WO2023075177A1 - Chimeric antigen receptor specifically binding to cadm1 and uses thereof - Google Patents

Chimeric antigen receptor specifically binding to cadm1 and uses thereof Download PDF

Info

Publication number
WO2023075177A1
WO2023075177A1 PCT/KR2022/014464 KR2022014464W WO2023075177A1 WO 2023075177 A1 WO2023075177 A1 WO 2023075177A1 KR 2022014464 W KR2022014464 W KR 2022014464W WO 2023075177 A1 WO2023075177 A1 WO 2023075177A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
chimeric antigen
antigen receptor
cadm1
Prior art date
Application number
PCT/KR2022/014464
Other languages
French (fr)
Korean (ko)
Inventor
진화섭
전태훈
양준
이창희
이나영
박인병
강석진
박시원
이현정
Original Assignee
주식회사 이뮤노로지컬디자이닝랩
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 주식회사 이뮤노로지컬디자이닝랩 filed Critical 주식회사 이뮤노로지컬디자이닝랩
Publication of WO2023075177A1 publication Critical patent/WO2023075177A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • 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
    • C07K14/70517CD8
    • 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
    • C07K14/70521CD28, CD152
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • 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
    • C12N5/0646Natural killers cells [NK], NKT cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment

Definitions

  • the present invention is a chimeric antigen receptor (chimeric antigen receptor, CAR) that specifically binds to CADM1; a polynucleotide encoding the chimeric antigen receptor protein; a vector containing the polynucleotide; It relates to a T cell or natural killer cell (NK cell) transformed with the vector and a cell therapy or pharmaceutical composition for cancer treatment containing the cell as an active ingredient.
  • CAR chimeric antigen receptor
  • Cell adhesion molecule 1 also called Necl2, TSLC1, IGSF4, RA175, or SynCam, is an integral membrane protein belonging to the immunoglobulin (Ig) superfamily (Kuramochi et al., 2001; Murakami, 2005; Takai et al., 2008).
  • CADM1 is observed in various cells and is known to be involved in cell-to-cell adhesion to regulate various life phenomena (Murakami, 2005).
  • CADM1 was first discovered in non-small lung cancer cell (NSCLC) and is expressed in various epithelial-derived carcinomas. et al., 2001; Murakami, 2005; Takai et al., 2008).
  • Carcinomas expressing CADM1 reported so far are non-small lung cancer cell (NSCLC) (Kuramochi et al., 2001), lung adenocarcinoma (Murakami, 2005), human T-cell lymphotropic virus Type 1 infected T cell leukemia (HTLV-1-infected T cell leukemia) (Nakahata et al., 2012), adult T cell leukemia (Nakahata et al., 2021), ovarian cancer ) (Si et al., 2020), breast cancer (Takahashi et al., 2012), cervical cancer (Steenbergen et al., 2004), prostate cancer (Fukuhara et al. , 2002), nasopharyngeal carcinoma (Hui et al., 2003), stomach cancer (Honda et al., 2002), and pancreatic cancer (Jansen et al., 2002).
  • NSCLC non-small lung cancer cell
  • cytotoxic T lymphocytes CTL
  • NK cells natural killer cells
  • a key obtained by grafting a single-chain variable fragment (scFv) of an antibody that recognizes an antigen to a domain grafted to CD3 zeta or the cytoplasmic signaling domain of another protein has been attempted as a method of delivering a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the chimeric antigen receptor is grafted onto T cells or natural killer cells, the anticancer activity of T cells or natural killer cells can be achieved only by recognizing the specific antigen of the single-chain Fv fragment, regardless of signal transduction by antigen presenting cells (APCs). It can be activated, and it is not limited to the HLA type, so it can be used as a more efficient treatment method that can be used universally by many people.
  • these chimeric antigen receptor-expressing T cells or natural killer cells show efficacy in various cancers (Holzinger et al., 2016; Pettitt et al., 2018; Wang et al., 2020
  • class-I MHC-restricted T cell associated molecule is an integral membrane protein, expressed in activated lymphocytes (Kennedy et al., 2000), and cytotoxic T lymphocytes. ) or natural killer cells (NK cells), when they induce apoptosis of target cells, they play a role in better adhesion to target cells (Boles et al., 2005; Takeuchi et al., 2009). ). It also promotes the secretion of IFN- ⁇ and IL-22 by helper T cells (Yeh et al., 2008).
  • an antibody-drug conjugate (ADC) using an anti-CADM1 antibody (Korean Patent Publication No. 10-2011-0126739) or an anti-CADM1 antibody recognizing v9, a specific part of CADM1 (US Patent Publication No. 2021-0079092) has been published, but for the invention of using a chimeric antigen receptor in which the antigen-binding domain is a CRTAM extracellular domain having binding specificity for cancer cells expressing CADM1 as an immunocancer treatment not listed
  • CADM1 expression in various cancer cells was identified or induced, and that CADM1 and CRTAM could bind with high affinity, to develop a cancer-specific expression of CADM1 using the extracellular domain of CRTAM.
  • Chimeric antigen receptor-expressing T cells and natural killer cells were prepared, and it was confirmed through experiments that the T cells or natural killer cells had cytotoxic ability specifically to cells expressing CADM1.
  • signal transduction intensity was amplified by making a fusion protein linking each of the extracellular domains of CRTAM with the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region).
  • chimeric antigen receptors have only one antigen recognition site by a single-chain Fv fragment
  • the chimeric antigen receptor designed by the present inventors uses each of the extracellular domains of CRTAM in human immunoglobulin G 1 heavy chain.
  • the constant region IgG 1 heavy chain constant region
  • an object of the present invention is to provide a chimeric antigen receptor that specifically binds to CADM1.
  • Another object of the present invention is to provide a polynucleotide encoding the chimeric antigen receptor protein, a vector containing the polynucleotide, and T cells or natural killer cells transformed with the vector.
  • Another object of the present invention is to provide a pharmaceutical composition for the treatment of cancer that kills cancer cells expressing cell therapy or CADM1, including the transformed T cells or natural killer cells.
  • the present invention is an antigen binding site, an extracellular domain; transmembrane domain; And a chimeric antigen receptor (CAR) comprising an intracellular signaling domain, wherein the antigen-binding domain comprises an extracellular domain of a class-I MHC-restricted T cell associated molecule (CRTAM) that specifically binds to CADM1. It provides a chimeric antigen receptor.
  • CAR chimeric antigen receptor
  • the present invention provides a polynucleotide encoding the chimeric antigen receptor protein, a vector containing the polynucleotide, and T cells or natural killer cells transformed with the vector.
  • the present invention provides a cell therapy agent or a pharmaceutical composition for treating cancer that kills cancer cells expressing CADM1, including the transformed T cells or natural killer cells.
  • a chimeric antigen receptor according to the present invention comprises an extracellular domain of CRTAM that specifically binds to CADM1. Therefore, in the case of cytotoxic T cells or natural killer cells transformed with a vector capable of overexpressing the chimeric antigen receptor, they have cytotoxicity specifically to carcinoma expressing CADM1, so they are useful as immune cell therapeutics for cancer treatment can be used appropriately.
  • FIG. 1 is a schematic diagram showing cDNA regions of each domain expressing a chimeric antigen receptor according to an embodiment of the present invention.
  • FIG. 2A is a schematic diagram of a lentiviral vector according to an embodiment of the present invention.
  • 2B is a schematic diagram of a retroviral vector according to an embodiment of the present invention.
  • FIG. 3 is a schematic diagram of a chimeric antigen receptor expressed on the surface of cytotoxic T cells or natural killer cells according to an embodiment of the present invention.
  • FIG. 4 is a diagram showing the results of measuring the expression level of CADM1 in cancer cells (A549 cell line) expressing CADM1, which is the target of the chimeric antigen receptor provided in the present invention, using flow cytometry.
  • FIG. 5 is a schematic diagram showing cDNA regions of each domain expressing a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
  • FIG. 6 is a schematic diagram of an expression vector (retroviral vector) expressing a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
  • FIG. 7 is a schematic diagram of a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
  • FIG. 8a is a diagram showing the result of expressing the prepared recombinant CRTAM fusion protein in Chinese hamster ovary (CHO) cells and then purifying it by affinity chromatography using a protein A column.
  • Figure 8b is a view showing the result of Western blotting using an antibody (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody) recognizing the human IgG heavy chain constant region of the prepared recombinant CRTAM fusion protein.
  • an antibody anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody
  • 8C is a diagram showing whether the prepared recombinant CRTAM fusion protein recognizes cancer cells (A549 cell line) expressing CADM1 as a target, using flow cytometry.
  • Figure 9a is a diagram showing the results of comparing the expression ratio of GFP in cytotoxic T cells (CRTAM CAR-T cells) transduced with an expression vector according to an embodiment of the present invention using a lentivirus system.
  • FIG. 9B is a diagram showing the result of comparing the expression ratio of CRTAM in natural killer cells (CRTAM CAR-NK cells) transduced with an expression vector according to an embodiment of the present invention using a retroviral system.
  • Figure 10a is a cytotoxic T cell (CRTAM CAR-T cell) according to an embodiment of the present invention or a cytotoxic T cell (Mock T cell) transduced with an empty vector as effector cells to express CADM1 It is a diagram showing the results of measuring cytotoxicity against cancer cells (A549 cell line).
  • 10B is cancer cells expressing CADM1 using CRTAM CAR-NK cells according to an embodiment of the present invention or Mock NK cells transduced with an empty vector as effector cells; It is a diagram showing the results of measuring cytotoxicity to (A549 cell line).
  • an antigen-binding domain comprises an extracellular domain of CRTAM that specifically binds to CADM1.
  • the "Chimeric antigen receptor (CAR)" naturally binds to a desired antigen without the mediation of an antigen-presenting cell (APC) or antibody necessary for the activation of T cells or natural killer cells, resulting in an antigen-antibody reaction. It refers to a fusion protein to be expressed in T cells or natural killer cells in order to induce activation of T cells and attack cells expressing the corresponding antigen. That is, it can be seen as a protein that binds to an antigen when expressed in T cells or natural killer cells and induces the activation of these cells. Through this, it may be a protein that recognizes an antigen specific to a cell to cause an immune response, and the cell to cause an immune response may refer to a cell present in a specific tissue or forming a tissue that causes a lesion.
  • APC antigen-presenting cell
  • Chimeric antigen receptor proteins may include functional equivalents.
  • 'Functional equivalent' means at least 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably, the amino acid sequence of the chimeric antigen receptor protein as a result of addition, substitution, or deletion of amino acids. has a sequence homology of 95% or more, and refers to a protein that exhibits substantially the same physiological activity.
  • 'Substantially homogeneous physiological activity means having an activity that can specifically bind to CADM1.
  • the present invention also includes fragments, derivatives and analogs of chimeric antigen receptors.
  • fragments, derivatives and analogs of chimeric antigen receptors include (1) a polypeptide in which one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted (the substituted amino acid residues are encoded by the genetic code).
  • polypeptide having substituent(s) on one or more amino acid residues (2) a polypeptide having substituent(s) on one or more amino acid residues, or (3) another compound (a compound capable of extending the half-life of a polypeptide, such as polyethylene glycol) a polypeptide derived from the combined mature polypeptide, or (4) an additional amino acid sequence (e.g., a leader sequence, a secreted sequence, a sequence used to purify the polypeptide, a proteinogen sequence, or a fusion protein) and It may be a polypeptide derived from the polypeptide to which it is linked.
  • additional amino acid sequence e.g., a leader sequence, a secreted sequence, a sequence used to purify the polypeptide, a proteinogen sequence, or a fusion protein
  • CRTAM class-I MHC-restricted T cell associated molecule
  • activated lymphocytes such as cytotoxic T lymphocytes or natural killer cells, When NK cell) induces apoptosis of target cells, it serves to better adhere to target cells. In addition, it promotes the secretion of IFN- ⁇ and IL-22 by helper T cells.
  • the antigen-binding domain may be in the form of a dimer in which a pair of extracellular domains of CRTAM are linked to a human immunoglobulin G 1 heavy chain constant region , respectively, to amplify signal transduction. It is not limited.
  • the extracellular domain of CRTAM may consist of SEQ ID NO: 1 or an amino acid sequence showing 95% or more homology thereto, but is not limited thereto.
  • the human immunoglobulin G 1 heavy chain constant region may consist of SEQ ID NO: 2 or an amino acid sequence exhibiting 95% or more homology thereto, but is not limited thereto.
  • CADM1 is known to be expressed in various epithelial tissues and immune cells, and its expression is increased in some carcinomas, so that it is used as a molecular marker of carcinoma or as a target for treatment.
  • the known CADM1-expressing carcinomas include non-small lung cancer cell (NSCLC), lung adenocarcinoma, and human T-cell lymphotropic virus type 1-infected T-cell leukemia (HTLV-1-infected T-cell). cell leukemia), adult T cell leukemia, ovarian cancer, breast cancer, cervical cancer, prostate cancer, nasopharyngeal carcinoma, stomach cancer (stomach cancer) and pancreatic cancer.
  • the "transmembrane domain” is a site that connects the extracellular domain of CRTAM and the co-stimulatory and essential signaling domains between the cell membrane, and the intracellular signaling domain is an immune cell by binding of the antigen-binding domain. refers to the site that activates the immune response of
  • the transmembrane domain is selected from the group consisting of CD28, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154 It may include one or more types, and the CD8 may be CD8 ⁇ or CD8 ⁇ , preferably a transmembrane domain of CD8 ⁇ , which may consist of SEQ ID NO: 3 or an amino acid sequence showing 95% or more homology thereto. However, it is not limited thereto.
  • intracellular signaling domain means a site that activates the immune response of immune cells by binding to an antigen binding domain.
  • the intracellular domain may be CD28, 4-1BB, CD3 zeta, or a combination thereof, but is not limited thereto.
  • the chimeric antigen receptor according to the present invention uses CD28, 4-1BB, and CD3 zeta as intracellular signaling domains, thereby exhibiting a high-activity killing effect on cancer cells, particularly cancer cells expressing CADM1.
  • the CD28 has SEQ ID NO: 4 or 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more sequence homology thereto, and the amino acid sequence represented by SEQ ID NO: 4 4-1BB (CD137) is SEQ ID NO: 5 or 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more.
  • sequence homology may consist of an amino acid sequence showing a function substantially equivalent to the amino acid sequence represented by SEQ ID NO: 5, CD3 zeta functions as an NK cell activation domain, and SEQ ID NO: 6 or 70 % or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more sequence homology, and an amino acid sequence that exhibits a function substantially equivalent to that of the amino acid sequence represented by SEQ ID NO: 6 It can be done.
  • the antigen-binding domain may include a CD8 ⁇ signal peptide, and the CD8 ⁇ signal peptide may consist of SEQ ID NO: 7 or an amino acid sequence showing 95% or more homology thereto, but is not limited thereto.
  • a polynucleotide encoding the chimeric antigen receptor protein is provided.
  • the polynucleotide encoding the antigen receptor of the present invention changes the amino acid sequence of the antigen receptor expressed from the coding region due to codon degeneracy or in consideration of codons preferred in organisms intended to express the antigen receptor.
  • Various modifications may be made to the coding region within a range not specified, and various modifications or modifications may be made to a part other than the coding region within a range that does not affect gene expression, and such modified genes are also within the scope of the present invention. It will be well understood by those skilled in the art.
  • nucleic acid bases may be mutated by substitution, deletion, insertion, or a combination thereof, and these are also included in the scope of the present invention.
  • a vector containing the polynucleotide and a cell transformed with the vector are provided.
  • Vectors used in the present invention may use various vectors known in the art, and depending on the type of host cell to produce the antigen receptor, a promoter, terminator, enhancer, etc. Expression control sequences, sequences for membrane targeting or secretion, etc. may be appropriately selected and combined in various ways depending on the purpose.
  • Vectors of the present invention include, but are not limited to, plasmid vectors, cosmid vectors, bacteriophage vectors and viral vectors. Suitable vectors include expression control elements such as promoters, operators, initiation codons, stop codons, polyadenylation signals and enhancers, as well as signal sequences or leader sequences for membrane targeting or secretion, and may be prepared in various ways depending on the purpose.
  • a lenti-virus vector or a retro-virus vector can be used, and in the following embodiment of the present invention, the pCDH-CMV-MCS-EF1-copGFP vector (lenti-virus vector) and pLNCX2 (retro-viral vector) was used, but is not limited thereto.
  • cells may be transformed by introducing a chimeric antigen receptor that specifically binds to CADM1 into cells through the vector.
  • the cells may be T cells, tumor-infiltrating lymphocytes, B cells, natural killer cells, or NKT cells, and preferably may be cytotoxic T cells or natural killer cells.
  • the cells may be obtained or prepared from bone marrow, peripheral blood, peripheral blood mononuclear cells or umbilical cord blood, and the cells may be human cells, but are not limited thereto.
  • cells transformed by introducing the chimeric antigen receptor of the present invention recognize CADM1 as an antigen and have a characteristic of strongly binding thereto.
  • chimeric antigen receptor T cell (hereinafter, simply abbreviated as 'CAR-T cell')" or “chimeric antigen receptor expressing NK cell (chimeric antigen receptor NK cell, hereinafter) Shortly abbreviated as 'CAR-NK cell')
  • 'CAR-NK cell' is a chimeric antigen receptor that specifically responds to cancer cells rather than the original T cell receptor or NK cell receptor by a method such as transduction of normal T cells or natural killer cells. refers to T cells or NK cells expressing T cells or NK cells having this receptor exhibit cytotoxicity by inducing apoptosis of target cells.
  • CAR-T cells or CAR-NK cells may be cells into which the chimeric antigen receptor of the present invention is introduced into cytotoxic T cells or NK cells.
  • the cells have the advantage of anticancer-specific targeted therapy, which is an existing advantage of CAR-T therapeutics.
  • the CAR-T cells or CAR-NK cells of the present invention can recognize and effectively destroy cancer cells expressing CADM1.
  • the present invention provides a cell therapeutic agent containing the cells or a pharmaceutical composition for treating cancer containing the same as an active ingredient.
  • cell therapy refers to cells and tissues prepared from a subject through isolation, culture, and special manipulation, and is a pharmaceutical product used for the purpose of treatment, and is a living autologous, allogeneic, or xenogeneic living agent to restore the function of a cell or tissue. It refers to drugs used for the purpose of treatment through a series of actions, such as proliferation and selection of cells in vitro or altering the biological characteristics of cells in other ways.
  • treatment refers to all activities in which symptoms caused by cancer are improved or beneficially changed by administration of the composition.
  • composition may include a pharmaceutically acceptable carrier.
  • the "pharmaceutically acceptable carrier” may refer to a carrier or diluent that does not inhibit the biological activity and properties of the compound to be injected without irritating living organisms.
  • the type of the carrier that can be used in the present invention is not particularly limited, and any carrier commonly used in the art and pharmaceutically acceptable can be used.
  • Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and the like. These may be used alone or in combination of two or more.
  • composition containing a pharmaceutically acceptable carrier may be in various oral or parenteral formulations. When formulated, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
  • solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, and these solid preparations contain at least one excipient such as starch, calcium carbonate, sucrose, and lactose in the compound. , can be prepared by mixing gelatin, etc.
  • lubricants such as magnesium stearate and talc may also be used.
  • Liquid preparations for oral use include suspensions, solutions for internal use, emulsions, and syrups.
  • various excipients such as wetting agents, sweeteners, aromatics, and preservatives may be included. there is.
  • Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried formulations, and suppositories.
  • Propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate may be used as non-aqueous solvents and suspending agents.
  • Witepsol, Macrogol, Tween 61, cacao butter, laurin paper, glycerogelatin, and the like may be used as a base for the suppository.
  • composition can be administered in a pharmaceutically effective amount.
  • the "pharmaceutically effective amount” means an amount sufficient to treat a disease with a reasonable benefit / risk ratio applicable to medical treatment, and the effective dose level is dependent on the type and severity of the subject, age, sex, infected virus type, and drug activity, sensitivity to the drug, time of administration, route of administration and rate of excretion, duration of treatment, factors including concomitantly used drugs and other factors well known in the medical arts.
  • the administration means introducing the composition of the present invention to a patient by any suitable method, and the administration route of the composition may be administered through any general route as long as it can reach the target tissue.
  • Intraperitoneal administration, intravenous administration, intramuscular administration, subcutaneous administration, intradermal administration, oral administration, topical administration, intranasal administration may be administered, but is not limited thereto.
  • composition of the present invention may be administered daily or intermittently, and the number of administrations per day may be administered once or divided into 2 to 3 times.
  • the number of administrations when the two active ingredients are each a single agent may be the same or may be different.
  • the composition of the present invention can be used alone or in combination with other drug treatments for the treatment of CADM1-expressing cancer. It is important to administer the amount that can obtain the maximum effect with the minimum amount without side effects in consideration of all the above factors, and can be easily determined by those skilled in the art.
  • the subject refers to all humans, including humans, monkeys, cows, horses, sheep, pigs, chickens, turkeys, quails, cats, dogs, mice, rats, rabbits, or guinea pigs that have or may develop cancer expressing CADM1. means animals.
  • the type of subject is included without limitation as long as the disease can be effectively treated by administering the pharmaceutical composition of the present invention to the subject.
  • Types of cancer expressing CADM1 to be treated in the present invention include non-small lung cancer cell (NSCLC), lung adenocarcinoma, and T cells infected with human T-cell lymphotropic virus type 1.
  • NSCLC non-small lung cancer cell
  • lung adenocarcinoma lung adenocarcinoma
  • Leukemia HTLV-1-infected T cell leukemia
  • adult T cell leukemia ovarian cancer, breast cancer, cervical cancer, prostate cancer
  • Examples include nasopharyngeal carcinoma, stomach cancer and pancreatic cancer, but are not limited thereto.
  • an antigen recognition site in which a pair of extracellular domains of CRTAM are linked to a human immunoglobulin G 1 heavy chain constant region, and a transmembrane domain of CD8 ⁇ , Protein coding sequences of the intracellular domains of each of CD28, 4-1BB, and CD3 zeta were identified in a gene database.
  • FIG. 1 A schematic diagram showing the cDNA region of each domain expressing the chimeric antigen receptor is shown in FIG. 1 .
  • the corresponding gene was used as a lentivirus-derived expression vector, pCDH-CMV -MCS-EF1-copGFP (System Biosciences) or retrovirus-derived expression vector pLNCX2 (Addgene) was cloned.
  • a primer that creates a restriction enzyme XbaI cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is created by polymerase chain reaction, and a restriction enzyme cleavage site is also made at the 3' end.
  • a restriction enzyme cleavage site was prepared by synthesizing a primer that creates the cleavage site sequence of the enzyme NotI and polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with XbaI, and the 3' end was treated with NotI. In addition, the multicloning site of the expression vector was treated with XbaI and NotI so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
  • a primer that creates a restriction enzyme Bgl II cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is made by polymerase chain reaction, and a restriction enzyme NotI cleavage site sequence is also added at the 3' end.
  • a primer was synthesized to make a restriction enzyme cleavage site by polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with BglII, and the 3' end was treated with NotI. Then, the multicloning site of the expression vector was treated with BglII and NotI so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
  • CADM1 was expressed in A549 (ATCC), a cell line derived from lung adenocarcinoma.
  • A549 A549
  • the cell lines were treated with an antibody (Biobyt) capable of binding to CADM1 to which a fluorescent protein was attached, and then fluorescence-activated cell sorting was used.
  • an antibody Biobyt capable of binding to CADM1 to which a fluorescent protein was attached, and then fluorescence-activated cell sorting was used.
  • CADM1 was expressed in the A549 cell line (see FIG. 4).
  • the A549 cell line expressing CADM1 was used as a target cell for CRTAM.CAR-T cells or CRTAM.CAR-NK cells.
  • a water-soluble recombinant CRTAM fusion protein was prepared.
  • protein coding sequences in which each of the extracellular domains of CRTAM were linked to the human immunoglobulin G1 heavy chain constant region were identified in a gene database.
  • gene synthesis was performed by connecting the remaining sequences except for the stop codon portion of the base sequence constituting the domains into one. The accuracy of gene synthesis was confirmed through sequencing after constructing a protein expression plasmid.
  • a schematic diagram showing the cDNA region of each domain expressing the recombinant protein is shown in FIG. 5 .
  • the corresponding gene was transferred to the retrovirus-derived expression vector, pLNCX2. (Addgene).
  • a primer that creates a restriction enzyme Xho I cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is created by polymerase chain reaction (PCR), and restriction is also made at the 3' end.
  • a restriction enzyme cleavage site was prepared by synthesizing a primer that creates the cleavage site sequence of the enzyme Not I and polymerase chain reaction.
  • the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with Xho I, and the 3' end was treated with Not I.
  • the multicloning site of the expression vector was treated with xho I and Not I so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
  • affinity chromatography was performed using a protein A column ( affinity chromatography) (GE healthcare) (see Fig. 8a).
  • the purified water-soluble chimeric antigen receptor was confirmed by Western blotting using an antibody (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody, abcam) recognizing the human IgG heavy chain constant region (Fig. see 8b).
  • the purified water-soluble recombinant CRTAM fusion protein was treated with a cell line (A549 cell line) expressing CADM1, and it was confirmed through flow cytometry whether the water-soluble recombinant CRTAM fusion protein could bind to the cells.
  • the results are shown in FIG. 8C.
  • A549 cells expressing CADM1 did not bind to control Ig (isotype control), but bound to soluble CRTAM fusion protein (CRTAM-Ig).
  • the prepared soluble CRTAM fusion protein had high affinity with human cell lines expressing CADM1, which recognized CADM1-expressing A549 cells and recognized human immunoglobulin G 1 Verification of CADM1 protein-specific cytotoxicity to cytotoxic T cells (CRTAM.CAR-T cells) or natural killer cells (CRTAM.CAR-NK cells) expressing a recombinant protein fused with a heavy chain constant region and a signaling protein domain means that it can be used for
  • cytotoxic T cells were first isolated from human peripheral blood mononuclear cells. After purchasing human peripheral blood mononuclear cells (Medi Lab Korea), magnetic-activated cell sorting was used to isolate cytotoxic T cells. Peripheral blood mononuclear cells are combined with an antibody (CD8 + T cell biotin-conjugated antibody cocktail, Miltenyi Biotec) capable of binding to other immune cells except for cytotoxic T cells, and then the antibodies are combined with magnetic microbeads (anti- biotin microbead) (Miltenyi Biotec).
  • an antibody CD8 + T cell biotin-conjugated antibody cocktail, Miltenyi Biotec
  • magnetic microbeads anti- biotin microbead
  • the microbeads, antibodies attached thereto, and cells were passed through a magnetic separation column (Miltenyi Biotec) to obtain cytotoxic T cells unlabeled with the antibodies.
  • flow cytometry was performed using CD8 and CD3 ⁇ , which are cell surface factors of cytotoxic T cells, and as a result, the purity was over 95%.
  • cytotoxic T cells Contains 1 ⁇ 10 6 /ml of human CD3 and CD28 antibody-coated magnetic beads (Thermo Fisher Scientific) and 10% calf serum supplemented with 100 U/ ⁇ l recombinant human IL-2 for activation of isolated cytotoxic T cells.
  • the cells were reconstituted in RPMI (Welgene) at a density of 1.5 ⁇ 10 6 cells/ml, inoculated into a 24-well cell culture dish, and cultured for 24 hours.
  • Example 8 Construction of chimeric antigen receptor-expressing cytotoxic T cells
  • Example 2 In order to introduce the recombinant vector prepared in Example 2 into cytotoxic T cells, a lentiviral system using 293FT cells (Thermo Fisher Scientific) was used.
  • 293FT cells were inoculated into 2.5 ⁇ 10 6 cells in a 100 ⁇ cell culture dish, and cultured in DMEM medium containing 10% calf serum. After 24 hours of incubation, when the cells have grown to cover 60-70% of the dish, 20 ⁇ g of CRTAM.CAR-T.pCDH-CMV-MCS-EF1-copGFP vector DNA was mixed with 10 ⁇ g of psPAX2 (Addgene) and 3 ⁇ g of pMD2. .G (Addgene) vector was crystallized using Calcium phosphate and Hepes-buffered solution and then introduced into 293FT cells.
  • culture supernatants containing lentivirus were collected at intervals of 24 hours after 48 hours of 293FT cells into which the expression vector was introduced.
  • the collected supernatant was centrifuged for 2 hours at 21,000 rpm in an ultra-high-speed centrifuge to concentrate the virus.
  • the concentrated virus was mixed with 4 ⁇ g/ml of polybrene and added to the culture medium of activated cytotoxic T cells, followed by centrifugation at 1,800 g for 75 minutes using a centrifuge for transduction.
  • the centrifuged cytotoxic T cells were further cultured for 4 hours and then replaced with RPMI culture medium containing 10% calf serum. After 48 hours, some of the transduced cytotoxic T cells were used to measure transduction efficiency.
  • the transduction efficiency was measured by flow cytometry to measure the amount of GFP expression inside the cell, and the transduced cytotoxic T cell (CRTAM CAR-T cell) was measured using a virus-transduced cytotoxic T cell (Mock CAR-T cell). Transduction rates of T cells) were compared (see FIG. 9a).
  • the collected retrovirus-containing supernatant was centrifuged at 21,000 rpm for 2 hours using an ultra-high-speed centrifuge, and then reconstituted in Myelocult H5100 culture medium (STEMCELL) containing 10% calf serum to be concentrated 100 times compared to before concentration.
  • NK92MI cells American type culture collection, ATCC
  • NK92MI cells were inoculated in a 24-well cell culture dish at a density of 5 ⁇ 10 5 /ml. Then, a mixed solution in which 8ug/ml of polybrene was added to the concentrated retrovirus was added to the culture medium of NK92MI cells, and cultured for 24 hours.
  • NK92MI cells treated with retrovirus were cultured using Myelocult H5100 culture medium supplemented with neomycin antibiotic (600 ⁇ g/ml) 24 hours after the culture medium change for selection of cells into which the gene was introduced. After 14 days of neomycin selection, NK92MI cells were transferred to a fresh culture medium and grown for one week.
  • NK92MI cells CRTAM CAR-NK cells introduced with CRTAM.CAR-NK.pLNCX2 and NK92MI cells (Mock CAR-NK cells) introduced with pLNCX2, an empty vector
  • NK92MI cells CRTAM.CAR-NK.pLNCX2
  • NK92MI cells NK92MI cells introduced with pLNCX2
  • anti-human CRTAM antibody rat IgG, abcam, #ab173178
  • anti-rat IgG-Alexa Fluor 488 goat anti-rat IgG, thermofisher, #a11006
  • A549 cells (CADM1 positive cells), the target cells selected in Example 3, were used as chimeric antigens.
  • Receptor-expressing cytotoxic T cells (CRTAM CAR-T cells) or empty vector-transduced cytotoxic T cells (Mock CAR-T cells) were co-cultured for 6 hours.
  • a non-radioactive cytotoxicity assay that measures the degree of cytotoxicity of cytotoxic T cells to target cells by the amount of lactate dehydrogenase present in the supernatant after co-culture used
  • cytotoxic T cells (1x10 5 cell) and target cells (1x10 4 cell) into wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, and inoculate so that the volume per well is 100 ⁇ l. Centrifuge for 4 minutes under the condition of 250g using a centrifuge to close the gap between cells. After 6 hours of incubation, 50 ⁇ l of the supernatant from each well is removed and transferred to a transparent 96-well dish for measuring absorbance, and then the enzyme reaction is progressed and stopped by treatment with analysis solution and 1M hydrochloric acid solution.
  • the degree of cytotoxicity of the cytotoxic T cells in each well was quantified by measuring and converting the absorbance in the 490 nm wavelength band using a fluorescence/emission/absorption meter (multi-detection plate reader).
  • the prepared chimeric antigen receptor-expressing cytotoxic T cell (CRTAM CAR-T cell) showed a high toxicity of 39% or more to the A549 cell line expressing CADM1, while introducing an empty vector as a control One cytotoxic T cell (Mock CAR-T cell) showed 3% toxicity to the A549 cell line.
  • A549 cells (CADM1 positive cells), the target cells selected in Example 3, were expressed as chimeric antigen receptors.
  • NK cells CTAM CAR-NK cells
  • NK cells Mock CAR-NK cells
  • a non-radioactive cytotoxicity assay was used to measure the degree of cytotoxicity of NK cells to target cells by the amount of lactate dehydrogenase present in the supernatant.
  • the degree of cytotoxicity of NK cells in each well was quantified by measuring and converting the absorbance in the 490 nm wavelength band using a fluorescence/luminescence/absorption meter (multi-detection plate reader).
  • the prepared chimeric antigen receptor-expressing natural killer cells (CRTAM CAR-NK cells) showed high toxicity of 27% or more to the CADM1-expressing A549 cell line, whereas the empty vector as a control was introduced.
  • Natural killer cells (Mock CAR-NK cells) showed toxicity of -2% or less to the A549 cell line.
  • the chimeric antigen recognition receptor-expressing NK cells containing the CRTAM extracellular domain showed specific cytotoxicity to the CADM1 protein, and thus the treatment of related cancer cells using the CAR-NK cells was possible. .

Abstract

The present invention relates to a chimeric antigen receptor (CAR) that specifically binds to CADM1; a polynucleotide encoding the chimeric antigen receptor protein; a vector comprising the polynucleotide; and a T cell or natural killer cell (NK cell) transformed with the vector, and a cellular therapeutic agent or a pharmaceutical composition for the treatment of cancer comprising the cell as an active ingredient. The chimeric antigen receptor according to the present invention comprises an extracellular domain of CRTAM that specifically binds to CADM1. Accordingly, as cytotoxic T cells or natural killer cells transformed with a vector capable of overexpressing the chimeric antigen receptor have cytotoxicity specifically to carcinoma expressing CADM1, the cytotoxic T cells or natural killer cells can be effectively used as an immune cell therapeutic agent for the treatment of cancer.

Description

CADM1에 특이적으로 결합하는 키메릭 항원 수용체 및 이의 용도Chimeric antigen receptor that specifically binds to CADM1 and uses thereof
본 발명은 CADM1에 특이적으로 결합하는 키메릭 항원 수용체(chimeric antigen receptor, CAR); 상기 키메릭 항원 수용체 단백질을 코딩하는 폴리 뉴클레오티드; 상기 폴리 뉴클레오티드를 포함하는 벡터; 상기 벡터로 형질전환된 T 세포 또는 자연살생세포(Natural killer cell, NK cell) 및 상기 세포를 유효성분으로 포함하는 세포 치료제 또는 암의 치료용 약학적 조성물에 관한 것이다.The present invention is a chimeric antigen receptor (chimeric antigen receptor, CAR) that specifically binds to CADM1; a polynucleotide encoding the chimeric antigen receptor protein; a vector containing the polynucleotide; It relates to a T cell or natural killer cell (NK cell) transformed with the vector and a cell therapy or pharmaceutical composition for cancer treatment containing the cell as an active ingredient.
Cell adhesion molecule 1 (CADM1)은 Necl2, TSLC1, IGSF4, RA175, 또는 SynCam이라고도 불리우며, 면역 글로불린(Ig) 수퍼 패밀리(immunoglobulin superfamily)에 속하는 내재성 막 단백질(integral membrane protein)이다(Kuramochi et al., 2001; Murakami, 2005; Takai et al., 2008). CADM1은 다양한 세포에서 관찰되며, 세포와 세포 간의 부착에 관여하여 여러가지 생명 현상을 조절할 수 있다고 알려져 있다(Murakami, 2005). CADM1은 비소세포 폐암(non-small lung cancer cell, NSCLC)에서 처음으로 발견되었고, 다양한 상피조직 유래 암종에서 발현되며, 몇몇 암종에서는 발현이 증가되어 암종의 분자 표지 또는 치료용 타겟으로 사용된다(Kuramochi et al., 2001; Murakami, 2005; Takai et al., 2008). 지금까지 보고된 CADM1을 발현하는 암종은 비소세포 폐암(non-small lung cancer cell, NSCLC)(Kuramochi et al., 2001), 폐 선암종(lung adenocarcinoma)(Murakami, 2005), 사람T세포림프친화바이러스 1형에 감염된 T 세포 백혈병 (HTLV-1-infected T cell leukemia)(Nakahata et al., 2012), 성인T세포백혈병(adult T cell leukemia)(Nakahata et al., 2021), 난소암(ovarian cancer)(Si et al., 2020), 유방암(breast cancer)(Takahashi et al., 2012), 자궁경부암(cervical cancer)(Steenbergen et al., 2004), 전립선암(prostate cancer)(Fukuhara et al., 2002), 상인두암(nasopharyngeal carcinoma)(Hui et al., 2003), 위암(stomach cancer)(Honda et al., 2002), 췌장암(pancreatic cancer)(Jansen et al., 2002) 으로 알려져있다.Cell adhesion molecule 1 (CADM1), also called Necl2, TSLC1, IGSF4, RA175, or SynCam, is an integral membrane protein belonging to the immunoglobulin (Ig) superfamily (Kuramochi et al., 2001; Murakami, 2005; Takai et al., 2008). CADM1 is observed in various cells and is known to be involved in cell-to-cell adhesion to regulate various life phenomena (Murakami, 2005). CADM1 was first discovered in non-small lung cancer cell (NSCLC) and is expressed in various epithelial-derived carcinomas. et al., 2001; Murakami, 2005; Takai et al., 2008). Carcinomas expressing CADM1 reported so far are non-small lung cancer cell (NSCLC) (Kuramochi et al., 2001), lung adenocarcinoma (Murakami, 2005), human T-cell lymphotropic virus Type 1 infected T cell leukemia (HTLV-1-infected T cell leukemia) (Nakahata et al., 2012), adult T cell leukemia (Nakahata et al., 2021), ovarian cancer ) (Si et al., 2020), breast cancer (Takahashi et al., 2012), cervical cancer (Steenbergen et al., 2004), prostate cancer (Fukuhara et al. , 2002), nasopharyngeal carcinoma (Hui et al., 2003), stomach cancer (Honda et al., 2002), and pancreatic cancer (Jansen et al., 2002).
효과적인 암 치료를 위해서 직접적으로 암 세포를 표적하는 세포독성 T 세포(cytotoxic T lymphocytes, CTL)와 자연살생세포(natural killer cell, NK cell)가 중요하다는 사실이 보고되어왔다. 지금까지 세포독성 T 세포 또는 자연살생세포를 이용한 항암 치료에 대한 연구는 환자 유래 특정 암 항원을 환자의 세포독성 T 세포에 전달하여 활성을 유발하거나, 항체 의존성 세포 독성(antibody dependent cellular cytotoxicity)을 유발하고자 하였다(Galluzzi et al., 2018; Lu et al., 2020). 하지만, 최근에는 세포독성 T 세포와 자연살생세포를 이용한 새로운 항암 치료 방법이 도입되었다. 즉, 항원을 인식하는 항체의 단일사슬 Fv 단편(single-chain variable fragment, scFv) 부분을 CD3 제타(zeta) 또는 다른 단백질의 세포질내 신호전달 도메인(cytoplasmic signaling domain)에 접목시킨 도메인에 접목시킨 키메릭 항원 수용체(chimeric antigen receptor, CAR)를 전달하는 방법으로 시도되었다. 키메릭 항원 수용체를 T 세포 또는 자연살생세포에 접목시키면 항원 제시 세포(antigen presenting cell, APC)에 의한 신호 전달과 관계없이 단일사슬 Fv 단편의 특정 항원 인지만으로 T 세포 또는 자연살생세포의 항암 작용을 활성화시킬 수 있으며, 또한 HLA type에 제한적이지 않아 많은 사람들이 보편적으로 사용할 수 있는 보다 효율적인 치료 방법으로 이용할 수 있다. 실제로, 이러한 키메릭 항원 수용체 발현 T 세포나 자연살생세포는 여러 암에서 효능을 보이고 있다(Holzinger et al., 2016; Pettitt et al., 2018; Wang et al., 2020).It has been reported that cytotoxic T lymphocytes (CTL) and natural killer cells (NK cells) that directly target cancer cells are important for effective cancer treatment. Until now, research on anticancer treatment using cytotoxic T cells or natural killer cells has been carried out by delivering patient-derived specific cancer antigens to the patient's cytotoxic T cells to induce activation or antibody dependent cellular cytotoxicity. (Galluzzi et al., 2018; Lu et al., 2020). However, recently, a new anti-cancer treatment method using cytotoxic T cells and natural killer cells has been introduced. That is, a key obtained by grafting a single-chain variable fragment (scFv) of an antibody that recognizes an antigen to a domain grafted to CD3 zeta or the cytoplasmic signaling domain of another protein. It has been attempted as a method of delivering a chimeric antigen receptor (CAR). When the chimeric antigen receptor is grafted onto T cells or natural killer cells, the anticancer activity of T cells or natural killer cells can be achieved only by recognizing the specific antigen of the single-chain Fv fragment, regardless of signal transduction by antigen presenting cells (APCs). It can be activated, and it is not limited to the HLA type, so it can be used as a more efficient treatment method that can be used universally by many people. In fact, these chimeric antigen receptor-expressing T cells or natural killer cells show efficacy in various cancers (Holzinger et al., 2016; Pettitt et al., 2018; Wang et al., 2020).
한편, class-I MHC-restricted T cell associated molecule (CRTAM)은 내재성 막 단백질(integral membrane protein)로, 활성화된 림프구에서 발현되며(Kennedy et al., 2000), 세포독성 T 림프구(cytotoxic T lymphocyte)나 자연살생세포(natural killer cell, NK cell)가 표적세포의 세포자살을 유도할 때, 표적세포에 좀 더 잘 부착되게 하는 역할을 한다(Boles et al., 2005; Takeuchi et al., 2009). 또한 보조 T 림프구(helper T cell)의 IFN-η와 IL-22 분비량을 촉진시킨다(Yeh et al., 2008).On the other hand, class-I MHC-restricted T cell associated molecule (CRTAM) is an integral membrane protein, expressed in activated lymphocytes (Kennedy et al., 2000), and cytotoxic T lymphocytes. ) or natural killer cells (NK cells), when they induce apoptosis of target cells, they play a role in better adhesion to target cells (Boles et al., 2005; Takeuchi et al., 2009). ). It also promotes the secretion of IFN-η and IL-22 by helper T cells (Yeh et al., 2008).
현재, 항-CADM1 항체를 이용한 항체-약물 접합체(antibody drug conjugate, ADC)(한국공개특허 제10-2011-0126739호)나 CADM1의 특정 부분인 v9을 인식하는 항-CADM1 항체(미국공개특허 제2021-0079092호)는 공개되어 있으나, CADM1이 발현되는 암세포에 대한 결합 특이성을 갖는 CRTAM 세포외 도메인(extracellular domain)을 항원 결합 도메인의 구성으로 한 키메릭 항원 수용체를 면역 항암 치료제로서 이용하는 발명에 대해서는 기재된 바 없다.Currently, an antibody-drug conjugate (ADC) using an anti-CADM1 antibody (Korean Patent Publication No. 10-2011-0126739) or an anti-CADM1 antibody recognizing v9, a specific part of CADM1 (US Patent Publication No. 2021-0079092) has been published, but for the invention of using a chimeric antigen receptor in which the antigen-binding domain is a CRTAM extracellular domain having binding specificity for cancer cells expressing CADM1 as an immunocancer treatment not listed
이러한 배경하에, 본 발명자들은 다양한 암세포에서의 CADM1 발현이 확인 또는 유도된다는 점과, 상기 CADM1과 CRTAM이 높은 친화도로 결합 가능하다는 점을 이용하여 CRTAM의 세포외 도메인을 사용한 CADM1을 발현하는 암 특이적 키메릭 항원 수용체 발현 T 세포와 자연살생세포를 제작하고, 상기 T 세포 또는 자연살생세포가 CADM1을 발현하는 세포에 특이적으로 세포독성 능력이 있음을 실험을 통해 확인하였다. 이 때, CRTAM의 세포외 도메인 각각을 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)과 연결하는 융합 단백질을 만들어 신호 전달 강도를 증폭하였다. 즉, 기존의 키메릭 항원 수용체는 단일사슬 Fv 단편에 의해 항원을 인식하는 부위가 하나인 반면에, 본 발명자들이 고안한 키메릭 항원 수용체는 CRTAM의 세포외 도메인 각각을 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)과 연결시켜, 키메릭 항원 수용체 하나당 2개의 CRTAM의 세포외 도메인이 CADM1 항원을 인식하게 하여, 신호 전달 강도를 증폭하고자 하였다.Under this background, the present inventors found that CADM1 expression in various cancer cells was identified or induced, and that CADM1 and CRTAM could bind with high affinity, to develop a cancer-specific expression of CADM1 using the extracellular domain of CRTAM. Chimeric antigen receptor-expressing T cells and natural killer cells were prepared, and it was confirmed through experiments that the T cells or natural killer cells had cytotoxic ability specifically to cells expressing CADM1. At this time, signal transduction intensity was amplified by making a fusion protein linking each of the extracellular domains of CRTAM with the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region). That is, existing chimeric antigen receptors have only one antigen recognition site by a single-chain Fv fragment, whereas the chimeric antigen receptor designed by the present inventors uses each of the extracellular domains of CRTAM in human immunoglobulin G 1 heavy chain. By linking to the constant region (IgG 1 heavy chain constant region), two extracellular domains of CRTAM per chimeric antigen receptor were allowed to recognize the CADM1 antigen, thereby amplifying signal transduction strength.
따라서 본 발명의 목적은 CADM1에 특이적으로 결합하는 키메릭 항원 수용체를 제공하는 것이다.Accordingly, an object of the present invention is to provide a chimeric antigen receptor that specifically binds to CADM1.
본 발명의 다른 목적은 상기 키메릭 항원 수용체 단백질을 코딩하는 폴리 뉴클레오티드, 상기 폴리 뉴클레오티드를 포함하는 벡터 및 상기 벡터로 형질전환된 T 세포 또는 자연살생세포를 제공하는 것이다.Another object of the present invention is to provide a polynucleotide encoding the chimeric antigen receptor protein, a vector containing the polynucleotide, and T cells or natural killer cells transformed with the vector.
본 발명의 또 다른 목적은 상기 형질전환된 T 세포 또는 자연살생세포를 포함하는, 세포 치료제 또는 CADM1을 발현하는 암세포를 사멸시키는 암의 치료용 약학적 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for the treatment of cancer that kills cancer cells expressing cell therapy or CADM1, including the transformed T cells or natural killer cells.
상기와 같은 본 발명의 목적을 달성하기 위해서, 본 발명은 항원 결합 부위인 세포외 도메인; 막관통 도메인; 및 세포 내 신호전달 도메인을 포함하는 키메릭 항원 수용체(CAR)로서, 상기 항원 결합 도메인은 CADM1에 특이적으로 결합하는 CRTAM(class-I MHC-restricted T cell associated molecule)의 세포외 도메인을 포함하는 것인, 키메릭 항원 수용체를 제공한다.In order to achieve the object of the present invention as described above, the present invention is an antigen binding site, an extracellular domain; transmembrane domain; And a chimeric antigen receptor (CAR) comprising an intracellular signaling domain, wherein the antigen-binding domain comprises an extracellular domain of a class-I MHC-restricted T cell associated molecule (CRTAM) that specifically binds to CADM1. It provides a chimeric antigen receptor.
또한, 본 발명은 상기 키메릭 항원 수용체 단백질을 코딩하는 폴리 뉴클레오티드, 상기 폴리 뉴클레오티드를 포함하는 벡터 및 상기 벡터로 형질전환된 T 세포 또는 자연살생세포를 제공한다.In addition, the present invention provides a polynucleotide encoding the chimeric antigen receptor protein, a vector containing the polynucleotide, and T cells or natural killer cells transformed with the vector.
또한, 본 발명은 상기 형질전환된 T 세포 또는 자연살생세포를 포함하는, 세포 치료제 또는 CADM1을 발현하는 암세포를 사멸시키는 암의 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a cell therapy agent or a pharmaceutical composition for treating cancer that kills cancer cells expressing CADM1, including the transformed T cells or natural killer cells.
본 발명에 따른 키메릭 항원 수용체는 CADM1에 특이적으로 결합하는 CRTAM의 세포외 도메인을 포함한다. 따라서, 상기 키메릭 항원 수용체를 과발현시킬 수 있는 벡터로 형질전환된 세포독성 T 세포 또는 자연살생세포의 경우 CADM1을 발현하는 암종에 특이적으로 세포독성을 갖게 되므로 암 치료를 위한 면역세포 치료제로서 유용하게 이용될 수 있다.A chimeric antigen receptor according to the present invention comprises an extracellular domain of CRTAM that specifically binds to CADM1. Therefore, in the case of cytotoxic T cells or natural killer cells transformed with a vector capable of overexpressing the chimeric antigen receptor, they have cytotoxicity specifically to carcinoma expressing CADM1, so they are useful as immune cell therapeutics for cancer treatment can be used appropriately.
도 1은 본 발명의 일 실시예에 따른 키메릭 항원 수용체를 발현시키는 각 도메인의 cDNA 구역을 나타낸 모식도이다. 1 is a schematic diagram showing cDNA regions of each domain expressing a chimeric antigen receptor according to an embodiment of the present invention.
도 2a는 본 발명의 일 실시예에 따른 렌티바이럴 벡터의 모식도이다. 2A is a schematic diagram of a lentiviral vector according to an embodiment of the present invention.
도 2b는 본 발명의 일 실시예에 따른 레트로바이럴 벡터의 모식도이다.2B is a schematic diagram of a retroviral vector according to an embodiment of the present invention.
도 3은 본 발명의 일 실시예에 따른 세포독성 T 세포 또는 자연살생세포 표면에 발현하는 키메릭 항원 수용체의 모식도이다.3 is a schematic diagram of a chimeric antigen receptor expressed on the surface of cytotoxic T cells or natural killer cells according to an embodiment of the present invention.
도 4는 본 발명에서 제공하는 키메릭 항원 수용체의 표적인 CADM1을 발현하는 암세포(A549 세포주)에서 CADM1의 발현량을 유세포 분석을 이용해 측정한 결과를 나타낸 도이다. 4 is a diagram showing the results of measuring the expression level of CADM1 in cancer cells (A549 cell line) expressing CADM1, which is the target of the chimeric antigen receptor provided in the present invention, using flow cytometry.
도 5는 본 발명의 일 실시예에 따른 CRTAM이 CADM1을 인식하는지를 측정하기 위해 제조한 재조합 CRTAM 융합 단백질을 발현시키는 각 도메인의 cDNA 구역을 나타낸 모식도이다. 5 is a schematic diagram showing cDNA regions of each domain expressing a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
도 6은 본 발명의 일 실시예에 따른 CRTAM이 CADM1을 인식하는지를 측정하기 위해 제조한 재조합 CRTAM 융합 단백질을 발현시키는 발현 벡터(레트로바이럴 벡터)의 모식도이다.6 is a schematic diagram of an expression vector (retroviral vector) expressing a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
도 7은 본 발명의 일 실시예에 따른 CRTAM이 CADM1을 인식하는지를 측정하기 위해 제조한 재조합 CRTAM 융합 단백질의 모식도이다.7 is a schematic diagram of a recombinant CRTAM fusion protein prepared to measure whether CRTAM recognizes CADM1 according to an embodiment of the present invention.
도 8a는 제조한 재조합 CRTAM 융합 단백질을 Chinese hamster ovary (CHO) 세포에서 발현시킨 후, protein A column을 이용한 친화크로마토그래피(affinity chromatography)로 정제한 결과를 나타낸 도이다.8a is a diagram showing the result of expressing the prepared recombinant CRTAM fusion protein in Chinese hamster ovary (CHO) cells and then purifying it by affinity chromatography using a protein A column.
도 8b는 제조한 재조합 CRTAM 융합 단백질을 인간 IgG 중쇄의 불변 영역을 인식하는 항체(항 인간 IgG 중쇄 불변영역 항체, anti-human IgG Fc region antibody)를 이용한 웨스턴 블로팅으로 확인한 결과를 나타낸 도이다.Figure 8b is a view showing the result of Western blotting using an antibody (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody) recognizing the human IgG heavy chain constant region of the prepared recombinant CRTAM fusion protein.
도 8c는 제조한 재조합 CRTAM 융합 단백질이 타겟인 CADM1을 발현하는 암세포(A549 세포주)를 인식하는지 여부를 유세포 분석을 이용해 나타낸 도이다.8C is a diagram showing whether the prepared recombinant CRTAM fusion protein recognizes cancer cells (A549 cell line) expressing CADM1 as a target, using flow cytometry.
도 9a는 본 발명의 일 실시예에 따른 발현 벡터를 렌티바이러스 시스템을 이용하여 형질도입시킨 세포독성 T 세포(CRTAM CAR-T cell)에서의 GFP의 발현 비율을 비교한 결과를 나타낸 도이다.Figure 9a is a diagram showing the results of comparing the expression ratio of GFP in cytotoxic T cells (CRTAM CAR-T cells) transduced with an expression vector according to an embodiment of the present invention using a lentivirus system.
도 9b는 본 발명의 일 실시예에 따른 발현 벡터를 레트로바이러스 시스템을 이용하여 형질도입시킨 자연살생세포(CRTAM CAR-NK cell)에서의 CRTAM의 발현 비율을 비교한 결과를 나타낸 도이다.9B is a diagram showing the result of comparing the expression ratio of CRTAM in natural killer cells (CRTAM CAR-NK cells) transduced with an expression vector according to an embodiment of the present invention using a retroviral system.
도 10a는 본 발명의 일 실시예에 따른 세포독성 T 세포(CRTAM CAR-T cell) 또는 공벡터를 형질도입시킨 세포독성 T 세포(Mock T cell)를 작동 세포(effector cell)로 하여 CADM1을 발현하는 암세포(A549 세포주)에 대한 세포독성을 측정한 결과를 나타낸 도이다.Figure 10a is a cytotoxic T cell (CRTAM CAR-T cell) according to an embodiment of the present invention or a cytotoxic T cell (Mock T cell) transduced with an empty vector as effector cells to express CADM1 It is a diagram showing the results of measuring cytotoxicity against cancer cells (A549 cell line).
도 10b는 본 발명의 일 실시예에 따른 자연살생세포(CRTAM CAR-NK cell) 또는 공벡터를 형질도입시킨 자연살생세포(Mock NK cell)를 작동 세포(effector cell)로 하여 CADM1을 발현하는 암세포(A549 세포주)에 대한 세포독성을 측정한 결과를 나타낸 도이다.10B is cancer cells expressing CADM1 using CRTAM CAR-NK cells according to an embodiment of the present invention or Mock NK cells transduced with an empty vector as effector cells; It is a diagram showing the results of measuring cytotoxicity to (A549 cell line).
본 발명은 하나의 양태로서, 항원 결합 도메인; 막관통 도메인; 및 세포 내 신호전달 도메인을 포함하는 키메릭 항원 수용체(CAR)로서, 상기 항원 결합 도메인은 CADM1에 특이적으로 결합하는 CRTAM의 세포외 도메인을 포함하는, 키메릭 항원 수용체를 제공한다.The present invention as one aspect, an antigen-binding domain; transmembrane domain; and an intracellular signaling domain, wherein the antigen binding domain comprises an extracellular domain of CRTAM that specifically binds to CADM1.
본 발명에서 "키메릭 항원 수용체(Chimeric antigen receptor, CAR)"는 자연적으로 T 세포 또는 자연살생세포가 활성화하는데 필요한 항원 제시 세포(APC)나 항체의 매개 없이 원하는 항원에 결합하여 항원-항체 반응을 통해 T 세포의 활성화를 유도하고 해당 항원을 발현하는 세포를 공격할 수 있도록 하기 위해 T 세포 또는 자연살생세포에 발현시키기 위한 융합 단백질을 의미한다. 즉, T 세포 또는 자연살생세포에 발현 시 항원에 결합하여 이들 세포들의 활성화를 유도하는 단백질이라고 볼 수 있다. 이를 통해 면역 반응을 일으키고자 하는 세포에 특이적인 항원을 인식하는 단백질일 수 있으며, 상기 면역 반응을 일으키고자 하는 세포는 특정 조직에 존재하거나 병변을 일으킨 조직을 이루는 세포를 의미할 수 있다.In the present invention, the "Chimeric antigen receptor (CAR)" naturally binds to a desired antigen without the mediation of an antigen-presenting cell (APC) or antibody necessary for the activation of T cells or natural killer cells, resulting in an antigen-antibody reaction. It refers to a fusion protein to be expressed in T cells or natural killer cells in order to induce activation of T cells and attack cells expressing the corresponding antigen. That is, it can be seen as a protein that binds to an antigen when expressed in T cells or natural killer cells and induces the activation of these cells. Through this, it may be a protein that recognizes an antigen specific to a cell to cause an immune response, and the cell to cause an immune response may refer to a cell present in a specific tissue or forming a tissue that causes a lesion.
본 발명의 일 실시예에 따른 키메릭 항원 수용체 단백질은 기능적 동등물을 포함할 수 있다. '기능적 동등물’이란 아미노산의 부가, 치환, 또는 결실의 결과, 상기 키메릭 항원 수용체 단백질의 아미노산 서열과 적어도 70% 이상, 바람직하게는 80% 이상, 더욱 바람직하게는 90% 이상, 더더욱 바람직하게는 95% 이상의 서열 상동성을 갖는 것으로, 실질적으로 동질의 생리활성을 나타내는 단백질을 말한다. '실질적으로 동질의 생리활성’이란 CADM1에 특이적으로 결합할 수 있는 활성을 가진 것을 의미한다.Chimeric antigen receptor proteins according to one embodiment of the present invention may include functional equivalents. 'Functional equivalent' means at least 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably, the amino acid sequence of the chimeric antigen receptor protein as a result of addition, substitution, or deletion of amino acids. has a sequence homology of 95% or more, and refers to a protein that exhibits substantially the same physiological activity. 'Substantially homogeneous physiological activity' means having an activity that can specifically bind to CADM1.
본 발명은 또한 키메릭 항원 수용체의 단편, 유도체 및 유사체(analogues)를 포함한다. 본원에 사용된, 용어 '단편', '유도체' 및 '유사체'는 본 발명의 키메릭 항원 수용체 단백질과 실질적으로 같은 생물학적 기능 또는 활성을 보유하는 폴리펩티드를 말한다. 본 발명의 단편, 유도체 및 유사체는 (1) 하나 이상의 보존적(conservative) 또는 비보존적 아미노산 잔기(바람직하게는 보존적 아미노산 잔기)가 치환된 폴리펩티드(상기 치환된 아미노산 잔기는 유전 암호에 의해 암호화될 수도, 되지 않을 수도 있다) 또는 (2) 하나 이상의 아미노산 잔기에서 치환기(들)를 가지는 폴리펩티드, 또는 (3) 또 다른 화합물(폴리펩티드의 반감기를 연장할 수 있는 화합물, 예를 들면 폴리에틸렌 글리콜)과 결합된 성숙 폴리펩티드로부터 유래된 폴리펩티드, 또는 (4) 부가적인 아미노산 서열(예를 들면, 선도 서열, 분비 서열, 상기 폴리펩티드를 정제하는데 사용된 서열, 프로테이노젠(proteinogen) 서열 또는 융합 단백질)과 결합된 상기 폴리펩티드로부터 유래된 폴리펩티드일 수 있다. 본 발명에 정의된 상기 단편, 유도체 및 유사체는 당업자에 잘 알려져있다.The present invention also includes fragments, derivatives and analogs of chimeric antigen receptors. As used herein, the terms 'fragment', 'derivative' and 'analogue' refer to a polypeptide that retains substantially the same biological function or activity as the chimeric antigen receptor protein of the invention. Fragments, derivatives and analogs of the present invention include (1) a polypeptide in which one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted (the substituted amino acid residues are encoded by the genetic code). (2) a polypeptide having substituent(s) on one or more amino acid residues, or (3) another compound (a compound capable of extending the half-life of a polypeptide, such as polyethylene glycol) a polypeptide derived from the combined mature polypeptide, or (4) an additional amino acid sequence (e.g., a leader sequence, a secreted sequence, a sequence used to purify the polypeptide, a proteinogen sequence, or a fusion protein) and It may be a polypeptide derived from the polypeptide to which it is linked. Such fragments, derivatives and analogues as defined herein are well known to those skilled in the art.
CRTAM(class-I MHC-restricted T cell associated molecule)은 내재성 막 단백질(integral membrane protein)로, 활성화된 림프구에서 발현되며, 세포독성 T 림프구(cytotoxic T lymphocyte)나 자연살생세포(natural killer cell, NK cell)가 표적세포의 세포자살을 유도할 때, 표적세포에 좀 더 잘 부착되게 하는 역할을 한다. 또한, 보조 T 림프구(helper T cell)의 IFN-η와 IL-22 분비량을 촉진시킨다.CRTAM (class-I MHC-restricted T cell associated molecule) is an integral membrane protein, which is expressed in activated lymphocytes, such as cytotoxic T lymphocytes or natural killer cells, When NK cell) induces apoptosis of target cells, it serves to better adhere to target cells. In addition, it promotes the secretion of IFN-η and IL-22 by helper T cells.
상기 항원 결합 도메인은 신호전달을 증폭하기 위해 CRTAM의 세포외 도메인 한 쌍을 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)에 각각 연결시킨 이합체(dimer) 형태일 수 있으나, 이에 제한되는 것은 아니다.The antigen-binding domain may be in the form of a dimer in which a pair of extracellular domains of CRTAM are linked to a human immunoglobulin G 1 heavy chain constant region , respectively, to amplify signal transduction. It is not limited.
상기 CRTAM의 세포외 도메인은 서열번호 1 또는 이와 95% 이상의 상동성을 나타내는 아미노산 서열로 이루어진 것일 수 있으나, 이에 제한되는 것은 아니다.The extracellular domain of CRTAM may consist of SEQ ID NO: 1 or an amino acid sequence showing 95% or more homology thereto, but is not limited thereto.
상기 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)은 서열번호 2 또는 이와 95% 이상의 상동성을 나타내는 아미노산 서열로 이루어진 것일 수 있으나, 이에 제한되는 것은 아니다.The human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region) may consist of SEQ ID NO: 2 or an amino acid sequence exhibiting 95% or more homology thereto, but is not limited thereto.
본 발명에서 "CADM1"은 다양한 상피 조직과 면역세포에서 발현하는 것으로 알려져 있으며, 몇몇 암종에서는 발현이 증가되어 암종의 분자 표지 또는 치료용 타겟으로 사용된다. 지금까지 알려진 CADM1을 발현하는 암종은 비소세포 폐암(non-small lung cancer cell, NSCLC), 폐 선암종(lung adenocarcinoma), 사람T세포림프친화바이러스 1형에 감염된 T 세포 백혈병(HTLV-1-infected T cell leukemia), 성인T세포백혈병(adult T cell leukemia), 난소암(ovarian cancer), 유방암(breast cancer), 자궁경부암(cervical cancer), 전립선암(prostate cancer), 상인두암(nasopharyngeal carcinoma), 위암(stomach cancer) 및 췌장암(pancreatic cancer) 등이 있다.In the present invention, "CADM1" is known to be expressed in various epithelial tissues and immune cells, and its expression is increased in some carcinomas, so that it is used as a molecular marker of carcinoma or as a target for treatment. The known CADM1-expressing carcinomas include non-small lung cancer cell (NSCLC), lung adenocarcinoma, and human T-cell lymphotropic virus type 1-infected T-cell leukemia (HTLV-1-infected T-cell). cell leukemia), adult T cell leukemia, ovarian cancer, breast cancer, cervical cancer, prostate cancer, nasopharyngeal carcinoma, stomach cancer (stomach cancer) and pancreatic cancer.
본 발명에서, "막관통 도메인(Transmembrane domain)"은 CRTAM의 세포외 도메인과 보조자극, 필수 신호전달 도메인을 세포막 사이로 연결하는 부위이며, 세포내 신호 전달 도메인은 항원 결합 도메인의 결합에 의해 면역 세포의 면역반응을 활성화시키는 부위를 의미한다.In the present invention, the "transmembrane domain" is a site that connects the extracellular domain of CRTAM and the co-stimulatory and essential signaling domains between the cell membrane, and the intracellular signaling domain is an immune cell by binding of the antigen-binding domain. refers to the site that activates the immune response of
일 실시예에서, 상기 막관통 도메인은 CD28, CD3엡실론, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 및 CD154로 이루어진 군으로부터 선택되는 1종 이상을 포함할 수 있으며, 상기 CD8은 CD8α 또는 CD8β일 수 있으며, 바람직하게는 CD8α의 막관통 도메인일 수 있고, 이는 서열번호 3 또는 이와 95% 이상의 상동성을 나타내는 아미노산 서열로 이루어진 것일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment, the transmembrane domain is selected from the group consisting of CD28, CD3epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154 It may include one or more types, and the CD8 may be CD8α or CD8β, preferably a transmembrane domain of CD8α, which may consist of SEQ ID NO: 3 or an amino acid sequence showing 95% or more homology thereto. However, it is not limited thereto.
본 발명에서 "세포내 신호전달 도메인"은 항원 결합 도메인에 결합에 의해 면역 세포의 면역반응을 활성화시키는 부위를 의미한다. 일 실시예에서, 상기 세포내 도메인은 CD28, 4-1BB, CD3 제타(zeta) 또는 이들의 조합일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, "intracellular signaling domain" means a site that activates the immune response of immune cells by binding to an antigen binding domain. In one embodiment, the intracellular domain may be CD28, 4-1BB, CD3 zeta, or a combination thereof, but is not limited thereto.
본 발명에 따른 키메릭 항원 수용체는 세포내 신호전달 도메인으로 CD28, 4-1BB, CD3 제타(zeta)를 이용함으로써 높은 활성으로 암 세포, 특히 CADM1을 발현하는 암세포에 대한 사멸 효과를 나타낼 수 있다.The chimeric antigen receptor according to the present invention uses CD28, 4-1BB, and CD3 zeta as intracellular signaling domains, thereby exhibiting a high-activity killing effect on cancer cells, particularly cancer cells expressing CADM1.
상기 CD28은 서열번호 4 또는 이와 70% 이상, 바람직하게는 80% 이상, 더욱 바람직하게는 90% 이상, 더더욱 바람직하게는 95% 이상의 서열 상동성을 갖는 것으로, 서열번호 4로 표시되는 아미노산 서열과 실질적으로 동등한 기능을 나타내는 아미노산 서열로 이루어질 수 있고, 4-1BB(CD137)은 서열번호 5 또는 이와 70% 이상, 바람직하게는 80% 이상, 더욱 바람직하게는 90% 이상, 더더욱 바람직하게는 95% 이상의 서열 상동성을 갖는 것으로, 서열번호 5로 표시되는 아미노산 서열과 실질적으로 동등한 기능을 나타내는 아미노산 서열로 이루어질 수 있으며, CD3 제타(zeta)는 NK 세포 활성화 도메인으로 기능하며, 서열번호 6 또는 이와 70% 이상, 바람직하게는 80% 이상, 더욱 바람직하게는 90% 이상, 더더욱 바람직하게는 95% 이상의 서열 상동성을 갖는 것으로, 서열번호 6으로 표시되는 아미노산 서열과 실질적으로 동등한 기능을 나타내는 아미노산 서열로 이루어 질 수 있다.The CD28 has SEQ ID NO: 4 or 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more sequence homology thereto, and the amino acid sequence represented by SEQ ID NO: 4 4-1BB (CD137) is SEQ ID NO: 5 or 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more. Having the above sequence homology, it may consist of an amino acid sequence showing a function substantially equivalent to the amino acid sequence represented by SEQ ID NO: 5, CD3 zeta functions as an NK cell activation domain, and SEQ ID NO: 6 or 70 % or more, preferably 80% or more, more preferably 90% or more, still more preferably 95% or more sequence homology, and an amino acid sequence that exhibits a function substantially equivalent to that of the amino acid sequence represented by SEQ ID NO: 6 It can be done.
상기 항원 결합 도메인은 CD8α 신호 펩타이드를 포함할 수 있으며, 상기 CD8α 신호 펩타이드는 서열번호 7 또는 이와 95% 이상의 상동성을 나타내는 아미노산 서열로 이루어진 것일 수 있으나, 이에 제한되는 것은 아니다.The antigen-binding domain may include a CD8α signal peptide, and the CD8α signal peptide may consist of SEQ ID NO: 7 or an amino acid sequence showing 95% or more homology thereto, but is not limited thereto.
또한, 본 발명에 따른 다른 하나의 양태로서, 상기 키메릭 항원 수용체 단백질을 코딩하는 폴리 뉴클레오티드를 제공한다.Also, as another aspect according to the present invention, a polynucleotide encoding the chimeric antigen receptor protein is provided.
본 발명의 항원 수용체를 암호화하는 폴리 뉴클레오티드는 코돈의 축퇴성 (degeneracy)으로 인하여 또는 상기 항원 수용체를 발현시키고자 하는 생물에서 선호되는 코돈을 고려하여, 코딩영역으로부터 발현되는 항원 수용체의 아미노산 서열을 변화시키지 않는 범위 내에서 코딩영역에 다양한 변형이 이루어질 수 있고, 코딩영역을 제외한 부분에서도 유전자의 발현에 영향을 미치지 않는 범위 내에서 다양한 변형 또는 수식이 이루어질 수 있으며, 그러한 변형 유전자 역시 본 발명의 범위에 포함됨을 당업자는 잘 이해할 수 있을 것이다. 즉, 본 발명의 폴리 뉴클레오티드는 이와 동등한 활성을 갖는 단백질을 코딩하는 한, 하나 이상의 핵산 염기가 치환, 결실, 삽입 또는 이들의 조합에 의해 변이될 수 있으며, 이들 또한 본 발명의 범위에 포함된다.The polynucleotide encoding the antigen receptor of the present invention changes the amino acid sequence of the antigen receptor expressed from the coding region due to codon degeneracy or in consideration of codons preferred in organisms intended to express the antigen receptor. Various modifications may be made to the coding region within a range not specified, and various modifications or modifications may be made to a part other than the coding region within a range that does not affect gene expression, and such modified genes are also within the scope of the present invention. It will be well understood by those skilled in the art. That is, as long as the polynucleotide of the present invention encodes a protein having an activity equivalent thereto, one or more nucleic acid bases may be mutated by substitution, deletion, insertion, or a combination thereof, and these are also included in the scope of the present invention.
또한, 본 발명에 따른 또 다른 하나의 양태로서 상기 폴리 뉴클레오티드를 포함하는 벡터, 상기 벡터로 형질전환된 세포를 제공한다.In addition, as another aspect of the present invention, a vector containing the polynucleotide and a cell transformed with the vector are provided.
본 발명에서 사용되는 벡터는 당 분야에 공지된 벡터를 다양하게 사용할 수 있고, 상기 항원 수용체를 생산하고자 하는 숙주세포의 종류에 따라 프로모터(promoter), 종결자(terminator), 인핸서(enhancer) 등과 같은 발현조절 서열, 막 표적화 또는 분비를 위한 서열 등을 적절히 선택하고 목적에 따라 다양하게 조합할 수 있다. 본 발명의 벡터는 플라스미드 벡터, 코즈미드 벡터, 박테리오 파아지 벡터 및 바이러스 벡터 등을 포함하나 이에 제한되지 않는다. 적합한 벡터는 프로모터, 오퍼레이터, 개시코돈, 종결코돈, 폴리아데닐화 시그널 및 인핸서 같은 발현 조절 엘리먼트 외에도 막 표적화 또는 분비를 위한 시그널 서열 또는 리더 서열을 포함하며 목적에 따라 다양하게 제조될 수 있다.Vectors used in the present invention may use various vectors known in the art, and depending on the type of host cell to produce the antigen receptor, a promoter, terminator, enhancer, etc. Expression control sequences, sequences for membrane targeting or secretion, etc. may be appropriately selected and combined in various ways depending on the purpose. Vectors of the present invention include, but are not limited to, plasmid vectors, cosmid vectors, bacteriophage vectors and viral vectors. Suitable vectors include expression control elements such as promoters, operators, initiation codons, stop codons, polyadenylation signals and enhancers, as well as signal sequences or leader sequences for membrane targeting or secretion, and may be prepared in various ways depending on the purpose.
본 발명에서는 바람직한 일 실시예로서, 렌티-바이러스용 벡터 또는 레트로-바이러스용 벡터를 사용할 수 있으며, 본 발명의 하기 실시예에서는 pCDH-CMV-MCS-EF1-copGFP 벡터(렌티-바이러스용 벡터)와 pLNCX2(레트로-바이러스용 벡터)를 사용하였으나, 이에 제한되는 것은 아니다.In the present invention, as a preferred embodiment, a lenti-virus vector or a retro-virus vector can be used, and in the following embodiment of the present invention, the pCDH-CMV-MCS-EF1-copGFP vector (lenti-virus vector) and pLNCX2 (retro-viral vector) was used, but is not limited thereto.
일 일시예에서, CADM1에 특이적으로 결합하는 키메릭 항원 수용체를 상기 벡터를 통해 세포에 도입하여 세포를 형질전환 시킬 수 있다. 상기 세포는 T 세포, 종양 침윤 림프구, B 세포, 자연살생세포, 또는 NKT 세포일 수 있으며, 바람직하게는, 세포독성 T 세포(cytotoxic T cell)또는 자연살생세포 일 수 있다. 상기 세포는 골수, 말초혈액, 말초혈액단핵세포 또는 제대혈로부터 얻거나 제조될 수 있으며, 세포는 인간 세포일 수 있으나, 이에 제한되는 것은 아니다.In one embodiment, cells may be transformed by introducing a chimeric antigen receptor that specifically binds to CADM1 into cells through the vector. The cells may be T cells, tumor-infiltrating lymphocytes, B cells, natural killer cells, or NKT cells, and preferably may be cytotoxic T cells or natural killer cells. The cells may be obtained or prepared from bone marrow, peripheral blood, peripheral blood mononuclear cells or umbilical cord blood, and the cells may be human cells, but are not limited thereto.
상기와 같이 본 발명의 키메릭 항원 수용체가 도입되어 형질전환된 세포는 CADM1을 항원으로 인식하고 이와 강하게 결합하는 특징을 가진다.As described above, cells transformed by introducing the chimeric antigen receptor of the present invention recognize CADM1 as an antigen and have a characteristic of strongly binding thereto.
본 발명에서, "키메릭 항원 수용체 발현 T 세포(chimeric antigen receptor T cell, 이하 간략하게 'CAR-T 세포'라 약칭함)" 또는 "키메릭 항원 수용체 발현 NK 세포(chimeric antigen receptor NK cell, 이하 간략하게 'CAR-NK 세포'라 약칭함)"란 정상의 T 세포 또는 자연살생세포를 형질도입 등의 방법으로 본래의 T 세포 수용체 또는 NK cell 수용체가 아닌 암세포에 특이적으로 반응하는 키메라 항원 수용체를 발현하는 T 세포 또는 NK cell을 의미한다. 이 수용체를 갖는 T 세포 또는 NK 세포는 표적세포(target cell)의 세포자살을 유도하여 세포독성을 나타낸다.In the present invention, "chimeric antigen receptor T cell (hereinafter, simply abbreviated as 'CAR-T cell')" or "chimeric antigen receptor expressing NK cell (chimeric antigen receptor NK cell, hereinafter) Shortly abbreviated as 'CAR-NK cell')" is a chimeric antigen receptor that specifically responds to cancer cells rather than the original T cell receptor or NK cell receptor by a method such as transduction of normal T cells or natural killer cells. refers to T cells or NK cells expressing T cells or NK cells having this receptor exhibit cytotoxicity by inducing apoptosis of target cells.
본 발명에 있어서, 특히 CAR-T 세포나 CAR-NK 세포는 세포독성 T 세포(cytotoxic T cell) 또는 NK cell에 본 발명의 키메릭 항원 수용체가 도입된 세포일 수 있다. 상기 세포는 CAR-T 치료제의 기존 장점인 항암 특이적 표적 치료의 장점을 가지며, 특히, 본 발명의 CAR-T 세포나 CAR-NK 세포는 CADM1을 발현하는 암세포를 인지하여 효과적으로 파괴할 수 있다.In the present invention, in particular, CAR-T cells or CAR-NK cells may be cells into which the chimeric antigen receptor of the present invention is introduced into cytotoxic T cells or NK cells. The cells have the advantage of anticancer-specific targeted therapy, which is an existing advantage of CAR-T therapeutics. In particular, the CAR-T cells or CAR-NK cells of the present invention can recognize and effectively destroy cancer cells expressing CADM1.
따라서 본 발명은 또 다른 하나의 양태로서, 상기 세포를 포함하는 세포 치료제 또는 이를 유효성분으로 포함하는 암의 치료용 약학적 조성물을 제공한다. Accordingly, in another aspect, the present invention provides a cell therapeutic agent containing the cells or a pharmaceutical composition for treating cancer containing the same as an active ingredient.
본 발명에서, "세포 치료제"는 개체로부터 분리, 배양 및 특수한 조작을 통해 제조된 세포 및 조직으로 치료의 목적으로 사용되는 의약품으로서, 세포 혹은 조직의 기능을 복원시키기 위하여 살아있는 자가, 동종, 또는 이종 세포를 체외에서 증식 선별하거나 다른 방법으로 세포의 생물학적 특성을 변화시키는 등의 일련의 행위를 통하여 치료의 목적으로 사용되는 의약품을 의미한다.In the present invention, "cell therapy" refers to cells and tissues prepared from a subject through isolation, culture, and special manipulation, and is a pharmaceutical product used for the purpose of treatment, and is a living autologous, allogeneic, or xenogeneic living agent to restore the function of a cell or tissue. It refers to drugs used for the purpose of treatment through a series of actions, such as proliferation and selection of cells in vitro or altering the biological characteristics of cells in other ways.
본 발명의 용어, "치료"는 상기 조성물의 투여에 의해 암에 의한 증세가 호전되거나 이롭게 변경되는 모든 행위를 의미한다.As used herein, the term "treatment" refers to all activities in which symptoms caused by cancer are improved or beneficially changed by administration of the composition.
상기 조성물은 약학적으로 허용 가능한 담체를 포함할 수 있다.The composition may include a pharmaceutically acceptable carrier.
상기 "약학적으로 허용 가능한 담체"란 생물체를 자극하지 않으면서, 주입되는 화합물의 생물학적 활성 및 특성을 저해하지 않는 담체 또는 희석제를 의미할 수 있다. 본 발명에 사용 가능한 상기 담체의 종류는 특별히 제한되지 아니하며 당해 기술 분야에서 통상적으로 사용되고 약학적으로 허용되는 담체라면 어느 것이든 사용할 수 있다. 상기 담체의 비제한적인 예로는, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사 용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 등을 들 수 있다. 이들은 단독으로 사용되거나 2종 이상을 혼합하여 사용될 수 있다.The "pharmaceutically acceptable carrier" may refer to a carrier or diluent that does not inhibit the biological activity and properties of the compound to be injected without irritating living organisms. The type of the carrier that can be used in the present invention is not particularly limited, and any carrier commonly used in the art and pharmaceutically acceptable can be used. Non-limiting examples of the carrier include saline, sterile water, Ringer's solution, buffered saline, albumin injection solution, dextrose solution, maltodextrin solution, glycerol, ethanol, and the like. These may be used alone or in combination of two or more.
약학적으로 허용 가능한 담체를 포함하는 상기 조성물은 경구 또는 비경구의 여러 가지 제형일 수 있다. 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다.The composition containing a pharmaceutically acceptable carrier may be in various oral or parenteral formulations. When formulated, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants, and surfactants.
상세하게는, 경구 투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 상기 화합물에 적어도 하나 이상의 부형제, 예를 들면, 전분, 칼슘카보네이트, 수크로오스, 락토오스, 젤라틴 등을 섞어 조제될 수 있다. 또한, 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용될 수 있다. 경구를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 흔히 사용되는 단순 희석제인 물, 액체 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제 및 좌제가 포함된다. 비수성용제, 현탁제로는 프로필렌글리콜, 폴리에틸렌 글리콜, 올리브 오일과 같은 식물성 오일, 에틸올레이트와 같은 주사 가능한 에스테르 등이 사용될 수 있다. 좌제의 기제로는 위텝솔, 마크로골, 트윈 61, 카카오지, 라우린지, 글리세로젤라틴 등이 사용될 수 있다.Specifically, solid preparations for oral administration include tablets, pills, powders, granules, capsules, and the like, and these solid preparations contain at least one excipient such as starch, calcium carbonate, sucrose, and lactose in the compound. , can be prepared by mixing gelatin, etc. In addition to simple excipients, lubricants such as magnesium stearate and talc may also be used. Liquid preparations for oral use include suspensions, solutions for internal use, emulsions, and syrups. In addition to water and liquid paraffin, which are commonly used simple diluents, various excipients such as wetting agents, sweeteners, aromatics, and preservatives may be included. there is. Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried formulations, and suppositories. Propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate may be used as non-aqueous solvents and suspending agents. As a base for the suppository, Witepsol, Macrogol, Tween 61, cacao butter, laurin paper, glycerogelatin, and the like may be used.
상기 조성물은 약학적으로 유효한 양으로 투여할 수 있다.The composition can be administered in a pharmaceutically effective amount.
상기 "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분한 양을 의미하며, 유효 용량 수준은 개체 종류 및 중증도, 연령, 성별, 감염된 바이러스 종류, 약물의 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다.The "pharmaceutically effective amount" means an amount sufficient to treat a disease with a reasonable benefit / risk ratio applicable to medical treatment, and the effective dose level is dependent on the type and severity of the subject, age, sex, infected virus type, and drug activity, sensitivity to the drug, time of administration, route of administration and rate of excretion, duration of treatment, factors including concomitantly used drugs and other factors well known in the medical arts.
상기 투여는 어떠한 적절한 방법으로 환자에게 본 발명의 조성물을 도입하는 것을 의미하며, 상기 조성물의 투여 경로는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 투여될 수 있다. 복강 내 투여, 정맥내 투여, 근육 내 투여, 피하 투여, 피내 투여, 경구 투여, 국소 투여, 비 내 투여될 수 있으나, 이에 제한되지는 않는다.The administration means introducing the composition of the present invention to a patient by any suitable method, and the administration route of the composition may be administered through any general route as long as it can reach the target tissue. Intraperitoneal administration, intravenous administration, intramuscular administration, subcutaneous administration, intradermal administration, oral administration, topical administration, intranasal administration may be administered, but is not limited thereto.
본 발명의 조성물을 매일 투여 또는 간헐적으로 투여해도 좋고, 1일당 투여 횟수는 1회 또는 2~3회로 나누어 투여하는 것이 가능하다. 두 유효성분이 각각 단제인 경우의 투여횟수는 같은 횟수여도 좋고, 다른 횟수로 해도 된다. 또한, 본 발명의 조성물은 CADM1을 발현하는 암의 치료를 위하여 단독으로, 또는 다른 약물 치료와 병용하여 사용할 수 있다. 상기 요소를 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 당업자에 의해 용이하게 결정될 수 있다.The composition of the present invention may be administered daily or intermittently, and the number of administrations per day may be administered once or divided into 2 to 3 times. The number of administrations when the two active ingredients are each a single agent may be the same or may be different. In addition, the composition of the present invention can be used alone or in combination with other drug treatments for the treatment of CADM1-expressing cancer. It is important to administer the amount that can obtain the maximum effect with the minimum amount without side effects in consideration of all the above factors, and can be easily determined by those skilled in the art.
상기 개체란, CADM1을 발현하는 암이 발병하였거나 발병할 수 있는 인간과, 원숭이, 소, 말, 양, 돼지, 닭, 칠면조, 메추라기, 고양이, 개, 마우스, 쥐, 토끼 또는 기니아 피그를 포함한 모든 동물을 의미한다. 본 발명의 약학적 조성물을 개체에게 투여함으로써 상기 질환을 효과적으로 치료할 수 있다면 개체의 종류는 제한 없이 포함된다.The subject refers to all humans, including humans, monkeys, cows, horses, sheep, pigs, chickens, turkeys, quails, cats, dogs, mice, rats, rabbits, or guinea pigs that have or may develop cancer expressing CADM1. means animals. The type of subject is included without limitation as long as the disease can be effectively treated by administering the pharmaceutical composition of the present invention to the subject.
본 발명에 있어 치료 대상이 되는 CADM1을 발현하는 암의 종류로는 비소세포 폐암(non-small lung cancer cell, NSCLC), 폐 선암종(lung adenocarcinoma), 사람T세포림프친화바이러스 1형에 감염된 T 세포 백혈병 (HTLV-1-infected T cell leukemia), 성인T세포백혈병(adult T cell leukemia), 난소암(ovarian cancer), 유방암(breast cancer), 자궁경부암(cervical cancer), 전립선암(prostate cancer), 상인두암(nasopharyngeal carcinoma), 위암(stomach cancer) 및 췌장암(pancreatic cancer) 등을 예시할 수 있으나, 이에 한정되는 것은 아니다.Types of cancer expressing CADM1 to be treated in the present invention include non-small lung cancer cell (NSCLC), lung adenocarcinoma, and T cells infected with human T-cell lymphotropic virus type 1. Leukemia (HTLV-1-infected T cell leukemia), adult T cell leukemia, ovarian cancer, breast cancer, cervical cancer, prostate cancer, Examples include nasopharyngeal carcinoma, stomach cancer and pancreatic cancer, but are not limited thereto.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로서, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다.Hereinafter, the present invention will be described in more detail through examples. These examples are only for explaining the present invention in more detail, and it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples according to the gist of the present invention. .
실시예 1. 유전자 합성 방법에 의한 키메릭 항원 수용체Example 1. Chimeric antigen receptor by gene synthesis method 유전자의 클로닝cloning of genes
본 발명의 키메릭 항원 수용체를 제조하기 위해서 CRTAM의 세포외 도메인 한 쌍을 각각 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)에 연결시킨 항원 인식 부위와 CD8α의 막관통 도메인, CD28, 4-1BB, CD3 제타(zeta) 각각의 세포내 도메인 부분의 단백질 암호화 서열을 유전자 데이터베이스(database)에서 확인하였다. In order to prepare the chimeric antigen receptor of the present invention, an antigen recognition site in which a pair of extracellular domains of CRTAM are linked to a human immunoglobulin G 1 heavy chain constant region, and a transmembrane domain of CD8α , Protein coding sequences of the intracellular domains of each of CD28, 4-1BB, and CD3 zeta were identified in a gene database.
그 후 상기 도메인들을 이루는 염기서열의 종결 코돈 부분을 제외한 나머지 서열들을 하나로 이어 유전자 합성을 진행하였다. 유전자 합성의 정확도는 단백질 발현 플라스미드를 제작한 후 시퀀싱을 통하여 확인하였다. 상기 키메릭 항원 수용체를 발현시키는 각 도메인의 cDNA 구역을 나타낸 모식도를 도 1에 나타내었다.Thereafter, gene synthesis was performed by connecting the remaining sequences except for the stop codon portion of the base sequence constituting the domains into one. The accuracy of gene synthesis was confirmed through sequencing after constructing a protein expression plasmid. A schematic diagram showing the cDNA region of each domain expressing the chimeric antigen receptor is shown in FIG. 1 .
실시예 2. 키메릭 항원 수용체Example 2. Chimeric Antigen Receptor 단백질 발현 플라스미드 제조Construction of protein expression plasmids
CADM1을 인식하고, 인간 이뮤노글로불린 G1 중쇄 불변 영역과 신호전달 단백질 도메인을 융합시킨 재조합 단백질을 세포독성 T 세포와 자연살생세포에 발현시키기 위해, 해당 유전자를 렌티바이러스 유래 발현벡터인 pCDH-CMV-MCS-EF1-copGFP(System Biosciences) 또는 레트로바이러스 유래 발현벡터인 pLNCX2(Addgene)에 클로닝하였다. In order to recognize CADM1 and express the recombinant protein fused with the human immunoglobulin G 1 heavy chain constant region and the signaling protein domain in cytotoxic T cells and natural killer cells, the corresponding gene was used as a lentivirus-derived expression vector, pCDH-CMV -MCS-EF1-copGFP (System Biosciences) or retrovirus-derived expression vector pLNCX2 (Addgene) was cloned.
먼저, pCDH-CMV-MCS-EF1-copGFP 벡터에 클로닝하기 위해서 5′말단에 제한효소 XbaI의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응으로 제한효소 절단 부위를 만들고, 3′말단에도 제한효소 NotI의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응으로 제한효소 절단 부위를 만들었다. 그 후 중합효소연쇄반응으로 인해 제한효소 절단 부위를 가진 유전자를 5′말단은 XbaI을 처리하였으며, 3′말단은 NotI을 처리하였다. 그리고 발현 벡터의 다중클로닝 자리를 XbaI과 NotI을 처리하여 유전자가 삽입될 수 있게 만들었다. 제한효소가 처리된 유전자와 발현 벡터를 섞어 준 다음, 결합 효소(ligase)를 처리하여 연결하였다. First, in order to clone into the pCDH-CMV-MCS-EF1-copGFP vector, a primer that creates a restriction enzyme XbaI cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is created by polymerase chain reaction, and a restriction enzyme cleavage site is also made at the 3' end. A restriction enzyme cleavage site was prepared by synthesizing a primer that creates the cleavage site sequence of the enzyme NotI and polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with XbaI, and the 3' end was treated with NotI. In addition, the multicloning site of the expression vector was treated with XbaI and NotI so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
다음, pLNCX2 벡터에 클로닝하기 위해서 5′말단에 제한효소 Bgl II의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응으로 제한효소 절단 부위를 만들고, 3′말단에도 제한효소 NotI의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응으로 제한효소 절단 부위를 만들었다. 그 후 중합효소연쇄반응으로 인해 제한효소 절단 부위를 가진 유전자를 5′말단은 BglII를 처리하였으며, 3′말단은 NotI을 처리하였다. 그리고 발현 벡터의 다중클로닝 자리를 BglII와 NotI을 처리하여 유전자가 삽입될 수 있게 만들었다. 제한효소가 처리된 유전자와 발현 벡터를 섞어 준 다음, 결합 효소(ligase)를 처리하여 연결하였다. Next, in order to clone into the pLNCX2 vector, a primer that creates a restriction enzyme Bgl II cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is made by polymerase chain reaction, and a restriction enzyme NotI cleavage site sequence is also added at the 3' end. A primer was synthesized to make a restriction enzyme cleavage site by polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with BglII, and the 3' end was treated with NotI. Then, the multicloning site of the expression vector was treated with BglII and NotI so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
그 결과, CADM1을 인식하고, 인간 이뮤노글로불린 G1 중쇄 불변 영역과 신호전달 단백질 도메인을 융합시킨 단백질(도 3 참조)을 발현하는 재조합 벡터 CRTAM.CAR-T.pCDH-CMV-MCS-EF1-copGFP 및 CRTAM.CAR-NK.pLNCX2를 완성하였다(도 2a 및 도 2b 참조).As a result, a recombinant vector CRTAM.CAR-T.pCDH-CMV-MCS-EF1- that recognizes CADM1 and expresses a protein in which a human immunoglobulin G 1 heavy chain constant region and a signaling protein domain are fused (see FIG. 3) is expressed. copGFP and CRTAM.CAR-NK.pLNCX2 were completed (see Figs. 2a and 2b).
실시예 3. CADM1을 발현하는 사람 세포주 screeningExample 3. Human cell line screening expressing CADM1
CADM1을 세포 표면에 발현하는 사람 세포주를 screening하기 위해서, 폐 선암종(lung adenocarcinoma) 유래 세포주인 A549 (ATCC)에서 CADM1의 발현을 확인하였다. 발현 확인을 위해 형광단백질이 부착된 CADM1과 결합이 가능한 항체(Biobyt)를 상기 세포주들에 처리한 후, 유세포 분석(fluorescence-activated cell sorting)을 이용하였다. 분석결과, A549 세포주에서 CADM1을 발현하는 것을 확인하였다(도 4 참조).To screen human cell lines expressing CADM1 on the cell surface, expression of CADM1 was confirmed in A549 (ATCC), a cell line derived from lung adenocarcinoma. To confirm expression, the cell lines were treated with an antibody (Biobyt) capable of binding to CADM1 to which a fluorescent protein was attached, and then fluorescence-activated cell sorting was used. As a result of the analysis, it was confirmed that CADM1 was expressed in the A549 cell line (see FIG. 4).
상기 결과를 근거로 CADM1을 발현하는 A549 세포주를 CRTAM.CAR-T 세포 또는 CRTAM.CAR-NK 세포의 타겟 세포로 사용하였다.Based on the above results, the A549 cell line expressing CADM1 was used as a target cell for CRTAM.CAR-T cells or CRTAM.CAR-NK cells.
실시예Example 4. 유전자 합성 방법에 의한 수용성 재조합 4. Soluble recombination by gene synthesis method CRTAMCRTAM 융합 단백질 유전자의 클로닝 Cloning of fusion protein genes
CRTAM이 CADM1에 결합하는 것을 확인하기 위해 수용성 재조합 CRTAM 융합 단백질을 제조하였다. 우선 CRTAM의 세포외 도메인(extracellular domain) 각각을 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)에 연결시킨 단백질 암호화 서열을 유전자 데이터베이스(database)에서 확인하였다. 그리고 상기 도메인들을 이루는 염기서열의 종결 코돈 부분을 제외한 나머지 서열들을 하나로 이어 유전자 합성을 진행하였다. 유전자 합성의 정확도는 단백질 발현 플라스미드를 제작한 후 시퀀싱을 통하여 확인하였다. 상기 재조합 단백질을 발현시키는 각 도메인의 cDNA 구역을 나타낸 모식도를 도 5에 나타내었다.To confirm that CRTAM binds to CADM1, a water-soluble recombinant CRTAM fusion protein was prepared. First, protein coding sequences in which each of the extracellular domains of CRTAM were linked to the human immunoglobulin G1 heavy chain constant region were identified in a gene database. In addition, gene synthesis was performed by connecting the remaining sequences except for the stop codon portion of the base sequence constituting the domains into one. The accuracy of gene synthesis was confirmed through sequencing after constructing a protein expression plasmid. A schematic diagram showing the cDNA region of each domain expressing the recombinant protein is shown in FIG. 5 .
실시예 5. 수용성 재조합 CRTAM 융합 단백질 발현 플라스미드 제조Example 5. Preparation of water soluble recombinant CRTAM fusion protein expression plasmid
실시예 4의 인간 CADM1을 인식하는 도메인과 인간 이뮤노글로불린 G1 중쇄 불변 영역을 융합시킨 재조합 CRTAM 융합 단백질을 Chinese hamster ovary (CHO) 세포에 발현시키기 위해, 해당 유전자를 레트로바이러스 유래 발현벡터인 pLNCX2(Addgene)에 클로닝하였다. In order to express the recombinant CRTAM fusion protein obtained by fusing the human CADM1 recognition domain of Example 4 with the human immunoglobulin G 1 heavy chain constant region in Chinese hamster ovary (CHO) cells, the corresponding gene was transferred to the retrovirus-derived expression vector, pLNCX2. (Addgene).
먼저, pLNCX2 벡터에 클로닝하기 위해서 5′말단에 제한효소 XhoI의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응(polymerase chain reaction, PCR)으로 제한효소 절단 부위를 만들고, 3′말단에도 제한효소 NotI의 절단 부위 서열을 만드는 프라이머를 합성하여 중합효소연쇄반응으로 제한효소 절단 부위를 만들었다. 그 후 중합효소연쇄반응으로 인해 제한효소 절단 부위를 가진 유전자를 5′말단은 XhoI을 처리하였으며, 3′말단은 NotI을 처리하였다. 그리고 발현 벡터의 다중클로닝 자리를 xhoI과 NotI을 처리하여 유전자가 삽입될 수 있게 만들었다. 제한효소가 처리된 유전자와 발현 벡터를 섞어 준 다음, 결합 효소(ligase)를 처리하여 연결하였다. First, in order to clone into the pLNCX2 vector, a primer that creates a restriction enzyme Xho I cleavage site sequence is synthesized at the 5' end, a restriction enzyme cleavage site is created by polymerase chain reaction (PCR), and restriction is also made at the 3' end. A restriction enzyme cleavage site was prepared by synthesizing a primer that creates the cleavage site sequence of the enzyme Not I and polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site due to polymerase chain reaction was treated with Xho I, and the 3' end was treated with Not I. In addition, the multicloning site of the expression vector was treated with xho I and Not I so that the gene could be inserted. After mixing the restriction enzyme-treated gene and the expression vector, they were ligated by treatment with a ligase.
그 결과, 인간 CADM1을 인식하는 CRTAM의 세포외 도메인과 인간 이뮤노글로불린 G1 중쇄 불변 영역을 융합시킨 단백질(도 7 참조)을 발현하는 재조합 벡터 CRTAM-IgG.pLNCX2를 완성하였다(도 6 참조).As a result, a recombinant vector CRTAM-IgG.pLNCX2 expressing a protein obtained by fusing the extracellular domain of CRTAM recognizing human CADM1 with the human immunoglobulin G 1 heavy chain constant region (see Fig. 7) was completed (see Fig. 6). .
실시예Example 6. 수용성 재조합 6. Soluble recombination CRTAMCRTAM 융합 단백질과 fusion protein and CADM1을CADM1 발현하는 사람 세포주의 친화도 측정 Affinity measurement of expressing human cell lines
실시예 5의 제작한 수용성 재조합 CRTAM 융합 단백질이 CADM1을 발현하는 사람 세포주와 친화도가 있는지 확인하기 위하여, 수용성 재조합 CRTAM 융합 단백질을 CHO 세포에서 발현시킨 후, protein A column을 이용하여 친화크로마토그래피(affinity chromatography) (GE healthcare)로 정제하였다(도 8a 참조).In order to confirm the affinity of the soluble recombinant CRTAM fusion protein prepared in Example 5 with the human cell line expressing CADM1, after expressing the soluble recombinant CRTAM fusion protein in CHO cells, affinity chromatography was performed using a protein A column ( affinity chromatography) (GE healthcare) (see Fig. 8a).
또한, 정제한 수용성 키메릭 항원 수용체를 인간 IgG 중쇄의 불변 영역을 인식하는 항체(항 인간 IgG 중쇄 불변영역 항체, anti-human IgG Fc region antibody, abcam)를 이용하여 웨스턴 블로팅으로 확인하였다(도 8b 참조).In addition, the purified water-soluble chimeric antigen receptor was confirmed by Western blotting using an antibody (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody, abcam) recognizing the human IgG heavy chain constant region (Fig. see 8b).
그 후, 상기 정제한 수용성 재조합 CRTAM 융합 단백질을 CADM1을 발현하는 세포주(A549 세포주)에 처리하여 수용성 재조합 CRTAM 융합 단백질이 해당 세포와 결합이 가능한지 유세포 분석을 통하여 확인한 결과를 도 8c에 나타내었다.Thereafter, the purified water-soluble recombinant CRTAM fusion protein was treated with a cell line (A549 cell line) expressing CADM1, and it was confirmed through flow cytometry whether the water-soluble recombinant CRTAM fusion protein could bind to the cells. The results are shown in FIG. 8C.
도 8c에 나타난 바와 같이, CADM1을 발현하는 A549 세포는 control Ig (isotype control)과 결합하지 않았으나, 수용성 CRTAM 융합 단백질(CRTAM-Ig)과는 결합하는 것을 확인하였다. As shown in FIG. 8c , it was confirmed that A549 cells expressing CADM1 did not bind to control Ig (isotype control), but bound to soluble CRTAM fusion protein (CRTAM-Ig).
상기 결과에서 확인된 바와 같이, 제작한 수용성 CRTAM 융합 단백질이 CADM1을 발현하는 사람 세포주와 친화도가 높음을 확인하였고, 이는 CADM1을 발현하는 A549 세포를, 제작한 CADM1을 인식하고 인간 이뮤노글로불린 G1 중쇄 불변 영역과 신호전달 단백질 도메인을 융합시킨 재조합 단백질을 발현시킨 세포독성 T 세포(CRTAM.CAR-T 세포) 또는 자연살생세포(CRTAM.CAR-NK 세포)에, CADM1 단백질 특이적 세포독성 검증을 위해 사용할 수 있다는 것을 의미한다.As confirmed from the above results, it was confirmed that the prepared soluble CRTAM fusion protein had high affinity with human cell lines expressing CADM1, which recognized CADM1-expressing A549 cells and recognized human immunoglobulin G 1 Verification of CADM1 protein-specific cytotoxicity to cytotoxic T cells (CRTAM.CAR-T cells) or natural killer cells (CRTAM.CAR-NK cells) expressing a recombinant protein fused with a heavy chain constant region and a signaling protein domain means that it can be used for
실시예Example 7. 세포독성 T 세포 분리 및 활성화 7. Cytotoxic T cell isolation and activation
본 발명의 일 실시예에 따른 키메릭 항원 수용체를 발현하는 T 세포를 제작하기 위해 먼저 세포 독성 T 세포를 사람의 말초 혈액 단핵세포에서 분리하였다. 사람의 말초 혈액 단핵세포(메디랩 코리아)를 구입한 후, 세포독성 T 세포를 분리하기 위해서 자력이용 세포 분리법(magnetic-activated cell sorting)을 이용하였다. 말초 혈액 단핵세포들을 세포독성 T 세포를 제외한 다른 면역세포들과 결합 가능한 항체(CD8+ T cell biotin-conjugated antibody cocktail, Miltenyi Biotec)와 결합시킨 다음, 상기 항체들을 다시 자성을 가진 마이크로비드(anti-biotin microbead)(Miltenyi Biotec)와 결합시켰다. 상기 마이크로비드와 그에 붙은 항체, 세포들을 자기장 분리 칼럼(magnetic seperation column) (Miltenyi Biotec)에 통과시켜 그 중 항체로 표지되지 않은 세포독성 T 세포를 얻었다. 분리한 세포독성 T 세포의 순도를 확인하기 위하여 세포독성 T 세포의 세포표면 인자인 CD8과 CD3ε을 이용한 유세포 분석을 실시하였고 그 결과 95% 이상의 순도를 보였다.In order to prepare T cells expressing a chimeric antigen receptor according to an embodiment of the present invention, cytotoxic T cells were first isolated from human peripheral blood mononuclear cells. After purchasing human peripheral blood mononuclear cells (Medi Lab Korea), magnetic-activated cell sorting was used to isolate cytotoxic T cells. Peripheral blood mononuclear cells are combined with an antibody (CD8 + T cell biotin-conjugated antibody cocktail, Miltenyi Biotec) capable of binding to other immune cells except for cytotoxic T cells, and then the antibodies are combined with magnetic microbeads (anti- biotin microbead) (Miltenyi Biotec). The microbeads, antibodies attached thereto, and cells were passed through a magnetic separation column (Miltenyi Biotec) to obtain cytotoxic T cells unlabeled with the antibodies. In order to confirm the purity of the isolated cytotoxic T cells, flow cytometry was performed using CD8 and CD3ε, which are cell surface factors of cytotoxic T cells, and as a result, the purity was over 95%.
분리한 세포독성 T 세포의 활성화를 위해 1Х106개/ml의 사람 CD3와 CD28 항체가 코팅된 자석비드(Thermo Fisher Scientific), 그리고 100 U/μl recombinant human IL-2가 첨가된 10% 송아지 혈청 함유 RPMI (Welgene)에 1.5Х106개/ml의 밀도로 세포를 재구성하여 24 웰(well) 세포 배양 접시에 접종 후 24시간 동안 배양하였다. Contains 1Х10 6 /ml of human CD3 and CD28 antibody-coated magnetic beads (Thermo Fisher Scientific) and 10% calf serum supplemented with 100 U/μl recombinant human IL-2 for activation of isolated cytotoxic T cells. The cells were reconstituted in RPMI (Welgene) at a density of 1.5Х10 6 cells/ml, inoculated into a 24-well cell culture dish, and cultured for 24 hours.
실시예 8. 키메릭 항원 수용체 발현 세포독성 T 세포 제작Example 8. Construction of chimeric antigen receptor-expressing cytotoxic T cells
상기 실시예 2를 통해 제작한 재조합 벡터를 세포독성 T 세포에 도입하기 위하여 293FT 세포(Thermo Fisher Scientific)를 사용한 렌티바이러스 시스템을 이용하였다. In order to introduce the recombinant vector prepared in Example 2 into cytotoxic T cells, a lentiviral system using 293FT cells (Thermo Fisher Scientific) was used.
먼저, 293FT 세포를 100π 세포 배양 접시에 2.5Х106 세포가 되도록 접종한 후, 10% 송아지 혈청을 함유한 DMEM 배지에서 배양하였다. 배양 24시간 후, 세포가 접시의 60~70% 정도를 덮을 정도로 자라면, 20μg의 CRTAM.CAR-T.pCDH-CMV-MCS-EF1-copGFP 벡터 DNA를 10μg의 psPAX2(Addgene)와 3μg의 pMD2.G(Addgene) 벡터를 Calcium phosphate 및 Hepes-buffered solution을 이용하여 결정화시킨 후 293FT 세포에 도입하였다. 그 후, 상기 발현 벡터가 도입된 293FT 세포를 48시간 후 24시간 간격으로 렌티바이러스를 포함하는 배양 상층액을 모았다. 모은 상층액을 초고속 원심분리기를 21,000rpm으로 2시간 동안 원심분리하여 바이러스를 농축시켰다. 농축된 바이러스를 polybrene 4μg/ml과 혼합하여 활성화시킨 세포독성 T 세포의 배양액에 추가한 후 원심분리기를 이용해 1,800g로 75분 동안 원심분리하여 형질도입을 진행하였다. 원심 분리를 마친 세포독성 T 세포는 4시간 동안 추가 배양 후 10% 송아지 혈청 함유 RPMI 배양액으로 교체하였으며 48시간 후에는 형질도입한 세포독성 T 세포의 일부를 형질도입 효율 측정에 사용하였다. 형질도입 효율은 세포 내부의 GFP 발현량을 유세포 분석을 이용해 측정했으며 형질도입된 세포독성 T 세포(CRTAM CAR-T cell)를 공벡터로 제작한 바이러스를 형질도입한 세포독성 T 세포(Mock CAR-T cell)의 형질도입 비율을 비교하였다(도 9a 참조).First, 293FT cells were inoculated into 2.5Х10 6 cells in a 100π cell culture dish, and cultured in DMEM medium containing 10% calf serum. After 24 hours of incubation, when the cells have grown to cover 60-70% of the dish, 20 μg of CRTAM.CAR-T.pCDH-CMV-MCS-EF1-copGFP vector DNA was mixed with 10 μg of psPAX2 (Addgene) and 3 μg of pMD2. .G (Addgene) vector was crystallized using Calcium phosphate and Hepes-buffered solution and then introduced into 293FT cells. Thereafter, culture supernatants containing lentivirus were collected at intervals of 24 hours after 48 hours of 293FT cells into which the expression vector was introduced. The collected supernatant was centrifuged for 2 hours at 21,000 rpm in an ultra-high-speed centrifuge to concentrate the virus. The concentrated virus was mixed with 4 μg/ml of polybrene and added to the culture medium of activated cytotoxic T cells, followed by centrifugation at 1,800 g for 75 minutes using a centrifuge for transduction. The centrifuged cytotoxic T cells were further cultured for 4 hours and then replaced with RPMI culture medium containing 10% calf serum. After 48 hours, some of the transduced cytotoxic T cells were used to measure transduction efficiency. The transduction efficiency was measured by flow cytometry to measure the amount of GFP expression inside the cell, and the transduced cytotoxic T cell (CRTAM CAR-T cell) was measured using a virus-transduced cytotoxic T cell (Mock CAR-T cell). Transduction rates of T cells) were compared (see FIG. 9a).
실시예 9. 키메릭 항원 수용체 발현 자연살해세포 제작Example 9. Construction of natural killer cells expressing chimeric antigen receptors
상기 실시예 2를 통해 제작한 재조합 벡터를 NK 세포에 도입하기 위하여 293GPG 세포를 사용한 레트로바이러스 시스템을 이용하였다. In order to introduce the recombinant vector prepared in Example 2 into NK cells, a retroviral system using 293GPG cells was used.
먼저, 293GPG 세포 3Х106개를 10ml의 10% 송아지 혈청 함유 DMEM 배양액 10ml에 풀어 100π 세포 배양 접시에 접종 후 24시간 동안 배양하였다. 앞서 제작한 재조합 벡터(CRTAM.CAR-NK.pLNCX2 벡터) 20μg을 Calcium phosphate 및 Hepes-buffered solution을 이용하여 결정화시킨 후 앞서 배양한 293GPG 세포의 배양액에 첨가해주었다. 이후 24시간 간격으로 72시간에 걸쳐 배양액을 교체하며 레트로바이러스를 함유한 293GPG 세포의 배양 상층액을 채취 및 보관하였다. First, 6 3Х10 293GPG cells were dissolved in 10 ml of DMEM culture medium containing 10% calf serum, inoculated into a 100π cell culture dish, and cultured for 24 hours. 20 μg of the previously prepared recombinant vector (CRTAM.CAR-NK.pLNCX2 vector) was crystallized using Calcium phosphate and Hepes-buffered solution, and then added to the previously cultured 293GPG cell culture medium. Thereafter, the culture medium was replaced at 24 hour intervals over 72 hours, and the culture supernatant of 293GPG cells containing the retrovirus was collected and stored.
상기 채취한 레트로바이러스 함유 상층액은 초고속 원심분리기를 이용하여 21,000 rpm으로 2시간 동안 원심분리 후 농축 이전 대비 100배로 농축될 수 있도록 10% 송아지 혈청 함유 Myelocult H5100 배양액(STEMCELL)에 재구성하였다. 농축한 레트로바이러스를 NK92MI 세포(American type culture collection, ATCC)에 형질도입하기 위해 NK92MI 세포를 24 well 세포 배양 접시에 5Х105/ml의 밀도로 접종하였다. 그 다음 농축된 레트로바이러스에 polybrene을 8ug/ml만큼 첨가한 혼합액을 배양 중인 NK92MI세포의 배양액에 추가한 후 24시간 동안 배양하였다. 배양 후 새 배양액으로 교체하였으며 유전자가 도입된 세포의 선별을 위해 배양액 교체 후 24시간 후부터는 neomycin 항생제(600μg/ml)가 첨가된 Myelocult H5100 배양액을 이용하여 레트로바이러스를 처리해준 NK92MI세포를 배양하였다. 14일 간의 neomycin 선별과정을 마친 NK92MI 세포는 다시 신선한 배양액에 옮겨 1주일간 증식시켰다. The collected retrovirus-containing supernatant was centrifuged at 21,000 rpm for 2 hours using an ultra-high-speed centrifuge, and then reconstituted in Myelocult H5100 culture medium (STEMCELL) containing 10% calf serum to be concentrated 100 times compared to before concentration. To transduce the concentrated retrovirus into NK92MI cells (American type culture collection, ATCC), NK92MI cells were inoculated in a 24-well cell culture dish at a density of 5Х10 5 /ml. Then, a mixed solution in which 8ug/ml of polybrene was added to the concentrated retrovirus was added to the culture medium of NK92MI cells, and cultured for 24 hours. After culturing, it was replaced with a new culture medium, and NK92MI cells treated with retrovirus were cultured using Myelocult H5100 culture medium supplemented with neomycin antibiotic (600 μg/ml) 24 hours after the culture medium change for selection of cells into which the gene was introduced. After 14 days of neomycin selection, NK92MI cells were transferred to a fresh culture medium and grown for one week.
증식 후 유세포 분석을 이용해 CRTAM.CAR-NK.pLNCX2를 도입한 NK92MI 세포(CRTAM CAR-NK cell)와 공벡터인 pLNCX2를 도입한 NK92MI 세포(Mock CAR-NK cell)의 CRTAM 발현량을 유세포분석기를 이용하여 비교하였다(도 9b 참조). 이때 1차 항체(primary antibody)로는 항 인간 CRTAM 항체(rat IgG, abcam, #ab17318)를 사용하였고, 2차 항체(secondary antibody)로는 항 rat IgG-Alexa Fluor 488 (goat anti-rat IgG, thermofisher, #a11006)을 사용하였다.After proliferation, the CRTAM expression level of NK92MI cells (CRTAM CAR-NK cells) introduced with CRTAM.CAR-NK.pLNCX2 and NK92MI cells (Mock CAR-NK cells) introduced with pLNCX2, an empty vector, was measured by flow cytometry using flow cytometry. Comparison was made using (see FIG. 9b). At this time, anti-human CRTAM antibody (rat IgG, abcam, #ab17318) was used as the primary antibody, and anti-rat IgG-Alexa Fluor 488 (goat anti-rat IgG, thermofisher, #a11006) was used.
실시예Example 10. CAR-T 세포의 10. CAR-T cells CADM1CADM1 + cell 특이적 세포독성 검증+ Verification of cell-specific cytotoxicity
제작한 키메릭 항원 수용체 발현 세포독성 T 세포가 CADM1을 발현하는 세포를 특이적으로 인지하여 독성을 나타내는지 확인하기 위해, 실시예 3에서 선정한 표적세포인 A549 세포(CADM1 positive cell)를 키메릭 항원 수용체 발현 세포독성 T 세포(CRTAM CAR-T cell) 혹은 공벡터를 도입한 세포독성 T 세포(Mock CAR-T cell)와 6시간 동안 공배양하였다. 공배양 후 상층액에 존재하는 젖산 탈수소 효소(lactate dehydrogenase)의 양으로 세포독성 T 세포의 표적세포(target cell)에 대한 세포독성 정도를 측정하는 비방사성 세포독성 분석법(non-radioactive cytotoxicity assay)을 이용하였다.In order to confirm that the produced chimeric antigen receptor-expressing cytotoxic T cells specifically recognize CADM1-expressing cells and exhibit toxicity, A549 cells (CADM1 positive cells), the target cells selected in Example 3, were used as chimeric antigens. Receptor-expressing cytotoxic T cells (CRTAM CAR-T cells) or empty vector-transduced cytotoxic T cells (Mock CAR-T cells) were co-cultured for 6 hours. A non-radioactive cytotoxicity assay that measures the degree of cytotoxicity of cytotoxic T cells to target cells by the amount of lactate dehydrogenase present in the supernatant after co-culture used
먼저, 96 well 세포 배양 접시의 well에 세포독성 T 세포(1x105 cell)와 표적세포(1x104 cell)를 각각 넣어 effector: target ratio를 10:1로 맞추고, well당 부피는 100μl가 되도록 접종 후 원심분리기를 이용해 250g 조건에서 4분 동안 원심분리하여 세포 간 간격을 가깝게 만든다. 6시간 배양 후 각 well의 상층액을 50μl씩 걷어내어 흡광도 측정용 투명 96 well 접시에 옮긴 후 분석용액 및 1M 염산용액을 처리하여 효소 반응을 진행 및 정지시킨다. 효소 반응을 정지시키고 난 후에는 형광/발광/흡광 측정기(multi-detection plate reader)를 이용하여 490nm 파장대의 흡광도를 측정 및 수치 변환하여 각 well의 세포독성 T 세포의 세포독성 정도를 정량화하였다.First, put cytotoxic T cells (1x10 5 cell) and target cells (1x10 4 cell) into wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, and inoculate so that the volume per well is 100 μl. Centrifuge for 4 minutes under the condition of 250g using a centrifuge to close the gap between cells. After 6 hours of incubation, 50 μl of the supernatant from each well is removed and transferred to a transparent 96-well dish for measuring absorbance, and then the enzyme reaction is progressed and stopped by treatment with analysis solution and 1M hydrochloric acid solution. After stopping the enzyme reaction, the degree of cytotoxicity of the cytotoxic T cells in each well was quantified by measuring and converting the absorbance in the 490 nm wavelength band using a fluorescence/emission/absorption meter (multi-detection plate reader).
그 결과 도 10a에서 나타낸 바와 같이, 제작한 키메릭 항원 수용체 발현 세포독성 T 세포(CRTAM CAR-T cell)는 CADM1을 발현하는 A549 세포주에 39% 이상의 높은 독성을 보인 반면, 대조군인 공벡터를 도입한 세포독성 T 세포(Mock CAR-T cell)의 경우 A549 세포주에 3%의 독성을 나타내었다. As a result, as shown in FIG. 10a, the prepared chimeric antigen receptor-expressing cytotoxic T cell (CRTAM CAR-T cell) showed a high toxicity of 39% or more to the A549 cell line expressing CADM1, while introducing an empty vector as a control One cytotoxic T cell (Mock CAR-T cell) showed 3% toxicity to the A549 cell line.
상기 결과를 통해 CRTAM 세포외 도메인을 포함하고 있는 키메릭 항원 인지 수용체 발현 세포독성 T 세포가 CADM1 단백질에 특이적인 세포독성을 보임으로써, 상기 CAR-T 세포를 이용하여 관련된 암 세포 치료가 가능함을 확인할 수 있었다.Through the above results, it was confirmed that the chimeric antigen recognition receptor-expressing cytotoxic T cells containing the extracellular domain of CRTAM exhibited cytotoxicity specific to the CADM1 protein, and thus the treatment of related cancer cells using the CAR-T cells was possible. could
실시예 11. CAR-NK 세포의 CADM1+ cell 특이적 세포독성 검증Example 11. Verification of CADM1+ cell specific cytotoxicity of CAR-NK cells
제작한 키메릭 항원 수용체 발현 NK 세포가 CADM1을 발현하는 세포를 특이적으로 인지하여 독성을 나타내는지 확인하기 위해, 실시예 3에서 선정한 표적세포인 A549 세포(CADM1 positive cell)를 키메릭 항원 수용체 발현 NK 세포(CRTAM CAR-NK cell) 혹은 공벡터를 도입한 NK 세포(Mock CAR-NK cell)와 공배양하였다. In order to confirm that the prepared chimeric antigen receptor-expressing NK cells specifically recognize CADM1-expressing cells and exhibit toxicity, A549 cells (CADM1 positive cells), the target cells selected in Example 3, were expressed as chimeric antigen receptors. NK cells (CRTAM CAR-NK cells) or NK cells (Mock CAR-NK cells) into which an empty vector was introduced were co-cultured.
공배양 후 상층액에 존재하는 젖산 탈수소 효소(lactate dehydrogenase)의 양으로 NK 세포의 표적세포에 대한 세포독성 정도를 측정하는 비방사성 세포독성 분석법(non-radioactive cytotoxicity assay)을 이용하였다. After co-culture, a non-radioactive cytotoxicity assay was used to measure the degree of cytotoxicity of NK cells to target cells by the amount of lactate dehydrogenase present in the supernatant.
먼저, 96 well 세포 배양 접시의 well에 자연살해세포(1x105 cell)와 표적세포(1x104 cell)를 각각 넣어 effector: target ratio를 10:1로 맞추고, well당 부피는 100μl가 되도록 접종 후 원심분리기를 이용해 250g의 조건에서 4분 동안 원심분리하여 세포간 간격을 가깝게 만든다. 6시간 배양 진행 후 각 well의 상층액을 50μl씩 걷어내어 흡광도 측정용 투명 96 well 접시에 옮긴 후 분석용액 및 1M 염산용액을 처리하여 효소 반응을 진행 및 정지시킨다. 효소 반응을 정지시키고 난 후에는 형광/발광/흡광 측정기(multi-detection plate reader)를 이용하여 490nm 파장대의 흡광도를 측정 및 수치 변환하여 각 well의 NK 세포의 세포독성 정도를 정량화하였다.First, put natural killer cells (1x10 5 cell) and target cells (1x10 4 cell) into wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, inoculate the well to a volume of 100 μl, and then centrifuge. Centrifuge for 4 minutes at 250g using a separator to close the space between cells. After 6 hours of incubation, 50 μl of the supernatant from each well is removed and transferred to a transparent 96-well dish for measuring absorbance, and then the enzyme reaction is progressed and stopped by treatment with analysis solution and 1M hydrochloric acid solution. After stopping the enzyme reaction, the degree of cytotoxicity of NK cells in each well was quantified by measuring and converting the absorbance in the 490 nm wavelength band using a fluorescence/luminescence/absorption meter (multi-detection plate reader).
그 결과 도 10b에서 나타낸 바와 같이, 제작한 키메릭 항원 수용체 발현 자연살해세포(CRTAM CAR-NK cell)는 CADM1을 발현하는 A549 세포주에 27% 이상의 높은 독성을 보인 반면, 대조군인 공벡터를 도입한 자연살해세포(Mock CAR-NK cell)의 경우 A549 세포주에 -2% 이하의 독성을 나타내었다. As a result, as shown in FIG. 10B, the prepared chimeric antigen receptor-expressing natural killer cells (CRTAM CAR-NK cells) showed high toxicity of 27% or more to the CADM1-expressing A549 cell line, whereas the empty vector as a control was introduced. Natural killer cells (Mock CAR-NK cells) showed toxicity of -2% or less to the A549 cell line.
상기 결과를 통해 CRTAM 세포외 도메인을 포함하고 있는 키메릭 항원 인지 수용체 발현 NK 세포가 CADM1 단백질에 특이적인 세포독성을 보임으로써, 상기 CAR-NK 세포를 이용하여 관련된 암 세포 치료가 가능함을 확인할 수 있었다.Through the above results, it was confirmed that the chimeric antigen recognition receptor-expressing NK cells containing the CRTAM extracellular domain showed specific cytotoxicity to the CADM1 protein, and thus the treatment of related cancer cells using the CAR-NK cells was possible. .
이제까지 본 발명에 대하여 그 바람직한 실시예들을 중심으로 살펴보았다. 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 특허청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.So far, the present invention has been looked at with respect to its preferred embodiments. Those skilled in the art to which the present invention pertains will be able to understand that the present invention can be implemented in a modified form without departing from the essential characteristics of the present invention. Therefore, the disclosed embodiments should be considered from an illustrative rather than a limiting point of view. The scope of the present invention is shown in the claims rather than the foregoing description, and all differences within the equivalent scope will be construed as being included in the present invention.

Claims (15)

  1. 항원 결합 도메인;antigen binding domain;
    막관통 도메인; 및transmembrane domain; and
    세포 내 신호전달 도메인을 포함하는 키메릭 항원 수용체(CAR)로서, 상기 항원 결합 도메인은 CADM1(Cell adhesion molecule 1)에 특이적으로 결합하는 CRTAM(class-I MHC-restricted T cell associated molecule)의 세포외 도메인을 포함하는 것인, 키메릭 항원 수용체.A chimeric antigen receptor (CAR) comprising an intracellular signaling domain, wherein the antigen-binding domain is a cell of a class-I MHC-restricted T cell associated molecule (CRTAM) that specifically binds to CADM1 (Cell adhesion molecule 1). A chimeric antigen receptor comprising an exodomain.
  2. 제1항에 있어서,According to claim 1,
    상기 항원 결합 도메인은 CRTAM의 세포외 도메인 한 쌍이 이합체(dimer)의 형태로 각각 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)에 연결된 것을 특징으로 하는 키메릭 항원 수용체.The antigen-binding domain is a chimeric antigen receptor, characterized in that a pair of extracellular domains of CRTAM are linked to a human immunoglobulin G 1 heavy chain constant region, respectively, in the form of a dimer.
  3. 제1항에 있어서,According to claim 1,
    상기 CRTAM의 세포외 도메인은 서열번호 1로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The chimeric antigen receptor, wherein the extracellular domain of CRTAM comprises the amino acid sequence represented by SEQ ID NO: 1.
  4. 제2항에 있어서,According to claim 2,
    인간 이뮤노글로불린 G1 중쇄 불변 영역부위(human IgG1 heavy chain constant region)는 서열번호 2로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.A human immunoglobulin G 1 heavy chain constant region region (human IgG 1 heavy chain constant region) comprising the amino acid sequence represented by SEQ ID NO: 2, chimeric antigen receptor.
  5. 제1항에 있어서,According to claim 1,
    상기 막관통 도메인은 CD8α이며, 서열번호 3으로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The transmembrane domain is CD8α and comprises the amino acid sequence represented by SEQ ID NO: 3, chimeric antigen receptor.
  6. 제1항에 있어서,According to claim 1,
    상기 세포 내 신호전달 도메인은 CD28, 4-1BB 및 CD3-zeta로 이루어진 것 또는 이들의 조합을 포함하는 것인, 키메릭 항원 수용체.The intracellular signaling domain is composed of CD28, 4-1BB and CD3-zeta, or a chimeric antigen receptor comprising a combination thereof.
  7. 제6항에 있어서, According to claim 6,
    상기 CD28은 서열번호 4; 4-1BB은 서열번호 5; 및 CD3-zeta은 서열번호 6으로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The CD28 is SEQ ID NO: 4; 4-1BB is SEQ ID NO: 5; and CD3-zeta is a chimeric antigen receptor comprising the amino acid sequence represented by SEQ ID NO: 6.
  8. 제1항에 있어서, According to claim 1,
    상기 항원 결합 도메인은 CD8α 신호 펩타이드를 포함하며, 서열번호 7로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The antigen-binding domain includes a CD8α signal peptide, and comprises an amino acid sequence represented by SEQ ID NO: 7, chimeric antigen receptor.
  9. 제1항 내지 제8항 중 어느 한 항의 키메릭 항원 수용체 단백질을 코딩하는 폴리 뉴클레오티드.A polynucleotide encoding the chimeric antigen receptor protein of any one of claims 1 to 8.
  10. 제9항의 폴리 뉴클레오티드를 포함하는 벡터.A vector comprising the polynucleotide of claim 9.
  11. 제10항의 벡터로 형질전환된 세포.A cell transformed with the vector of claim 10.
  12. 제11항에 있어서,According to claim 11,
    상기 세포는 세포독성 T 세포 또는 NK 세포인 것을 특징으로 하는 세포.Cells, characterized in that the cells are cytotoxic T cells or NK cells.
  13. 제12항의 세포를 유효성분으로 포함하는 CADM1을 발현하는 암의 치료용 약학적 조성물.A pharmaceutical composition for the treatment of cancer expressing CADM1 comprising the cell of claim 12 as an active ingredient.
  14. 제13항에 있어서,According to claim 13,
    상기 CADM1을 발현하는 암은 비소세포 폐암(non-small lung cancer cell, NSCLC), 폐 선암종(lung adenocarcinoma), 사람T세포림프친화바이러스 1형에 감염된 T 세포 백혈병 (HTLV-1-infected T cell leukemia), 성인T세포백혈병(adult T cell leukemia), 난소암(ovarian cancer), 유방암(breast cancer), 자궁경부암(cervical cancer), 전립선암(prostate cancer), 상인두암(nasopharyngeal carcinoma), 위암(stomach cancer) 및 췌장암(pancreatic cancer)으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 조성물.Cancers expressing the CADM1 include non-small lung cancer cell (NSCLC), lung adenocarcinoma, and human T-cell lymphotropic virus type 1-infected T-cell leukemia (HTLV-1-infected T-cell leukemia). ), adult T cell leukemia, ovarian cancer, breast cancer, cervical cancer, prostate cancer, nasopharyngeal carcinoma, stomach cancer cancer) and pancreatic cancer (pancreatic cancer) characterized in that the composition is selected from the group consisting of.
  15. 제11항의 세포를 포함하는 세포 치료제.A cell therapy product comprising the cell of claim 11.
PCT/KR2022/014464 2021-10-29 2022-09-27 Chimeric antigen receptor specifically binding to cadm1 and uses thereof WO2023075177A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020210146245A KR20230062691A (en) 2021-10-29 2021-10-29 Chimeric antigen receptor specifically binding to CADM1 and use thereof
KR10-2021-0146245 2021-10-29

Publications (1)

Publication Number Publication Date
WO2023075177A1 true WO2023075177A1 (en) 2023-05-04

Family

ID=86158283

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/014464 WO2023075177A1 (en) 2021-10-29 2022-09-27 Chimeric antigen receptor specifically binding to cadm1 and uses thereof

Country Status (2)

Country Link
KR (1) KR20230062691A (en)
WO (1) WO2023075177A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180081011A (en) * 2017-01-05 2018-07-13 한국생명공학연구원 Anti-ceacam6 chimeric antigen receptor specifically binding to ceacam6
KR20210025905A (en) * 2019-08-28 2021-03-10 고려대학교 산학협력단 Chimeric antigen receptor specifically binding to VLA-4 and use thereof
US20210079092A1 (en) * 2018-03-16 2021-03-18 The University Of Tokyo CADM1 V9-Recognizing Antibody

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010102175A1 (en) 2009-03-05 2010-09-10 Medarex, Inc. Fully human antibodies specific to cadm1
KR102566453B1 (en) 2019-12-19 2023-09-06 주식회사 에스아이웨어 Apparatus for steering wheel multi test with pin mapping

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180081011A (en) * 2017-01-05 2018-07-13 한국생명공학연구원 Anti-ceacam6 chimeric antigen receptor specifically binding to ceacam6
US20210079092A1 (en) * 2018-03-16 2021-03-18 The University Of Tokyo CADM1 V9-Recognizing Antibody
KR20210025905A (en) * 2019-08-28 2021-03-10 고려대학교 산학협력단 Chimeric antigen receptor specifically binding to VLA-4 and use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BOLES KENT S ET AL: "The tumor suppressor TSLC1/NECL-2 triggers NK-cell and CD8(+) T-cell responses through the ceff-surface receptor CRTAM", BLOOD, vol. 106, no. 3, 1 August 2005 (2005-08-01), US , pages 779 - 786, XP002550646, ISSN: 0006-4971, DOI: 10.1182/blood-2005-02-0817 *
ZECHMANN MARIA, REESE SVEN, GÖBEL THOMAS W.: "Chicken CRTAM Binds Nectin-Like 2 Ligand and Is Upregulated on CD8+ αβ and γδ T Lymphocytes with Different Kinetics", PLOS ONE, vol. 8, no. 12, pages e81942, XP093060774, DOI: 10.1371/journal.pone.0081942 *

Also Published As

Publication number Publication date
KR20230062691A (en) 2023-05-09

Similar Documents

Publication Publication Date Title
CN111849910B (en) Engineered immune cells and uses thereof
WO2021057823A1 (en) Ror1 specific chimeric antigen receptors and their therapeutic applications
WO2021256724A1 (en) Chimeric antigen receptor targeting bcma and use thereof
US20240082306A1 (en) Novel chimeric antigen receptor and use thereof
WO2022218226A1 (en) Engineered immune cell and use thereof
WO2023003404A1 (en) Novel chimeric antigen receptor and immune cells expressing same
WO2023075177A1 (en) Chimeric antigen receptor specifically binding to cadm1 and uses thereof
WO2023068584A1 (en) Chimeric antigen receptor specifically binding to cd138 and uses thereof
WO2022250440A1 (en) Receptor tyrosine-protein kinase erbb3-specific chimeric antigen receptor and immune cell therapy product expressing same
WO2023287049A1 (en) Chimeric antigen receptor binding specifically to cd30 and use thereof
WO2022215919A1 (en) Chimeric antigen receptor specifically binding to cd47 and use thereof
WO2022239986A1 (en) Chimeric antigen receptor comprising extracellular domain of baff and use thereof
WO2022220433A1 (en) Chimeric antigen receptor binding specifically to programmed death-ligand 1 (pd-l1) and use thereof
WO2021235697A1 (en) Cd22-specific antibody and use thereof
WO2022186682A1 (en) Chimeric antigen receptor specifically binding to rank ligand, and use thereof
WO2022182059A1 (en) Chimeric antigen receptor specifically binding to hla class ii and use thereof
WO2023113292A1 (en) Transformed antigen-specific professional antigen-presenting cell comprising chimeric antigen receptor (car) and use thereof
KR102231285B1 (en) Chimeric antigen receptor specifically binding to VLA-4 and use thereof
WO2023113293A1 (en) Transformed antigen-specific professional antigen-presenting cell containing chimeric antigen receptor (car) and use thereof
KR102231284B1 (en) Chimeric antigen receptor specifically binding to CD38 and use thereof
WO2022203226A1 (en) Transformed antigen-specific professional antigen presenting cell including chimeric antigen receptor (car) and use thereof
WO2022211376A1 (en) Transformed antigen-specific professional antigen presenting cells comprising chimeric antigen receptor (car) and use thereof
WO2022203227A1 (en) Transformed antigen-specific professional antigen-presenting cell comprising chimeric antigen receptor (car) and use thereof
WO2022234976A1 (en) Transformed antigen-specific professional antigen-presenting cell containing chimeric antigen receptor (car) and use thereof
WO2022215920A1 (en) Transformed antigen-specific professional antigen-presenting cells comprising chimeric antigen receptor (car) and use thereof

Legal Events

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

Ref document number: 22887381

Country of ref document: EP

Kind code of ref document: A1