use thereof
TW202237638A (en) Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
JP2020530766A (en) Antigen binding region for fibronectin type III domain and its usage
US20240018262A1 (en) Antibody specific for gpc3 and uses thereof
CN114258402B (en) Anti-CD123 antibodies, anti-CD123 chimeric antigen receptors, and anti-CD123 chimeric antigen receptor T cells
WO2024241208A1 (en) Cd3-targeting switchable car-t cells of which cd3 gene is edited
US20250144214A1 (en) Cd28 hinge and transmembrane containing chimeric antigen receptors targeting gpc2 and use thereof
JP7531603B2 (en) CD28H domain-containing chimeric antigen receptors and methods of use
TW202323295A (en) Process for producing armed immune cells
WO2022020945A1 (en) Anti-cd22 single domain antibodies and therapeutic constructs
EP4620972A1 (en) Dual gene-transduced immune cell using ctla-4 variant and chimeric antigen receptor and use thereof
US20250381225A1 (en) Recombinant antibodies, chimeric antigen receptors, and uses thereof in treating cancers
EP4257608A1 (en) Antibody specific for cd47 and use thereof
KR20250117586A (en) Combination therapy of bispecific antibody and car-t
CN118184798A (en) Double-target chimeric antigen receptor targeting CD19 and CD81 and application 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: 24810563

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 20257015183

Country of ref document: KR

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 1020257015183

Country of ref document: KR