WO2022182059A1 - Chimeric antigen receptor specifically binding to hla class ii and use thereof - Google Patents

Chimeric antigen receptor specifically binding to hla class ii and use thereof Download PDF

Info

Publication number
WO2022182059A1
WO2022182059A1 PCT/KR2022/002352 KR2022002352W WO2022182059A1 WO 2022182059 A1 WO2022182059 A1 WO 2022182059A1 KR 2022002352 W KR2022002352 W KR 2022002352W WO 2022182059 A1 WO2022182059 A1 WO 2022182059A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
chimeric antigen
antigen receptor
hla
Prior art date
Application number
PCT/KR2022/002352
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
Priority claimed from KR1020210026090A external-priority patent/KR102671408B1/en
Application filed by 주식회사 이뮤노로지컬디자이닝랩 filed Critical 주식회사 이뮤노로지컬디자이닝랩
Publication of WO2022182059A1 publication Critical patent/WO2022182059A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C07K14/70521CD28, 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to a chimeric antigen receptor (CAR) that specifically binds to HLA class II; a polynucleotide encoding the chimeric antigen receptor protein; a vector comprising the polynucleotide; T cells or natural killer cells transformed with the vector (Natural killer cell, NK cell); It relates to a cell therapeutic agent or a pharmaceutical composition for the treatment of cancer comprising the cell as an active ingredient.
  • CAR chimeric antigen receptor
  • Human major histocompatibility complex (MHC) antigen class II consists of HLA-DP, HLA-DQ, and HLA-DR, and consists of a heterodimer. Integral membrane protein composed of HLA-DP ⁇ and HLA-DP ⁇ for HLA-DP, HLA-DQ ⁇ and HLA-DQ ⁇ for HLA-DQ, and HLA-DR ⁇ and HLA-DR ⁇ for HLA-DR to be.
  • HLA class II is mainly expressed in professional antigen-presenting cells and plays a role in delivering peptide antigens to CD4 + T cells (helper T cells). However, HLA class II is expressed abnormally high in various carcinomas when normal cells are tumorigenic.
  • HLA class II can block the action of immune checkpoint inhibitors (Johnson et al., 2016; Johnson et al., 2018). That is, HLA class II binds to HLA class II ligand LAG-3 or Fc receptor-like 6 (FCRL6), respectively, to form natural killer cells (NK cells) or cytotoxic T lymphocytes (CTLs). Inhibits the activity of or helper T lymphocytes (Johnson et al., 2016; Johnson et al., 2018).
  • immune checkpoint inhibitors Johnson et al., 2016; Johnson et al., 2018. That is, HLA class II binds to HLA class II ligand LAG-3 or Fc receptor-like 6 (FCRL6), respectively, to form natural killer cells (NK cells) or cytotoxic T lymphocytes (CTLs). Inhibits the activity of or helper T lymphocytes (Johnson et al., 2016; Johnson et al., 2018).
  • cytotoxic T lymphocytes CTL
  • NK cells natural killer cells
  • a single-chain variable fragment (scFv) portion of an antigen-recognizing antibody is grafted onto the cytoplasmic signaling domain of CD3 ⁇ or other protein, chimeric antigen receptor, CAR) was tried as a method of delivering.
  • the chimeric antigen receptor is grafted onto T cells or natural killer cells, the anticancer action of T cells or natural killer cells is enhanced by the specific antigen recognition of single-chain Fv fragments 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. Indeed, these chimeric antigen receptor-expressing T cells or natural killer cells have shown efficacy in several cancers (Holzinger et al., 2016; Pettitt et al., 2018; Wang et al., 2020).
  • lymphocyte activation gene-3 (LAG-3; CD223) is an integral membrane protein belonging to the immunoglobulin (Ig) superfamily that is expressed on the cell surface when T cells are activated.
  • LAG-3 expressed in activated T cells is known to inhibit the activity of T cells.
  • LAG-3 is structurally similar to CD4 and binds to MHC class II, but has a much higher affinity for MHC class II than CD4.
  • human LAG-3 shows very high affinity to all of human MHC class II, HLA-DP, HLA-DQ, and HLA-DR (Niehrs et al., 2019).
  • the present inventors used the fact that HLA class II expression in various cancer cells is confirmed or induced, and that the HLA class II and LAG-3 can bind to the extracellular domain of LAG-3 (No. 1 to 4) were used to prepare cancer-specific chimeric antigen receptor-expressing T cells and natural killer cells expressing HLA class II, and the T cells or natural killer cells specifically target cells expressing HLA class II. It was confirmed through an experiment that it has a toxic ability. At this time, the signal transduction strength was amplified by making a fusion protein linking each of the extracellular domains (No. 1 to No. 4) of LAG-3 with the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region).
  • the existing chimeric antigen receptor has one site for recognizing an antigen by a single chain Fv fragment
  • the chimeric antigen receptor designed by the present inventors has the extracellular domain of LAG-3 (No. 1 to No. 4).
  • human human immunoglobulin G 1 heavy chain constant region IgG 1 heavy chain constant region
  • two extracellular domains of LAG-3 (No. 1 to No. 4) per chimeric antigen receptor recognize antigen Therefore, it was attempted to amplify the signal transduction strength.
  • 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 therapeutics or HLA class II, including the transformed T cells or natural killer cells.
  • the present invention provides an antigen-binding domain; transmembrane domain; and an intracellular signaling domain, wherein the antigen binding domain comprises an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II.
  • a phosphorus, chimeric antigen receptor is provided.
  • 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 pharmaceutical composition for the treatment of cancer comprising the transformed T cells or natural killer cells, a cell therapeutic agent or HLA class II expressing cancer cells.
  • the chimeric antigen receptor according to the present invention includes an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II (HLA-DP, HLA-DQ, HLA-DR). Therefore, in the case of cytotoxic T cells or natural killer cells transformed with a vector capable of overexpressing the chimeric antigen receptor, it is specific for HLA class II (HLA-DP, HLA-DQ, HLA-DR) expressing carcinoma. Because it has cytotoxicity, it can be usefully used as an immune cell therapeutic for cancer treatment.
  • FIG. 1 is a schematic diagram showing the cDNA region 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.
  • HLA class II is a view of HLA class II in cancer cells expressing HLA class II (HLA class II positive cells) and cancer cells not expressing HLA class II (HLA class II negative cells), which are targets of the chimeric antigen receptor provided in the present invention. It is a diagram showing the result of measuring the expression level using flow cytometry.
  • 5a is a diagram showing the results of purification by affinity chromatography using a protein A column after making the chimeric antigen receptor provided in the present invention water-soluble and expressing it in Chinese hamster ovary (CHO) cells.
  • Figure 5b is a water-soluble chimeric antigen receptor according to an embodiment of the present invention using an antibody recognizing the constant region of a human IgG heavy chain (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody) Western blotting It is a diagram showing the results confirmed by .
  • FIG. 5c is a diagram showing whether the water-soluble chimeric antigen receptor according to an embodiment of the present invention recognizes cancer cells (HLA class II positive cells) expressing the extracellular domain target, HLA class II, using flow cytometry.
  • cancer cells HLA class II positive cells
  • Figure 6a is a diagram showing the result of comparing the expression rate of GFP in cytotoxic T cells (LAG-3 CAR-T cell) transduced with the expression vector according to an embodiment of the present invention using a lentiviral system. .
  • Figure 6b is a diagram showing the result of comparing the expression ratio of LAG-3 in natural killer cells (LAG-3 CAR-NK cells) transduced with the expression vector according to an embodiment of the present invention using a retroviral system to be.
  • Figure 7a shows HLA using cytotoxic T cells (LAG-3 CAR-T cells) or empty vector-transduced cytotoxic T cells (Mock T cells) as effector cells according to an embodiment of the present invention; It is a diagram showing the results of measuring the cytotoxicity to cancer cells expressing class II (HLA class II positive cells).
  • HLA class II using natural killer cells (LAG-3 CAR-NK cells) or mock NK cells transduced with an empty vector as effector cells according to an embodiment of the present invention; It is a diagram showing the result of measuring the cytotoxicity to cancer cells (HLA class II positive cells) expressing.
  • the present invention provides an antigen-binding domain; transmembrane domain; and an intracellular signaling domain, wherein the antigen binding domain comprises an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II.
  • a chimeric antigen receptor is provided.
  • chimeric antigen receptor binds to a desired antigen without the mediation of antigen presenting cells (APCs) or antibodies necessary for naturally activating T cells or natural killer cells, thereby producing an antigen-antibody reaction. It refers to a fusion protein for expression in T cells or natural killer cells in order to induce activation of T cells and attack cells expressing the corresponding antigen. That is, when expressed in T cells or natural killer cells, it can be considered as a protein that binds to an antigen and induces activation of these cells. Through this, it may be a protein recognizing an antigen specific to a cell to cause an immune response, and the cell to cause an immune response may refer to a cell existing in a specific tissue or constituting a tissue causing a lesion.
  • APCs antigen presenting cells
  • the chimeric antigen receptor protein according to an embodiment of the present invention may include a functional equivalent.
  • “Functional equivalent” means at least 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably the amino acid sequence of the chimeric antigen receptor protein as a result of the addition, substitution, or deletion of amino acids. refers to a protein having a sequence homology of 95% or more and exhibiting substantially homogeneous physiological activity. “Substantially homogenous physiological activity” means that it has an activity capable of specifically binding to HLA class II (HLA-DP, HLA-DQ, HLA-DR).
  • the present invention also includes fragments, derivatives and analogues of chimeric antigen receptors.
  • fragments, derivatives and analogues of chimeric antigen receptors As used herein, the terms 'fragment', 'derivative' and 'analog' refer to a polypeptide that retains substantially the same biological function or activity as the chimeric antigen receptor protein of the present invention.
  • Fragments, derivatives and analogs of the present invention include 1) polypeptides in which one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted, wherein the substituted amino acid residues are encoded by the genetic code.
  • polypeptide may or may not) or 2) a polypeptide having substituent(s) at one or more amino acid residues, or 3) maturation associated with another compound (a compound capable of extending the half-life of the polypeptide, such as polyethylene glycol) a polypeptide derived from a polypeptide, or 4) the polypeptide associated with an additional amino acid sequence (eg, a leader sequence, a secretory sequence, a sequence used to purify the polypeptide, a proteinogen sequence or a fusion protein) It may be a polypeptide derived from The fragments, derivatives and analogs as defined herein are well known to those skilled in the art.
  • Lymphocyte-activation gene 3 (LAG-3) protein, called lymphocyte activation gene-3, belongs to the immunoglobulin (Ig) superfamily, and has an extracellular domain consisting of four domains designated D1 to D4. It is an integral membrane protein.
  • the antigen - binding domain is a dimer ( dimer), but is not limited thereto.
  • the extracellular domain (No. 1 to No. 4) of the LAG-3 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 showing 95% or more homology thereto, but is not limited thereto.
  • HLA class II is an intrinsic membrane protein that is mainly expressed in professional antigen presenting cells and delivers peptide antigens to CD4+ T cells (helper T cells), but HLA class II is When it becomes oncogenic, it has abnormally high expression in various carcinomas.
  • carcinomas expressing HLA class II have been classified as head and neck squamous cell carcinoma (Meissner et al., 2008), thymoma (Strobel et al., 2008) and esophageal squamous cell carcinoma ( Esophageal squamous cell carcinoma) (Hu et al., 2014), melanoma (Johnson et al., 2016; Rodig et al., 2018; Johnson et al., 2018), breast cancer (Park et al.) al., 2017; Loi et al., 2016; Bartek et al., 1987), colorectal cancer (Michel et al., 2010; Bustin et al., 2001), ovarian cancer (callahan et al.) al., 2008, Turner et al., 2017), prostate cancer (Younger et al., 2008), neuroblstoma (Yazawa et al., 2002), glio
  • the transmembrane domain is selected from the group consisting of CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. It may include one or more, 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, but is not limited thereto.
  • intracellular signaling domain refers to a site that activates an immune response of an immune cell 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 high activity in cancer cells, particularly HLA class II ((HLA-DP, HLA-DQ, HLA- DR) expressing cancer cells.
  • HLA class II ((HLA-DP, HLA-DQ, HLA- DR) expressing cancer cells.
  • the CD28 is SEQ ID NO: 4 or 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably 95% or more sequence homology thereto, and the amino acid sequence represented by SEQ ID NO: 4 and may consist of an amino acid sequence exhibiting a substantially equivalent function
  • 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% thereof Having the above sequence homology, it may consist of an amino acid sequence exhibiting a function substantially equivalent to the amino acid sequence shown in SEQ ID NO: 5, CD3 zeta functions as an NK cell activation domain, and SEQ ID NO: 6 or 70 thereof % or more, preferably 80% or more, more preferably 90% or more, even more preferably 95% or more, and an amino acid sequence that exhibits a substantially equivalent function to the amino acid sequence represented by SEQ ID NO: 6.
  • the antigen-binding domain may include a LAG-3 signal peptide, and the LAG-3 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 in another aspect according to the present invention, there is provided a polynucleotide encoding the chimeric antigen receptor protein.
  • 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 the codon preferred in the organism to express the antigen receptor.
  • Various modifications may be made to the coding region within the range that does not occur, and various modifications or modifications may be made within the range that does not affect the expression of the gene in parts other than the coding region, 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 comprising the polynucleotide, and a cell transformed with the vector.
  • the vector used in the present invention may use a variety of vectors known in the art, and may include a promoter, a terminator, an enhancer, etc., depending on the type of host cell to produce the antigen receptor. Expression control sequences, sequences for membrane targeting or secretion, etc. can be appropriately selected and variously combined according to the purpose.
  • the vector of the present invention includes, but is not limited to, a plasmid vector, a cosmid vector, a bacteriophage vector, and a viral vector. Suitable vectors include a signal sequence or leader sequence for membrane targeting or secretion in addition to expression control elements such as promoter, operator, start codon, stop codon, polyadenylation signal and enhancer, and may be prepared in various ways depending on the purpose.
  • a vector for a lenti-virus or a vector for a retro-virus can be used.
  • pLNCX2 retro-virus vector
  • a cell can be transformed by introducing a chimeric antigen receptor that specifically binds to HLA class II into the cell through the vector.
  • the cell may be a T cell, a tumor infiltrating lymphocyte, a B cell, a natural killer cell, or an NKT cell, preferably a cytotoxic T cell or a natural killer cell.
  • the cell may be obtained or prepared from bone marrow, peripheral blood, peripheral blood mononuclear cells or umbilical cord blood, and the cell may be a human cell, but is not limited thereto.
  • the transformed cells into which the chimeric antigen receptor of the present invention is introduced recognizes HLA class II ((HLA-DP, HLA-DQ, HLA-DR) as an antigen and strongly binds thereto.
  • HLA-DP HLA-DP, HLA-DQ, HLA-DR
  • chimeric antigen receptor T cells (hereinafter abbreviated as 'CAR-T cells')" or “chimeric antigen receptor NK cells (hereinafter referred to as chimeric antigen receptor NK cells)" 'CAR-NK cells') is a chimeric antigen receptor that specifically responds to cancer cells other than the original T cell receptor or NK cell receptor by transducing normal T cells or natural killer cells, etc. refers to T cells or NK cells expressing T cells or NK cells having this receptor induce apoptosis of target cells, thereby exhibiting cytotoxicity.
  • CAR-T cells or CAR-NK cells may be cytotoxic T cells or cells in which the chimeric antigen receptor of the present invention is introduced into NK cells.
  • the cells have the advantage of anticancer-specific targeted therapy, which is the existing advantage of CAR-T therapeutics.
  • the CAR-T cells or CAR-NK cells of the present invention are HLA class II (HLA-DP, HLA-DQ, HLA- It can recognize and effectively destroy cancer cells expressing DR).
  • the present invention provides a cell therapeutic agent comprising the cell or a pharmaceutical composition for the treatment of cancer comprising the same as an active ingredient.
  • cell therapeutic refers to cells and tissues prepared through isolation, culture, and special manipulation from an individual, and as pharmaceuticals used for therapeutic purposes, living autologous, allogeneic, or xenogeneic cells or tissues are restored to function. It refers to a drug used for therapeutic purposes through a series of actions, such as in vitro proliferation and selection or changing the biological properties of cells in other ways.
  • treatment refers to any action in which symptoms of cancer are improved or beneficially changed by administration of the composition.
  • composition may include a pharmaceutically acceptable carrier.
  • the "pharmaceutically acceptable carrier” may mean a carrier or diluent that does not inhibit the biological activity and properties of the injected compound without irritating the organism.
  • the type of carrier usable in the present invention is not particularly limited, and any carrier commonly used in the art and pharmaceutically acceptable may 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 may be used in combination of two or more.
  • composition comprising a pharmaceutically acceptable carrier may be in various oral or parenteral formulations.
  • formulation it is prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants that are usually used.
  • solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations include at least one excipient to the compound, for example, starch, calcium carbonate, sucrose, lactose. , gelatin, etc. may be mixed and prepared.
  • excipients for example, starch, calcium carbonate, sucrose, lactose. , gelatin, etc.
  • lubricants such as magnesium stearate and talc may also be used.
  • Liquid formulations for oral use include suspensions, solutions, emulsions, and syrups.
  • various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. have.
  • Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized formulations and suppositories.
  • Non-aqueous solvents and suspending agents include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
  • Witepsol, Macrogol, Tween 61, cacao butter, laurin fat, glycerogelatin, etc. may be used as the base of the suppository.
  • composition may 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 subject's type and severity, age, sex, infected virus type, and drug. Activity, sensitivity to drug, time of administration, route of administration and excretion rate, duration of treatment, factors including concomitant drugs and other factors well known in the medical field.
  • Intraperitoneal administration, intravenous administration, intramuscular administration, subcutaneous administration, intradermal administration, oral administration, topical administration, may be administered intranasally, 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 two to three times.
  • the two active ingredients are single drugs, the number of administrations may be the same or different.
  • the composition of the present invention can be used alone or in combination with other drug treatments for the treatment of HLA class II-expressing cancer. Taking all of the above factors into consideration, it is important to administer an amount that can obtain the maximum effect with a minimum amount without side effects, and can be easily determined by those skilled in the art.
  • the subject is a human, monkey, cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig that has or can develop cancer expressing HLA class II. all animals, including If the disease can be effectively treated by administering the pharmaceutical composition of the present invention to the subject, the type of subject is included without limitation.
  • the types of cancer to be treated include head and neck squamous cell carcinoma, thymoma, esophageal squamous cell carcinoma, melanoma, Breast cancer, colorectal cancer, ovarian cancer, prostate cancer, neuroblstoma, glioma, non-small cell lung cancer , classic Hodgkin lymphoma, T-cell leukemia/lymphoma, B cell lymphoma, chronic myeloid leukemia, chronic lymphocytic leukemia), acute myeloid leukemia, or acute lymphoid leukemia, but is not limited thereto.
  • a pair of extracellular domains (No. 1 to No. 4) of LAG-3 are each linked to the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region) for antigen recognition
  • FIG. 1 A schematic diagram showing the cDNA region of each domain expressing the chimeric antigen receptor is shown in FIG. 1 .
  • HLA-DP HLA-DQ, HLA-DR
  • a recombinant protein in which the human immunoglobulin G 1 heavy chain constant region and the signaling protein domain are fused to cytotoxic T cells and natural killer cells To do this, the gene was cloned into pCDH-CMV-MCS-EF1-copGFP (System Biosciences), a lentivirus-derived expression vector, or pLNCX2 (Addgene), a retrovirus-derived expression vector.
  • a primer that creates a cleavage site sequence of restriction enzyme XbaI at the 5' end was synthesized, and a restriction enzyme cleavage site was created by polymerase chain reaction, and also restricted at the 3' end. Restriction enzyme cleavage site was created by polymerase chain reaction by synthesizing a primer making the cleavage site sequence of the enzyme NotI.
  • the 5' end of the gene having a restriction enzyme cleavage site was treated with XbaI, and the 3' end was treated with NotI due to the polymerase chain reaction.
  • the multicloning site of the expression vector was treated with XbaI and NotI to allow the gene to be inserted. After mixing the restriction enzyme-treated gene with the expression vector, it was ligated by treatment with a ligase.
  • a primer that creates a cleavage site sequence of restriction enzyme Bgl II at the 5' end was synthesized to create a restriction enzyme cleavage site by polymerase chain reaction, and the cleavage site sequence of restriction enzyme NotI was also synthesized at the 3' end.
  • a restriction enzyme cleavage site was created by synthesizing the primers to be made and by polymerase chain reaction.
  • the 5' end of the gene having a restriction enzyme cleavage site was treated with BglII, and the 3' end was treated with NotI due to the polymerase chain reaction.
  • the multicloning site of the expression vector was treated with BglII and NotI to allow the gene to be inserted. After mixing the restriction enzyme-treated gene with the expression vector, it was ligated by treatment with a ligase.
  • a recombinant vector LAG-3 that recognizes HLA class II (HLA-DP, HLA-DQ, HLA-DR) and expresses a protein obtained by fusion of a human immunoglobulin G 1 heavy chain constant region and a signaling protein domain.
  • CAR-T.pCDH-CMV-MCS-EF1-copGFP and LAG-3.CAR-NK.pLNCX2 were completed (see FIGS. 2A and 2B ).
  • HLA class II In order to screen human cell lines expressing HLA class II on the cell surface, acute promyelocytic leukemia-derived cell line HL-60 (ATCC) and T cell lymphoma-derived cell line Jurkat (ATCC) Expression of HLA class II was confirmed in the cell line. To confirm expression, the cell lines were treated with an antibody (biolegend) capable of binding to HLA class II to which a fluorescent protein was attached, and then flow cytometry (fluorescence-activated cell sorting) was used. As a result of the analysis, it was confirmed that HLA class II was expressed in the HL-60 cell line, and it was confirmed that the Jurkat cell line did not express HLA class II (see FIG. 4 ).
  • the HL-60 cell line expressing HLA class II among the two cells was used as a target cell, and Jurkat cells not expressing HLA class II were used as a negative control.
  • Example 4 chimeric antigen receptors and HLA Affinity measurement of human cell lines expressing class II
  • each of the extracellular domains No. IgG 1 heavy chain constant region
  • two extracellular domains of LAG-3 No. 1 to No. 4
  • the protein was purified by affinity chromatography (GE healthcare) using a protein A column (see FIG. 5a ).
  • the purified water-soluble chimeric antigen receptor was confirmed by Western blotting using an antibody recognizing the constant region of human IgG heavy chain (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody, abcam) (Fig. 5b).
  • the purified water-soluble chimeric antigen receptor was treated with a cell line expressing HLA class II (HL-60) and a cell line not expressing HLA class II (Jurkat) to prevent the water-soluble chimeric antigen receptor from binding to the cell.
  • HL-60 HLA class II
  • Jurkat Jurkat
  • the prepared chimeric antigen receptor had a high affinity with the human cell line expressing HLA class II, which was the MHC class II of the immune cell therapy prepared with HL-60 cells and Jurkat cells. This means that it can be used for specific cytotoxicity verification of proteins.
  • cytotoxic T cells were first isolated from human peripheral blood mononuclear cells. After purchasing human peripheral blood mononuclear cells (Medilab Korea), magnetic-activated cell sorting was used to isolate cytotoxic T cells. Peripheral blood mononuclear cells were 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 were re-conjugated 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 and cells attached thereto were passed through a magnetic seperation column (Miltenyi Biotec) to obtain cytotoxic T cells not labeled with the antibody.
  • a magnetic seperation column Miltenyi Biotec
  • flow cytometry using CD8 and CD3 ⁇ , which are cell surface factors of cytotoxic T cells was performed, and as a result, the purity was greater than 95%.
  • cytotoxic T cells For activation of isolated cytotoxic T cells, 1 ⁇ 10 6 pieces/ml of magnetic beads coated with human CD3 and CD28 antibodies (Thermo Fisher Scientific), and 10% calf serum supplemented with 100 U/ ⁇ l recombinant human IL-2 Cells were reconstituted in RPMI (Welgene) at a density of 1.5 ⁇ 10 6 pieces/ml and cultured for 24 hours after inoculation in a 24-well cell culture dish.
  • 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 to become 2.5 ⁇ 10 6 cells in a 100 ⁇ cell culture dish, and then 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 LAG-3.CAR-T.pCDH-CMV-MCS-EF1-copGFP vector DNA is mixed with 10 ⁇ g of psPAX2 (Addgene) and 3 ⁇ g pMD2.G (Addgene) vector was crystallized using calcium phosphate and Hepes-buffered solution, and then introduced into 293FT cells.
  • the culture supernatant containing the lentivirus was collected at intervals of 24 hours after 48 hours of the 293FT cells introduced with the expression vector.
  • the collected supernatant was centrifuged at 21,000 rpm in an ultra-high speed centrifuge for 2 hours to concentrate the virus.
  • the concentrated virus was mixed with 4 ⁇ g/ml of polybrene and added to the culture of activated cytotoxic T cells, the transduction was performed by centrifugation at 1,800 g for 75 minutes using a centrifuge. After centrifugation, the cytotoxic T cells were further cultured for 4 hours and then replaced with an RPMI culture medium containing 10% calf serum. After 48 hours, some of the transduced cytotoxic T cells were used to measure the transduction efficiency.
  • Transduction efficiency was measured by using flow cytometry to measure the expression level of GFP inside the cells, and cytotoxic T cells (Mock) transduced with a virus prepared by using the transduced cytotoxic T cells (LAG-3 CAR-T cells) as an empty vector. /T cells) and the results compared with the transduction ratio is shown in Figure 6a.
  • 293GPG cells were dissolved in 10 ml of 10 ml of 10% calf serum-containing DMEM culture medium and inoculated in a 100 ⁇ cell culture dish and then cultured for 24 hours.
  • 20 ⁇ g of the previously prepared recombinant vector (LAG-3.CAR-NK.pLNCX2 vector) was crystallized using calcium phosphate and Hepes-buffered solution, and then added to the culture medium of the previously cultured 293GPG cells. Thereafter, the culture medium was changed over 72 hours at 24 hour intervals, and the culture supernatant of 293GPG cells containing retrovirus was collected and stored.
  • 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 into a 24 well cell culture dish at a density of 5 ⁇ 10 5 /ml.
  • a mixed solution of 8 ug/ml of polybrene added to the concentrated retrovirus was added to the culture medium of NK92MI cells in culture and cultured for 24 hours. After culturing, the culture medium was replaced with a new culture medium.
  • the Myelocult H5100 culture medium supplemented with neomycin antibiotic 600 ⁇ g/ml was used to culture the retrovirus-treated NK92MI cells. After 14 days of neomycin selection, NK92MI cells were transferred to fresh culture medium and proliferated for 1 week.
  • NK92MI cells After proliferation, the LAG-3 expression level of NK92MI cells was measured using flow cytometry. NK92MI cells introduced with LAG-3.CAR-NK.pLNCX2 (LAG-3 CAR-NKcell) and NK92MI cells introduced with the empty vector pLNCX2 ( The results of comparing the LAG-3 expression level of Mock CAR-NK cells using a flow cytometer using an anti-human LAG-3 antibody (Novus biologicals) are shown in FIG. 6b .
  • Example 8 MHC class II+ cell-specific cytotoxicity verification of CAR-T cells
  • the target cells selected in Example 3 HL-60 cells (HLA class II positive cells) and Jurkat cells (HLA class II negative cells) were co-cultured with chimeric antigen receptor-expressing cytotoxic T cells (LAG-3 CAR-T cells) or cytotoxic T cells (Mock T cells) introduced with an empty vector for 6 hours.
  • a non-radioactive cytotoxicity assay was used to measure the degree of cytotoxicity of cytotoxic T cells to target cells by the amount of lactate dehydrogenase present in the supernatant after co-culture.
  • cytotoxic T cells (1x10 5 cells) and target cells (1x10 4 cells) into the wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, and inoculate so that the volume per well becomes 100 ⁇ l Centrifuge for 4 minutes at 250 g using a centrifuge to close the intercellular space. After culturing for 6 hours, 50 ⁇ l of the supernatant from each well is removed, transferred to a transparent 96-well dish for absorbance measurement, and treated with an analysis solution and 1M hydrochloric acid solution to proceed and stop the enzyme reaction.
  • the absorbance in the 490 nm wavelength band was measured and numerically converted using a fluorescence/luminescence/absorption meter (multi-detection plate reader) to quantify the degree of cytotoxicity of cytotoxic T cells in each well.
  • the prepared chimeric antigen receptor-expressing cytotoxic T cells (LAG-3 CAR-T cells) showed about 0.5% cytotoxicity to Jurkat cells (HLA class II negative cells), whereas , It was confirmed that HL-60 cells (HLA class II positive cell) showed high cytotoxicity of 22% or more (p ⁇ 0.01). On the other hand, in the case of cytotoxic T cells introduced with the empty vector as a control, it was confirmed not to have cytotoxicity against Jurkat cells (HLA class II negative cells) or HL-60 cells (HLA class II positive cells).
  • the chimeric antigen recognition receptor-expressing cytotoxic T cells containing the LAG-3 extracellular domain showed cytotoxicity specific to the MHC class II protein, and thus the related cancer cells using the CAR-T T cells. It was confirmed that treatment was possible.
  • Example 9 MHC class II+ cell-specific cytotoxicity verification of CAR-NK cells
  • HL-60 cells HLA class II positive cells
  • Jurkat cells selected in Example 3 HLA class II negative cells
  • NK cells chimeric antigen receptor-expressing 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 after co-culture.
  • the absorbance of the 490 nm wavelength band was measured and numerically converted using a fluorescence/luminescence/absorption meter (multi-detection plate reader) to quantify the degree of cytotoxicity of NK cells in each well.
  • the prepared chimeric antigen receptor-expressing NK cells (LAG-3 CAR-NK cells) showed almost no cytotoxicity to Jurkat cells (HLA class II negative cells), whereas HL-60 It was confirmed that the cells (HLA class II positive cell) showed a very high cytotoxicity of more than 47% (p ⁇ 0.01).
  • cytotoxic T cells introduced with the empty vector as a control, it was confirmed not to have cytotoxicity against Jurkat cells (HLA class II negative cells) or HL-60 cells (HLA class II positive cells).
  • the chimeric antigen-recognizing receptor-expressing NK cells containing the LAG-3 extracellular domain showed cytotoxicity specific to the MHC class II protein, so that it is possible to treat the related cancer cells using the CAR-NK cells. was able to confirm

Landscapes

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

Abstract

The present invention relates to: a chimeric antigen receptor specifically binding to HLA class II; a polynucleotide encoding a protein of the chimeric antigen receptor; a vector comprising the polynucleotide; a cell transformed with the vector; and a cell therapy product or a pharmaceutical composition for treating cancer, comprising the cell as an active ingredient. The chimeric antigen receptor according to the present invention comprises extracellular domains (1 to 4) of LAG-3 that specifically binds to HLA class II (HLA-DP, HLA-DQ, HLA-DR). Therefore, in the case of cytotoxic T cells or natural killer cells that are transformed with a vector that can overexpress the chimeric antigen receptor, the cells have cytotoxicity specifically to carcinoma expressing HLA class II (HLA-DP, HLA-DQ, HLA-DR), and thus the cells may be usefully utilized as immune cell therapeutic agents for cancer treatment.

Description

HLA CLASS II에 특이적으로 결합하는 키메릭 항원 수용체 및 이의 용도Chimeric antigen receptors specifically binding to HLA CLASS II and uses thereof
본 발명은 HLA class II에 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); 상기 세포를 유효성분으로 포함하는 세포 치료제 또는 암의 치료용 약학적 조성물에 관한 것이다.The present invention relates to a chimeric antigen receptor (CAR) that specifically binds to HLA class II; a polynucleotide encoding the chimeric antigen receptor protein; a vector comprising the polynucleotide; T cells or natural killer cells transformed with the vector (Natural killer cell, NK cell); It relates to a cell therapeutic agent or a pharmaceutical composition for the treatment of cancer comprising the cell as an active ingredient.
인간의 주조직적합성(major histocompatibility complex, MHC) 항원 class II는 HLA-DP, HLA-DQ, HLA-DR로 구성되어 있고, 이형이합체(heterodimer)로 구성되어 있다. HLA-DP의 경우 HLA-DPα와 HLA-DPβ, HLA-DQ의 경우 HLA-DQα와 HLA-DQβ, HLA-DR의 경우 HLA-DRα와 HLA-DRβ로 구성되어진 내재성 막 단백질(integral membrane protein)이다. HLA class II는 주로 전문적 항원표출세포(professional antigen-presenting cell)에서 발현하며 펩티드 항원을 CD4+ T 세포(helper T cell)에 전달해 주는 역할을 한다. 하지만, HLA class II는 정상 세포가 종양화되면 다양한 암종에서 비정상적으로 높은 발현을 하게 된다. 다양한 암종에서 HLA class II는 면역관문억제제(immune checkpoint inhibitor)의 작용을 block할 수 있다(Johnson et al., 2016; Johnson et al., 2018). 즉 HLA class II는 HLA class II의 리간드인 LAG-3 또는 Fc receptor-like 6(FCRL6)와 각각 결합하여 자연살생세포(natural killer cell, NK cell) 또는 세포독성 T 세포(cytotoxic T lymphocytes, CTL)나 도움 T 세포(helper T lymphocytes)의 활성을 억제한다(Johnson et al., 2016; Johnson et al., 2018). Human major histocompatibility complex (MHC) antigen class II consists of HLA-DP, HLA-DQ, and HLA-DR, and consists of a heterodimer. Integral membrane protein composed of HLA-DPα and HLA-DPβ for HLA-DP, HLA-DQα and HLA-DQβ for HLA-DQ, and HLA-DRα and HLA-DRβ for HLA-DR to be. HLA class II is mainly expressed in professional antigen-presenting cells and plays a role in delivering peptide antigens to CD4 + T cells (helper T cells). However, HLA class II is expressed abnormally high in various carcinomas when normal cells are tumorigenic. In various carcinomas, HLA class II can block the action of immune checkpoint inhibitors (Johnson et al., 2016; Johnson et al., 2018). That is, HLA class II binds to HLA class II ligand LAG-3 or Fc receptor-like 6 (FCRL6), respectively, to form natural killer cells (NK cells) or cytotoxic T lymphocytes (CTLs). Inhibits the activity of or helper T lymphocytes (Johnson et al., 2016; Johnson et al., 2018).
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 CD3ε 또는 다른 단백질의 세포질 내 신호전달 도메인(cytoplasmic signaling domain)에 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, studies on anticancer treatment using cytotoxic T cells or natural killer cells have either delivered specific cancer antigens derived from a patient to the patient's cytotoxic T cells to induce activity, or induce antibody-dependent cellular cytotoxicity. (Galluzzi et al., 2018; Lu et al., 2020). However, recently, a new anticancer treatment method using cytotoxic T cells and natural killer cells has been introduced. That is, a single-chain variable fragment (scFv) portion of an antigen-recognizing antibody is grafted onto the cytoplasmic signaling domain of CD3ε or other protein, chimeric antigen receptor, CAR) was tried as a method of delivering. When the chimeric antigen receptor is grafted onto T cells or natural killer cells, the anticancer action of T cells or natural killer cells is enhanced by the specific antigen recognition of single-chain Fv fragments 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. Indeed, these chimeric antigen receptor-expressing T cells or natural killer cells have shown efficacy in several cancers (Holzinger et al., 2016; Pettitt et al., 2018; Wang et al., 2020).
한편, lymphocyte activation gene-3 (LAG-3; CD223)은 T 세포가 활성화될 때 세포 표면에 발현하는 면역 글로불린(Ig) 수퍼 패밀리(immunoglobulin superfamily)에 속하는 내재성 막 단백질(integral membrane protein)이다. 활성화된 T 세포에서 발현되는 LAG-3는 T 세포의 활성을 억제시키는 것으로 알려져 있다. LAG-3는 구조적으로 CD4와 유사하여 MHC class II에 결합하지만, CD4보다 MHC class II에 대한 훨씬 높은 친화도를 가진다. 또한, 최근 연구 결과에 의하면 사람 LAG-3는 사람 MHC class II인 HLA-DP, HLA-DQ, HLA-DR에 모두 매우 높은 친화도를 보인다(Niehrs et al., 2019).On the other hand, lymphocyte activation gene-3 (LAG-3; CD223) is an integral membrane protein belonging to the immunoglobulin (Ig) superfamily that is expressed on the cell surface when T cells are activated. LAG-3 expressed in activated T cells is known to inhibit the activity of T cells. LAG-3 is structurally similar to CD4 and binds to MHC class II, but has a much higher affinity for MHC class II than CD4. In addition, according to recent research results, human LAG-3 shows very high affinity to all of human MHC class II, HLA-DP, HLA-DQ, and HLA-DR (Niehrs et al., 2019).
현재, 암 면역요법을 위한 키메릭 항원 수용체(한국공개특허 제10-2017-0062446호)나 LAG-3의 막관통 부위(transmembrane region)를 포함하는 키메릭 항원 수용체가 공개되어 있으나(미국공개특허 제2017-0275362호), HLA class II가 발현되는 암세포에 대한 결합특이성을 갖는 LAG-3 세포외 도메인(extracellular domain)을 항원 결합 도메인에 포함하여 면역 항암 치료제로서 이용하는 발명에 대해서는 기재된 바 없다.Currently, a chimeric antigen receptor for cancer immunotherapy (Korean Patent Application Laid-Open No. 10-2017-0062446) or a chimeric antigen receptor including a transmembrane region of LAG-3 has been disclosed (US Patent Publication) No. 2017-0275362), the invention of using the LAG-3 extracellular domain having binding specificity for HLA class II-expressing cancer cells as an antigen-binding domain as an immunocancer therapeutic agent has not been described.
이러한 배경 하에, 본 발명자들은 다양한 암세포에서의 HLA class II 발현이 확인 또는 유도된다는 점과, 상기 HLA class II와 LAG-3가 결합이 가능하다는 점을 이용하여 LAG-3의 세포외 도메인(1번부터 4번)을 사용한 HLA class II를 발현하는 암 특이적 키메릭 항원 수용체 발현 T 세포와 자연살생세포를 제작하고, 상기 T 세포 또는 자연살생세포가 HLA class II를 발현하는 세포에 특이적으로 세포독성 능력이 있음을 실험을 통해 확인하였다. 이때, LAG-3의 세포외 도메인(1번부터 4번) 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 LAG-3의 세포외 도메인(1번부터 4번) 각각을 인간 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)과 연결시켜, 키메릭 항원 수용체 하나당 2개의 LAG-3의 세포외 도메인(1번부터 4번)이 항원을 인식하게 하여, 신호 전달 강도를 증폭하고자 하였다.Under this background, the present inventors used the fact that HLA class II expression in various cancer cells is confirmed or induced, and that the HLA class II and LAG-3 can bind to the extracellular domain of LAG-3 (No. 1 to 4) were used to prepare cancer-specific chimeric antigen receptor-expressing T cells and natural killer cells expressing HLA class II, and the T cells or natural killer cells specifically target cells expressing HLA class II. It was confirmed through an experiment that it has a toxic ability. At this time, the signal transduction strength was amplified by making a fusion protein linking each of the extracellular domains (No. 1 to No. 4) of LAG-3 with the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region). That is, the existing chimeric antigen receptor has one site for recognizing an antigen by a single chain Fv fragment, whereas the chimeric antigen receptor designed by the present inventors has the extracellular domain of LAG-3 (No. 1 to No. 4). By linking each with human human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region), two extracellular domains of LAG-3 (No. 1 to No. 4) per chimeric antigen receptor recognize antigen Therefore, it was attempted to amplify the signal transduction strength.
따라서 본 발명의 목적은 HLA class II에 특이적으로 결합하는 키메릭 항원 수용체를 제공하는 것이다.Accordingly, it is an object of the present invention to provide a chimeric antigen receptor that specifically binds to HLA class II.
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 HLA class II를 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 therapeutics or HLA class II, including the transformed T cells or natural killer cells.
상기와 같은 본 발명의 목적을 달성하기 위해서, 본 발명은 항원 결합 도메인; 막관통 도메인; 및 세포 내 신호전달 도메인을 포함하는 키메릭 항원 수용체(CAR)로서, 상기 항원 결합 도메인은 HLA class II에 특이적으로 결합하는 LAG-3의 세포외 도메인(1번부터 4번)을 포함하는 것인, 키메릭 항원 수용체를 제공한다.In order to achieve the object of the present invention as described above, the present invention provides an antigen-binding domain; transmembrane domain; and an intracellular signaling domain, wherein the antigen binding domain comprises an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II. A phosphorus, chimeric antigen receptor is provided.
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 HLA class II를 발현하는 암세포를 사멸시키는 암의 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for the treatment of cancer comprising the transformed T cells or natural killer cells, a cell therapeutic agent or HLA class II expressing cancer cells.
본 발명에 따른 키메릭 항원 수용체는 HLA class II(HLA-DP, HLA-DQ, HLA-DR)에 특이적으로 결합하는 LAG-3의 세포외 도메인(1번부터 4번)을 포함한다. 따라서, 상기 키메릭 항원 수용체를 과발현시킬 수 있는 벡터로 형질전환된 세포독성 T 세포 또는 자연살생세포의 경우 HLA class II(HLA-DP, HLA-DQ, HLA-DR)를 발현하는 암종에 특이적으로 세포독성을 갖게되므로 암 치료를 위한 면역세포 치료제로서 유용하게 이용될 수 있다.The chimeric antigen receptor according to the present invention includes an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II (HLA-DP, HLA-DQ, HLA-DR). Therefore, in the case of cytotoxic T cells or natural killer cells transformed with a vector capable of overexpressing the chimeric antigen receptor, it is specific for HLA class II (HLA-DP, HLA-DQ, HLA-DR) expressing carcinoma. Because it has cytotoxicity, it can be usefully used as an immune cell therapeutic for cancer treatment.
도 1은 본 발명의 일 실시예에 따른 키메릭 항원 수용체를 발현시키는 각 도메인의 cDNA 구역을 나타낸 모식도이다. 1 is a schematic diagram showing the cDNA region 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 타겟인 HLA class II를 발현하는 암세포(HLA class II positive cell)와 HLA class II를 발현하지 않는 암세포(HLA class II negative cell)에서 HLA class II의 발현량을 유세포 분석을 이용해 측정한 결과를 나타낸 도이다.4 is a view of HLA class II in cancer cells expressing HLA class II (HLA class II positive cells) and cancer cells not expressing HLA class II (HLA class II negative cells), which are targets of the chimeric antigen receptor provided in the present invention. It is a diagram showing the result of measuring the expression level using flow cytometry.
도 5a는 본 발명에서 제공하는 키메릭 항원 수용체를 수용성으로 만들어 Chinese hamster ovary (CHO) 세포에서 발현시킨 후, protein A column을 이용하여 친화크로마토그래피(affinity chromatography)로 정제한 결과를 나타낸 도이다.5a is a diagram showing the results of purification by affinity chromatography using a protein A column after making the chimeric antigen receptor provided in the present invention water-soluble and expressing it in Chinese hamster ovary (CHO) cells.
도 5b는 본 발명의 일 실시예에 따른 수용성 키메릭 항원 수용체를 인간 IgG 중쇄의 불변 영역을 인식하는 항체(항 인간 IgG 중쇄 불변영역 항체, anti-human IgG Fc region antibody)를 이용하여 웨스턴 블로팅으로 확인한 결과를 나타낸 도이다.Figure 5b is a water-soluble chimeric antigen receptor according to an embodiment of the present invention using an antibody recognizing the constant region of a human IgG heavy chain (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody) Western blotting It is a diagram showing the results confirmed by .
도 5c는 본 발명의 일 실시예에 따른 수용성 키메릭 항원 수용체를 세포외 도메인 타겟인 HLA class II를 발현하는 암세포(HLA class II positive cell)를 인식하는지 여부를 유세포 분석을 이용해 나타낸 도이다.FIG. 5c is a diagram showing whether the water-soluble chimeric antigen receptor according to an embodiment of the present invention recognizes cancer cells (HLA class II positive cells) expressing the extracellular domain target, HLA class II, using flow cytometry.
도 6a는 본 발명의 일 실시예에 따른 발현 벡터를 렌티바이러스 시스템을 이용하여 형질도입시킨 세포독성 T 세포(LAG-3 CAR-T cell)에서의 GFP의 발현 비율을 비교한 결과를 나타낸 도이다.Figure 6a is a diagram showing the result of comparing the expression rate of GFP in cytotoxic T cells (LAG-3 CAR-T cell) transduced with the expression vector according to an embodiment of the present invention using a lentiviral system. .
도 6b는 본 발명의 일 실시예에 따른 발현 벡터를 레트로바이러스 시스템을 이용하여 형질도입시킨 자연살생세포(LAG-3 CAR-NK cell)에서의 LAG-3의 발현 비율을 비교한 결과를 나타낸 도이다.Figure 6b is a diagram showing the result of comparing the expression ratio of LAG-3 in natural killer cells (LAG-3 CAR-NK cells) transduced with the expression vector according to an embodiment of the present invention using a retroviral system to be.
도 7a는 본 발명의 일 실시예에 따른 세포독성 T 세포(LAG-3 CAR-T cell) 또는 공벡터를 형질도입시킨 세포독성 T 세포(Mock T cell)를 작동 세포(effector cell)로 하여 HLA class II를 발현하는 암세포(HLA class II positive cell)에 대한 세포독성을 측정한 결과를 나타낸 도이다.Figure 7a shows HLA using cytotoxic T cells (LAG-3 CAR-T cells) or empty vector-transduced cytotoxic T cells (Mock T cells) as effector cells according to an embodiment of the present invention; It is a diagram showing the results of measuring the cytotoxicity to cancer cells expressing class II (HLA class II positive cells).
도 7b는 본 발명의 일 실시예에 따른 자연살생세포(LAG-3 CAR-NK cell) 또는 공벡터를 형질도입시킨 자연살생세포(Mock NK cell)를 작동 세포(effector cell)로 하여 HLA class II를 발현하는 암세포(HLA class II positive cell)에 대한 세포독성을 측정한 결과를 나타낸 도이다.7b is HLA class II using natural killer cells (LAG-3 CAR-NK cells) or mock NK cells transduced with an empty vector as effector cells according to an embodiment of the present invention; It is a diagram showing the result of measuring the cytotoxicity to cancer cells (HLA class II positive cells) expressing.
본 발명은 하나의 양태로서, 항원 결합 도메인; 막관통 도메인; 및 세포 내 신호전달 도메인을 포함하는 키메릭 항원 수용체(CAR)로서, 상기 항원 결합 도메인은 HLA class II에 특이적으로 결합하는 LAG-3의 세포외 도메인(1번부터 4번)을 포함하는, 키메릭 항원 수용체를 제공한다.The present invention provides an antigen-binding domain; transmembrane domain; and an intracellular signaling domain, wherein the antigen binding domain comprises an extracellular domain (Nos. 1 to 4) of LAG-3 that specifically binds to HLA class II. A chimeric antigen receptor is provided.
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 발현시 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, "chimeric antigen receptor (CAR)" binds to a desired antigen without the mediation of antigen presenting cells (APCs) or antibodies necessary for naturally activating T cells or natural killer cells, thereby producing an antigen-antibody reaction. It refers to a fusion protein for expression in T cells or natural killer cells in order to induce activation of T cells and attack cells expressing the corresponding antigen. That is, when expressed in T cells or natural killer cells, it can be considered as a protein that binds to an antigen and induces activation of these cells. Through this, it may be a protein recognizing an antigen specific to a cell to cause an immune response, and the cell to cause an immune response may refer to a cell existing in a specific tissue or constituting a tissue causing 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 HLA class II(HLA-DP, HLA-DQ, HLA-DR)에 특이적으로 결합할 수 있는 활성을 가진 것을 의미한다.The chimeric antigen receptor protein according to an embodiment of the present invention may include a functional equivalent. "Functional equivalent" means at least 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably the amino acid sequence of the chimeric antigen receptor protein as a result of the addition, substitution, or deletion of amino acids. refers to a protein having a sequence homology of 95% or more and exhibiting substantially homogeneous physiological activity. “Substantially homogenous physiological activity” means that it has an activity capable of specifically binding to HLA class II (HLA-DP, HLA-DQ, HLA-DR).
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 알려져 있다.The present invention also includes fragments, derivatives and analogues of chimeric antigen receptors. As used herein, the terms 'fragment', 'derivative' and 'analog' refer to a polypeptide that retains substantially the same biological function or activity as the chimeric antigen receptor protein of the present invention. Fragments, derivatives and analogs of the present invention include 1) polypeptides in which one or more conservative or non-conservative amino acid residues (preferably conservative amino acid residues) are substituted, wherein the substituted amino acid residues are encoded by the genetic code. may or may not) or 2) a polypeptide having substituent(s) at one or more amino acid residues, or 3) maturation associated with another compound (a compound capable of extending the half-life of the polypeptide, such as polyethylene glycol) a polypeptide derived from a polypeptide, or 4) the polypeptide associated with an additional amino acid sequence (eg, a leader sequence, a secretory sequence, a sequence used to purify the polypeptide, a proteinogen sequence or a fusion protein) It may be a polypeptide derived from The fragments, derivatives and analogs as defined herein are well known to those skilled in the art.
LAG-3(Lymphocyte-activation gene 3) 단백질은 림프구 활성화 유전자-3라고 불리며, 면역 글로불린(Ig) 수퍼패밀리(immunoglobulin superfamily)에 속하며, D1에서 D4로 지정된 4개의 도메인으로 구성된 세포외 도메인을 갖는 내재성 막 단백질(integral membrane protein)이다.Lymphocyte-activation gene 3 (LAG-3) protein, called lymphocyte activation gene-3, belongs to the immunoglobulin (Ig) superfamily, and has an extracellular domain consisting of four domains designated D1 to D4. It is an integral membrane protein.
상기 항원 결합 도메인은 신호전달을 증폭하기 위해 LAG-3의 세포외 도메인(1번부터 4번) 한쌍을 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 is a dimer ( dimer), but is not limited thereto.
상기 LAG-3의 세포외 도메인(1번부터 4번)은 서열번호 1 또는 이와 95% 이상의 상동성을 나타내는 아미노산 서열로 이루어진 것일 수 있으나, 이에 제한되는 것은 아니다.The extracellular domain (No. 1 to No. 4) of the LAG-3 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 showing 95% or more homology thereto, but is not limited thereto.
본 발명에서 "HLA class II"는 주로 전문적 항원표출세포(professional antigen presenting cell)에서 발현하며 펩티드 항원을 CD4+ T 세포(helper T cell)에 전달해 주는 내재성 막 단백질이지만, HLA class II는 정상 세포가 종양화되면 다양한 암종에서 비정상적으로 높은 발현을 하게 된다. 지금까지 HLA class II를 발현하는 암종은 머리 및 목 편평 세포 암종(Head and neck squamous cell carcinoma)(Meissner et al., 2008), 흉선종(thymoma) (Strobel et al., 2008) 식도 편평 세포 암종(Esophageal squamous cell carcinoma) (Hu et al., 2014), 흑색종(melanoma) (Johnson et al., 2016; Rodig et al., 2018; Johnson et al., 2018), 유방암(breast cancer) (Park et al., 2017; Loi et al., 2016; Bartek et al., 1987), 결장암(colorectal cancer) (Michel et al., 2010; Bustin et al., 2001), 난소암(ovarian cancer) (callahan et al., 2008, Turner et al., 2017), 전립선암(prostate cancer) (Younger et al., 2008), 신경아 세포종(neuroblstoma) (Yazawa et al., 2002), 신경교종(glioma) (Soos et al., 2001), 비소세포형 폐암(non-small cell lung cancer) (Yazawa et al., 1999), 클래식 호지킨 림프종(classic Hodgkin lymphoma) (Roemer et al., 2018), T 세포 백혈병/림프종(T-cell leukemia/lymphoma) (Takeuchi et al., 2019), B세포 림프종 (B cell lymphoma) (Steidl et al., 2011), 만성골수성 백혈병(chronic myeloid leukemia) (Petersdorf et al., 2001), 만성림프성 백혈병(chronic lymphocytic leukemia) (Hojjat-Farsangi et al., 2008), 급성 골수성 백혈병(acute myeloid leukemia) (van den Ancker et al., 2014), 급성 림프구성 백혈병(acute lymphoid leukemia) (Hurwitz et al., 1988) 등이 있다. 또한, 모든 HLA class II(HLA-DP, HLA-DQ, HLA-DR)는 LAG-3의 세포외 도메인(도메인 1번부터 4번)과 강하게 결합하는 특징을 가진다.In the present invention, "HLA class II" is an intrinsic membrane protein that is mainly expressed in professional antigen presenting cells and delivers peptide antigens to CD4+ T cells (helper T cells), but HLA class II is When it becomes oncogenic, it has abnormally high expression in various carcinomas. To date, carcinomas expressing HLA class II have been classified as head and neck squamous cell carcinoma (Meissner et al., 2008), thymoma (Strobel et al., 2008) and esophageal squamous cell carcinoma ( Esophageal squamous cell carcinoma) (Hu et al., 2014), melanoma (Johnson et al., 2016; Rodig et al., 2018; Johnson et al., 2018), breast cancer (Park et al.) al., 2017; Loi et al., 2016; Bartek et al., 1987), colorectal cancer (Michel et al., 2010; Bustin et al., 2001), ovarian cancer (callahan et al.) al., 2008, Turner et al., 2017), prostate cancer (Younger et al., 2008), neuroblstoma (Yazawa et al., 2002), glioma (Soos et al.) al., 2001), non-small cell lung cancer (Yazawa et al., 1999), classic Hodgkin lymphoma (Roemer et al., 2018), T cell leukemia/lymphoma (T-cell leukemia/lymphoma) (Takeuchi et al., 2019), B cell lymphoma (Steidl et al., 2011), chronic myeloid leukemia (Petersdorf et al., 2001) , chronic lymphocytic leukemia (Hojjat-Farsangi et al., 2008), acute bone acute myeloid leukemia (van den Ancker et al., 2014), acute lymphoid leukemia (Hurwitz et al., 1988), and the like. In addition, all HLA class II (HLA-DP, HLA-DQ, HLA-DR) has the characteristic of strongly binding to the extracellular domain of LAG-3 (domains 1 to 4).
일 실시예에서, 상기 막관통 도메인은 CD28, CD3 엡실론, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 및 CD154로 이루어진 군으로부터 선택되는 1종 이상을 포함할 수 있으며, 바람직하게는, CD8의 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, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137 and CD154. It may include one or more, 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, but 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" refers to a site that activates an immune response of an immune cell 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 암세포, 특히 HLA class II((HLA-DP, HLA-DQ, HLA-DR)를 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 high activity in cancer cells, particularly HLA class II ((HLA-DP, HLA-DQ, HLA- DR) expressing cancer cells.
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 이루어질 수 있다.The CD28 is SEQ ID NO: 4 or 70% or more, preferably 80% or more, more preferably 90% or more, even more preferably 95% or more sequence homology thereto, and the amino acid sequence represented by SEQ ID NO: 4 and may consist of an amino acid sequence exhibiting a substantially equivalent function, and 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% thereof Having the above sequence homology, it may consist of an amino acid sequence exhibiting a function substantially equivalent to the amino acid sequence shown in SEQ ID NO: 5, CD3 zeta functions as an NK cell activation domain, and SEQ ID NO: 6 or 70 thereof % or more, preferably 80% or more, more preferably 90% or more, even more preferably 95% or more, and an amino acid sequence that exhibits a substantially equivalent function to the amino acid sequence represented by SEQ ID NO: 6. can be done
상기 항원 결합 도메인은 LAG-3 신호 펩타이드를 포함할 수 있으며, 상기 LAG-3 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 LAG-3 signal peptide, and the LAG-3 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. In another aspect according to the present invention, there is provided a polynucleotide encoding the chimeric antigen receptor protein.
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 the codon preferred in the organism to express the antigen receptor. Various modifications may be made to the coding region within the range that does not occur, and various modifications or modifications may be made within the range that does not affect the expression of the gene in parts other than the coding region, 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 equivalent activity, 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 according to the present invention, there is provided a vector comprising the polynucleotide, and a cell transformed with the vector.
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.The vector used in the present invention may use a variety of vectors known in the art, and may include a promoter, a terminator, an enhancer, etc., depending on the type of host cell to produce the antigen receptor. Expression control sequences, sequences for membrane targeting or secretion, etc. can be appropriately selected and variously combined according to the purpose. The vector of the present invention includes, but is not limited to, a plasmid vector, a cosmid vector, a bacteriophage vector, and a viral vector. Suitable vectors include a signal sequence or leader sequence for membrane targeting or secretion in addition to expression control elements such as promoter, operator, start codon, stop codon, polyadenylation signal and enhancer, 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.As a preferred embodiment of the present invention, a vector for a lenti-virus or a vector for a retro-virus can be used. pLNCX2 (retro-virus vector) was used, but is not limited thereto.
일 일시예에서, HLA class II에 특이적으로 결합하는 키메릭 항원 수용체를 상기 벡터를 통해 세포에 도입하여 세포를 형질전환시킬 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 자연살생세포일 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, a cell can be transformed by introducing a chimeric antigen receptor that specifically binds to HLA class II into the cell through the vector. The cell may be a T cell, a tumor infiltrating lymphocyte, a B cell, a natural killer cell, or an NKT cell, preferably a cytotoxic T cell or a natural killer cell. The cell may be obtained or prepared from bone marrow, peripheral blood, peripheral blood mononuclear cells or umbilical cord blood, and the cell may be a human cell, but is not limited thereto.
상기와 같이 본 발명의 키메릭 항원 수용체가 도입되어 형질전환된 세포는 HLA class II((HLA-DP, HLA-DQ, HLA-DR)를 항원으로 인식하고 이와 강하게 결합하는 특징을 가진다.As described above, the transformed cells into which the chimeric antigen receptor of the present invention is introduced recognizes HLA class II ((HLA-DP, HLA-DQ, HLA-DR) as an antigen and strongly binds 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 타겟 세포의 세포자살을 유도하여 세포독성을 나타낸다.In the present invention, "chimeric antigen receptor T cells (hereinafter abbreviated as 'CAR-T cells')" or "chimeric antigen receptor NK cells (hereinafter referred to as chimeric antigen receptor NK cells)" 'CAR-NK cells') is a chimeric antigen receptor that specifically responds to cancer cells other than the original T cell receptor or NK cell receptor by transducing normal T cells or natural killer cells, etc. refers to T cells or NK cells expressing T cells or NK cells having this receptor induce apoptosis of target cells, thereby exhibiting cytotoxicity.
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 HLA class II(HLA-DP, HLA-DQ, HLA-DR)를 발현하는 암세포를 인지하여 효과적으로 파괴할 수 있다.In the present invention, in particular, CAR-T cells or CAR-NK cells may be cytotoxic T cells or cells in which the chimeric antigen receptor of the present invention is introduced into NK cells. The cells have the advantage of anticancer-specific targeted therapy, which is the existing advantage of CAR-T therapeutics. In particular, the CAR-T cells or CAR-NK cells of the present invention are HLA class II (HLA-DP, HLA-DQ, HLA- It can recognize and effectively destroy cancer cells expressing DR).
따라서 본 발명은 또 다른 하나의 양태로서, 상기 세포를 포함하는 세포 치료제 또는 이를 유효성분으로 포함하는 암의 치료용 약학적 조성물을 제공한다. Therefore, as another aspect, the present invention provides a cell therapeutic agent comprising the cell or a pharmaceutical composition for the treatment of cancer comprising 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 therapeutic" refers to cells and tissues prepared through isolation, culture, and special manipulation from an individual, and as pharmaceuticals used for therapeutic purposes, living autologous, allogeneic, or xenogeneic cells or tissues are restored to function. It refers to a drug used for therapeutic purposes through a series of actions, such as in vitro proliferation and selection or changing the biological properties 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 any action in which symptoms of 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 2 종 이상을 혼합하여 사용될 수 있다.The "pharmaceutically acceptable carrier" may mean a carrier or diluent that does not inhibit the biological activity and properties of the injected compound without irritating the organism. The type of carrier usable in the present invention is not particularly limited, and any carrier commonly used in the art and pharmaceutically acceptable may 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 may be used 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 comprising a pharmaceutically acceptable carrier may be in various oral or parenteral formulations. In the case of formulation, it is prepared using diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrants, and surfactants that are usually used.
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, etc., and these solid preparations include at least one excipient to the compound, for example, starch, calcium carbonate, sucrose, lactose. , gelatin, etc. may be mixed and prepared. In addition to simple excipients, lubricants such as magnesium stearate and talc may also be used. Liquid formulations for oral use include suspensions, solutions, emulsions, and syrups. In addition to water and liquid paraffin, which are commonly used simple diluents, various excipients such as wetting agents, sweeteners, fragrances, and preservatives may be included. have. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilized formulations and suppositories. Non-aqueous solvents and suspending agents include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate. As the base of the suppository, Witepsol, Macrogol, Tween 61, cacao butter, laurin fat, glycerogelatin, etc. may be used.
상기 조성물은 약학적으로 유효한 양으로 투여할 수 있다.The composition may 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 subject's type and severity, age, sex, infected virus type, and drug. Activity, sensitivity to drug, time of administration, route of administration and excretion rate, duration of treatment, factors including concomitant drugs and other factors well known in the medical field.
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 the 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, may be administered intranasally, 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 HLA class II를 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 two to three times. When the two active ingredients are single drugs, the number of administrations may be the same or different. In addition, the composition of the present invention can be used alone or in combination with other drug treatments for the treatment of HLA class II-expressing cancer. Taking all of the above factors into consideration, it is important to administer an amount that can obtain the maximum effect with a minimum amount without side effects, and can be easily determined by those skilled in the art.
상기 개체란, HLA class II를 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 is a human, monkey, cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig that has or can develop cancer expressing HLA class II. all animals, including If the disease can be effectively treated by administering the pharmaceutical composition of the present invention to the subject, the type of subject is included without limitation.
본 발명에 있어 치료 대상이 되는 암의 종류로는 머리 및 목 편평 세포 암종(Head and neck squamous cell carcinoma), 흉선종(thymoma), 식도 편평 세포 암종(Esophageal squamous cell carcinoma), 흑색종(melanoma), 유방암(breast cancer), 결장암(colorectal cancer), 난소암(ovarian cancer), 전립선암(prostate cancer), 신경아 세포종(neuroblstoma), 신경교종(glioma), 비소세포형 폐암(non-small cell lung cancer), 클래식 호지킨 림프종(classic Hodgkin lymphoma), T 세포 백혈병/림프종(T-cell leukemia/lymphoma), B세포 림프종 (B cell lymphoma), 만성골수성 백혈병(chronic myeloid leukemia), 만성림프성 백혈병(chronic lymphocytic leukemia) , 급성 골수성 백혈병(acute myeloid leukemia) 또는 급성 림프구성 백혈병(acute lymphoid leukemia)일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the types of cancer to be treated include head and neck squamous cell carcinoma, thymoma, esophageal squamous cell carcinoma, melanoma, Breast cancer, colorectal cancer, ovarian cancer, prostate cancer, neuroblstoma, glioma, non-small cell lung cancer , classic Hodgkin lymphoma, T-cell leukemia/lymphoma, B cell lymphoma, chronic myeloid leukemia, chronic lymphocytic leukemia), acute myeloid leukemia, or acute lymphoid leukemia, but is 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 illustrating 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 LAG-3의 세포외 도메인(1번부터 4번) 한쌍을 각각 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)에 연결시킨 항원 인식 부위와 CD8α의 막관통 도메인, CD28, 4-1BB, CD3-zeta 각각의 세포내 도메인 부분의 단백질 암호화 서열을 유전자 데이터베이스(database)에서 확인하였다. 상기 LAG-3의 세포외 도메인 1번부터 4번의 구체적인 서열 정보는 다음과 같다.In order to prepare the chimeric antigen receptor of the present invention, a pair of extracellular domains (No. 1 to No. 4) of LAG-3 are each linked to the human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region) for antigen recognition The protein coding sequence of the region and the transmembrane domain of CD8α, the intracellular domain of CD28, 4-1BB, and CD3-zeta, respectively, was identified in the gene database. Specific sequence information of extracellular domains 1 to 4 of the LAG-3 is as follows.
LAG-3 세포외 도메인 1번부터 4번의 서열목록Sequence list of LAG-3 extracellular domains 1 to 4
D1D1 CTC CAG CCA GGG GCT GAG GTC CCG GTG GTG TGG GCC CAG GAG GGG GCT CCT GCC CAG CTC CCC TGC AGC CCC ACA ATC CCC CTC CAG GAT CTC AGC CTT CTG CGA AGA GCA GGG GTC ACT TGG CAG CAT CAG CCA GAC AGT GGC CCG CCC GCT GCC GCC CCC GGC CAT CCC CTG GCC CCC GGC CCT CAC CCG GCG GCG CCC TCC TCC TGG GGG CCC AGG CCC CGC CGC TAC ACG GTG CTG AGC GTG GGT CCC GGA GGC CTG CGC AGC GGG AGG CTG CCC CTG CAG CCC CGC GTC CAG CTG GAT GAG CGC GGC CGG CAG CGC GGG GAC TTC TCG CTA TGG CTG CGC CCA GCC CGG CGC GCG GAC GCC GGC GAG TAC CGC GCC GCG GTG CAC CTC AGG GAC CGC GCC CTC TCC TGC CGC CTC CGT CTG CGC CTG GGCCTC CAG CCA GGG GCT GAG GTC CCG GTG GTG TGG GCC CAG GAG GGG GCT CCT GCC CAG CTC CCC TGC AGC CCC ACA ATC CCC CTC CAG GAT CTC AGC CTT CTG CGA AGA GCA GGG GTC ACT TGG CAG CAT CAG CCA GAC AGT GGC CCG C GCT GCC GCC CCC GGC CAT CCC CTG GCC CCC GGC CCT CAC CCG GCG GCG CCC TCC TCC TGG GGG CCC AGG CCC CGC CGC TAC ACG GTG CTG AGC GTG GGT CCC GGA GGC CTG CGC AGC GGG AGG CTG CCC CTG CAG CTGGC GTC CAG CCC CGC GTC GAT GAG CGC GGC CGG CAG CGC GGG GAC TTC TCG CTA TGG CTG CGC CCA GCC CGG CGC GCG GAC GCC GGC GAG TAC CGC GCC GCG GTG CAC CTC AGG GAC CGC GCC CTC TCC TGC CGC CTC CGT CTG CGC CTG GGC Ig-like V domain (435bp)Ig-like V domain (435bp)
D2D2 CAG GCC TCG ATG ACT GCC AGC CCC CCA GGA TCT CTC AGA GCC TCC GAC TGG GTC ATT TTG AAC TGC TCC TTC AGC CGC CCT GAC CGC CCA GCC TCT GTG CAT TGG TTC CGG AAC CGG GGC CAG GGC CGA GTC CCT GTC CGG GAG TCC CCC CAT CAC CAC TTA GCG GAA AGC TTC CTC TTC CTG CCC CAA GTC AGC CCC ATG GAC TCT GGG CCC TGG GGC TGC ATC CTC ACC TAC AGA GAT GGC TTC AAC GTC TCC ATC ATG TAT AAC CTC ACT GTT CTG GGT CTG GAG CCCCAG GCC TCG ATG ACT GCC AGC CCC CCA GGA TCT CTC AGA GCC TCC GAC TGG GTC ATT TTG AAC TGC TCC TTC AGC CGC CCT GAC CGC CCA GCC TCT GTG CAT TGG TTC CGG AAC CGG GGC CAG GGC CGA GTC CCC GTC CGG GAG T CAT CAC CAC TTA GCG GAA AGC TTC CTC TTC CTG CCC CAA GTC AGC CCC ATG GAC TCT GGG CCC TGG GGC TGC ATC CTC ACC TAC AGA GAT GGC TTC AAC GTC TCC ATC ATG TAT AAC CTC ACT GTT CTG GGT CTG GAG CCC Ig-like C2 type-1 domain (291bp)Ig-like C2 type-1 domain (291bp)
D3D3 CCA ACT CCC TTG ACA GTG TAC GCT GGA GCA GGT TCC AGG GTG GGG CTG CCC TGC CGC CTG CCT GCT GGT GTG GGG ACC CGG TCT TTC CTC ACT GCC AAG TGG ACT CCT CCT GGG GGA GGC CCT GAC CTC CTG GTG ACT GGA GAC AAT GGC GAC TTT ACC CTT CGA CTA GAG GAT GTG AGC CAG GCC CAG GCT GGG ACC TAC ACC TGC CAT ATC CAT CTG CAG GAA CAG CAG CTC AAT GCC ACT GTC ACACCA ACT CCC TTG ACA GTG TAC GCT GGA GCA GGT TCC AGG GTG GGG CTG CCC TGC CGC CTG CCT GCT GGT GTG GGG ACC CGG TCT TTC CTC ACT GCC AAG TGG ACT CCT CCT GGG GGA GGC CCT GAC CTC CTG GTG ACT GGA GAC AAT GGC GGA GAC GAC TTT ACC CTT CGA CTA GAG GAT GTG AGC CAG GCC CAG GCT GGG ACC TAC ACC TGC CAT ATC CAT CTG CAG GAA CAG CAG CTC AAT GCC ACT GTC ACA Ig-like C2 type-2 domain (249bp)Ig-like C2 type-2 domain (249bp)
D4D4 TTG GCA ATC ATC ACA GTG ACT CCC AAA TCC TTT GGG TCA CCT GGA TCC CTG GGG AAG CTG CTT TGT GAG GTG ACT CCA GTA TCT GGA CAA GAA CGC TTT GTG TGG AGC TCT CTG GAC ACC CCA TCC CAG AGG AGT TTC TCA GGA CCT TGG CTG GAG GCA CAG GAG GCC CAG CTC CTT TCC CAG CCT TGG CAA TGC CAG CTG TAC CAG GGG GAG AGG CTT CTT GGA GCA GCA GTG TAC TTC ACA GAG CTG TCT AGC CCA GGT GCC CAA CGC TCT GGG AGA GCC CCA GGT GCC CTC CCA GCA GGC CAC CTCTTG GCA ATC ATC ACA GTG ACT CCC AAA TCC TTT GGG TCA CCT GGA TCC CTG GGG AAG CTG CTT TGT GAG GTG ACT CCA GTA TCT GGA CAA GAA CGC TTT GTG TGG AGC TCT CTG GAC ACC CCA TCC CAG AGG AGT TTC TCA GGA CGG CTG GAG GCA CAG GAG GCC CAG CTC CTT TCC CAG CCT TGG CAA TGC CAG CTG TAC CAG GGG GAG AGG CTT CTT GGA GCA GCA GTG TAC TTC ACA GAG CTG TCT AGC CCA GGT GCC CAA CGC TCT GGG AGA GCC CCA GGT GCC CTC CTC GGC CAC CTC Ig-like C2 type-3 domain (309bp)Ig-like C2 type-3 domain (309bp)
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, the remaining sequences except for the stop codon portion of the nucleotide sequence constituting the domains were joined together to perform gene synthesis. The accuracy of gene synthesis was confirmed through sequencing after a protein expression plasmid was prepared. 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 제조Protein expression plasmid preparation
HLA class II(HLA-DP, HLA-DQ, HLA-DR)를 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. Recognizing HLA class II (HLA-DP, HLA-DQ, HLA-DR) and expressing a recombinant protein in which the human immunoglobulin G 1 heavy chain constant region and the signaling protein domain are fused to cytotoxic T cells and natural killer cells To do this, the gene was cloned into pCDH-CMV-MCS-EF1-copGFP (System Biosciences), a lentivirus-derived expression vector, or pLNCX2 (Addgene), a retrovirus-derived expression vector.
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, for cloning into the pCDH-CMV-MCS-EF1-copGFP vector, a primer that creates a cleavage site sequence of restriction enzyme XbaI at the 5' end was synthesized, and a restriction enzyme cleavage site was created by polymerase chain reaction, and also restricted at the 3' end. Restriction enzyme cleavage site was created by polymerase chain reaction by synthesizing a primer making the cleavage site sequence of the enzyme NotI. After that, the 5' end of the gene having a restriction enzyme cleavage site was treated with XbaI, and the 3' end was treated with NotI due to the polymerase chain reaction. Then, the multicloning site of the expression vector was treated with XbaI and NotI to allow the gene to be inserted. After mixing the restriction enzyme-treated gene with the expression vector, it was 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 BglII과 NotI을 처리하여 유전자가 삽입될 수 있게 만들었다. 제한효소가 처리된 유전자와 발현 벡터를 섞어 준 다음, 결합 효소(ligase)를 처리하여 연결하였다. Next, for cloning into the pLNCX2 vector, a primer that creates a cleavage site sequence of restriction enzyme Bgl II at the 5' end was synthesized to create a restriction enzyme cleavage site by polymerase chain reaction, and the cleavage site sequence of restriction enzyme NotI was also synthesized at the 3' end. A restriction enzyme cleavage site was created by synthesizing the primers to be made and by polymerase chain reaction. After that, the 5' end of the gene having a restriction enzyme cleavage site was treated with BglII, and the 3' end was treated with NotI due to the polymerase chain reaction. And the multicloning site of the expression vector was treated with BglII and NotI to allow the gene to be inserted. After mixing the restriction enzyme-treated gene with the expression vector, it was ligated by treatment with a ligase.
그 결과, HLA class II(HLA-DP, HLA-DQ, HLA-DR)를 인식하고, 인간 이뮤노글로불린 G1 중쇄 불변 영역과 신호전달 단백질 도메인을 융합시킨 단백질을 발현하는 재조합 벡터 LAG-3.CAR-T.pCDH-CMV-MCS-EF1-copGFP 및 LAG-3.CAR-NK.pLNCX2를 완성하였다(도 2a와 2b 참조).As a result, a recombinant vector LAG-3 that recognizes HLA class II (HLA-DP, HLA-DQ, HLA-DR) and expresses a protein obtained by fusion of a human immunoglobulin G 1 heavy chain constant region and a signaling protein domain. CAR-T.pCDH-CMV-MCS-EF1-copGFP and LAG-3.CAR-NK.pLNCX2 were completed (see FIGS. 2A and 2B ).
실시예 3. HLA class II를 발현하는 사람 세포주 screeningExample 3. Screening of human cell lines expressing HLA class II
HLA class II를 세포 표면에 발현하는 사람 세포주를 screening하기 위해서, 급성전골수성백혈병(acute promyelocytic leukemia) 유래 세포주인 HL-60 (ATCC) 및 T 세포 림프종(T cell lymphoma)유래 세포주인 Jurkat (ATCC) 세포주에서 HLA class II의 발현을 확인하였다. 발현 확인을 위해 형광 단백질이 부착된 HLA class II와 결합이 가능한 항체(biolegend)를 상기 세포주들에 처리한 후, 유세포 분석(fluorescence-activated cell sorting)을 이용하였다. 분석결과, HL-60 세포주에서 HLA class II를 발현하는 것을 확인하였으며, Jurkat 세포주에서는 HLA class II를 발현하지 못하는 것으로 확인하였다(도 4 참조). In order to screen human cell lines expressing HLA class II on the cell surface, acute promyelocytic leukemia-derived cell line HL-60 (ATCC) and T cell lymphoma-derived cell line Jurkat (ATCC) Expression of HLA class II was confirmed in the cell line. To confirm expression, the cell lines were treated with an antibody (biolegend) capable of binding to HLA class II to which a fluorescent protein was attached, and then flow cytometry (fluorescence-activated cell sorting) was used. As a result of the analysis, it was confirmed that HLA class II was expressed in the HL-60 cell line, and it was confirmed that the Jurkat cell line did not express HLA class II (see FIG. 4 ).
상기 결과를 근거로 두 세포 중 HLA class II를 발현하는 HL-60 세포주를 타겟 세포로 이용하였으며 HLA class II를 발현하지 않는 Jurkat 세포를 음성 대조군으로 사용하였다.Based on the above results, the HL-60 cell line expressing HLA class II among the two cells was used as a target cell, and Jurkat cells not expressing HLA class II were used as a negative control.
실시예Example 4. 4. 키메릭chimeric 항원 수용체와 antigen receptors and HLAHLA class II를 발현하는 사람 세포주의 친화도 측정 Affinity measurement of human cell lines expressing class II
제작한 키메릭 항원 수용체가 HLA class II를 발현하는 사람 세포주와 친화도가 있는지 확인하기 위하여, LAG-3의 세포외 도메인(1번부터 4번) 각각을 인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)과 연결시켜, 키메릭 항원 수용체 하나당 2개의 LAG-3의 세포외 도메인(1번부터 4번)이 항원을 인식하게 제작한 키메릭 항원 수용체를 수용성으로 만들어 Chinese hamster ovary(CHO) 세포에서 발현시킨 후, protein A column을 이용하여 친화크로마토그래피(affinity chromatography) (GE healthcare)로 정제하였다(도 5a 참조). In order to confirm whether the constructed chimeric antigen receptor has affinity with a human cell line expressing HLA class II, each of the extracellular domains (No. IgG 1 heavy chain constant region), two extracellular domains of LAG-3 (No. 1 to No. 4) per one chimeric antigen receptor made the chimeric antigen receptor water soluble to recognize the antigen, Chinese hamster ovary After expression in (CHO) cells, the protein was purified by affinity chromatography (GE healthcare) using a protein A column (see FIG. 5a ).
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 5b 참조). In addition, the purified water-soluble chimeric antigen receptor was confirmed by Western blotting using an antibody recognizing the constant region of human IgG heavy chain (anti-human IgG heavy chain constant region antibody, anti-human IgG Fc region antibody, abcam) (Fig. 5b).
그 후, 상기 정제한 수용성 키메릭 항원 수용체를 HLA class II를 발현하는 세포주(HL-60)와 HLA class II를 발현하지 않는 세포주(Jurkat)에 처리하여 수용성 키메릭 항원 수용체가 해당 세포와 결합이 가능한지 유세포 분석을 통하여 확인한 결과를 도 5c에 나타내었다.Thereafter, the purified water-soluble chimeric antigen receptor was treated with a cell line expressing HLA class II (HL-60) and a cell line not expressing HLA class II (Jurkat) to prevent the water-soluble chimeric antigen receptor from binding to the cell. The results confirmed through flow cytometry analysis are shown in FIG. 5c.
도 5c에 나타난 바와 같이, MHC class II를 발현하는 HL-60 세포는 수용성 키메릭 항원 수용체와 결합하였으나, MHC class II를 발현하지 않는 Jurkat 세포에서는 수용성 키메릭 항원 수용체와 결합하지 않는 것을 확인하였다. As shown in FIG. 5c , it was confirmed that HL-60 cells expressing MHC class II bound to the water-soluble chimeric antigen receptor, but Jurkat cells not expressing MHC class II did not bind to the water-soluble chimeric antigen receptor.
상기 결과에서 확인된 바와 같이, 제작한 키메릭 항원 수용체가 HLA class II를 발현하는 사람 세포주와 친화도가 높음을 확인하였고, 이는 HL-60 세포와 Jurkat 세포를 제작한 면역세포 치료제의 MHC class II 단백질의 특이적 세포독성 검증을 위해 사용할 수 있다는 것을 의미한다.As confirmed from the above results, it was confirmed that the prepared chimeric antigen receptor had a high affinity with the human cell line expressing HLA class II, which was the MHC class II of the immune cell therapy prepared with HL-60 cells and Jurkat cells. This means that it can be used for specific cytotoxicity verification of proteins.
실시예 5. 세포독성 T 세포 분리 및 활성화Example 5. 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.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 (Medilab Korea), magnetic-activated cell sorting was used to isolate cytotoxic T cells. Peripheral blood mononuclear cells were 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 were re-conjugated with magnetic microbeads (anti- biotin microbead) (Miltenyi Biotec). The microbeads, antibodies and cells attached thereto were passed through a magnetic seperation column (Miltenyi Biotec) to obtain cytotoxic T cells not labeled with the antibody. In order to confirm the purity of the isolated cytotoxic T cells, flow cytometry using CD8 and CD3ε, which are cell surface factors of cytotoxic T cells, was performed, and as a result, the purity was greater than 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. For activation of isolated cytotoxic T cells, 1Х10 6 pieces/ml of magnetic beads coated with human CD3 and CD28 antibodies (Thermo Fisher Scientific), and 10% calf serum supplemented with 100 U/μl recombinant human IL-2 Cells were reconstituted in RPMI (Welgene) at a density of 1.5Х10 6 pieces/ml and cultured for 24 hours after inoculation in a 24-well cell culture dish.
실시예 6. 키메릭 항원 수용체 발현 세포독성 T 세포 제작Example 6. 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 LAG-3.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. 원심분리를 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(LAG-3 CAR-T cell)를 공벡터로 제작한 바이러스를 형질도입한 세포독성 T 세포(Mock/T cell)의 형질도입 비율과 비교한 결과를 도 6a에 나타내었다.First, 293FT cells were inoculated to become 2.5Х10 6 cells in a 100π cell culture dish, and then 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 LAG-3.CAR-T.pCDH-CMV-MCS-EF1-copGFP vector DNA is mixed with 10 µg of psPAX2 (Addgene) and 3 µg pMD2.G (Addgene) vector was crystallized using calcium phosphate and Hepes-buffered solution, and then introduced into 293FT cells. Thereafter, the culture supernatant containing the lentivirus was collected at intervals of 24 hours after 48 hours of the 293FT cells introduced with the expression vector. The collected supernatant was centrifuged at 21,000 rpm in an ultra-high speed centrifuge for 2 hours to concentrate the virus. After the concentrated virus was mixed with 4 μg/ml of polybrene and added to the culture of activated cytotoxic T cells, the transduction was performed by centrifugation at 1,800 g for 75 minutes using a centrifuge. After centrifugation, the cytotoxic T cells were further cultured for 4 hours and then replaced with an RPMI culture medium containing 10% calf serum. After 48 hours, some of the transduced cytotoxic T cells were used to measure the transduction efficiency. Transduction efficiency was measured by using flow cytometry to measure the expression level of GFP inside the cells, and cytotoxic T cells (Mock) transduced with a virus prepared by using the transduced cytotoxic T cells (LAG-3 CAR-T cells) as an empty vector. /T cells) and the results compared with the transduction ratio is shown in Figure 6a.
실시예 7. 키메릭 항원 수용체 발현 자연살해세포 제작Example 7. Preparation of chimeric antigen receptor-expressing natural killer cells
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(LAG-3.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. 채취한 레트로바이러스 함유 상층액은 초고속 원심분리기를 이용하여 21,000rpm으로 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. First, 3Х10 6 293GPG cells were dissolved in 10 ml of 10 ml of 10% calf serum-containing DMEM culture medium and inoculated in a 100π cell culture dish and then cultured for 24 hours. 20 μg of the previously prepared recombinant vector (LAG-3.CAR-NK.pLNCX2 vector) was crystallized using calcium phosphate and Hepes-buffered solution, and then added to the culture medium of the previously cultured 293GPG cells. Thereafter, the culture medium was changed over 72 hours at 24 hour intervals, and the culture supernatant of 293GPG cells containing retrovirus was collected and stored. 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 into a 24 well cell culture dish at a density of 5Х10 5 /ml. Then, a mixed solution of 8 ug/ml of polybrene added to the concentrated retrovirus was added to the culture medium of NK92MI cells in culture and cultured for 24 hours. After culturing, the culture medium was replaced with a new culture medium. For the selection of the cells into which the gene was introduced, from 24 hours after the culture medium replacement, the Myelocult H5100 culture medium supplemented with neomycin antibiotic (600 μg/ml) was used to culture the retrovirus-treated NK92MI cells. After 14 days of neomycin selection, NK92MI cells were transferred to fresh culture medium and proliferated for 1 week.
증식 후 유세포 분석을 이용해 NK92MI세포의 LAG-3 발현량을 측정하였으며 LAG-3.CAR-NK.pLNCX2를 도입한 NK92MI 세포(LAG-3 CAR-NKcell)와 공벡터인 pLNCX2를 도입한 NK92MI 세포(Mock CAR-NK cell)의 LAG-3 발현량을 항 인간 LAG-3 항체(anti-human LAG-3 antibody, Novus biologicals)를 이용한 유세포분석기를 이용하여 비교한 결과를 도 6b에 나타내었다.After proliferation, the LAG-3 expression level of NK92MI cells was measured using flow cytometry. NK92MI cells introduced with LAG-3.CAR-NK.pLNCX2 (LAG-3 CAR-NKcell) and NK92MI cells introduced with the empty vector pLNCX2 ( The results of comparing the LAG-3 expression level of Mock CAR-NK cells using a flow cytometer using an anti-human LAG-3 antibody (Novus biologicals) are shown in FIG. 6b .
실시예 8. CAR-T 세포의 MHC class II+ cell 특이적 세포독성 검증Example 8. MHC class II+ cell-specific cytotoxicity verification of CAR-T cells
제작한 키메릭 항원 수용체 발현 세포독성 T 세포가 MHC class II 세포를 특이적으로 인지하여 독성을 나타내는지 확인하기 위해, 실시예 3에서 선정한 타겟 세포인 HL-60 세포(HLA class II positive cell)와 Jurkat 세포(HLA class II negative cell)를 키메릭 항원 수용체 발현 세포독성 T 세포(LAG-3 CAR-T cell) 혹은 공벡터를 도입한 세포독성 T 세포(Mock T cell)와 6시간 동안 공배양하였다. 공배양 후 상층액에 존재하는 젖산 탈수소 효소(lactate dehydrogenase)의 양으로 세포독성 T 세포의 표적세포에 대한 세포독성 정도를 측정하는 비방사성 세포독성 분석법(non-radioactive cytotoxicity assay)을 이용하였다. In order to confirm whether the prepared chimeric antigen receptor-expressing cytotoxic T cells specifically recognize MHC class II cells and show toxicity, the target cells selected in Example 3, HL-60 cells (HLA class II positive cells) and Jurkat cells (HLA class II negative cells) were co-cultured with chimeric antigen receptor-expressing cytotoxic T cells (LAG-3 CAR-T cells) or cytotoxic T cells (Mock T cells) introduced with an empty vector for 6 hours. . A non-radioactive cytotoxicity assay was used to measure the degree of cytotoxicity of cytotoxic T cells to target cells by the amount of lactate dehydrogenase present in the supernatant after co-culture.
먼저, 96 well 세포 배양 접시의 well에 세포독성 T 세포(1x105 cell)와 표적 세포(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 cells) and target cells (1x10 4 cells) into the wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, and inoculate so that the volume per well becomes 100μl Centrifuge for 4 minutes at 250 g using a centrifuge to close the intercellular space. After culturing for 6 hours, 50 μl of the supernatant from each well is removed, transferred to a transparent 96-well dish for absorbance measurement, and treated with an analysis solution and 1M hydrochloric acid solution to proceed and stop the enzyme reaction. After stopping the enzymatic reaction, the absorbance in the 490 nm wavelength band was measured and numerically converted using a fluorescence/luminescence/absorption meter (multi-detection plate reader) to quantify the degree of cytotoxicity of cytotoxic T cells in each well.
그 결과 도 7a에서 나타낸 바와 같이, 제작한 키메릭 항원 수용체 발현 세포독성 T 세포(LAG-3 CAR-T cell)는 Jurkat 세포(HLA class II negative cell)에 대해서 0.5% 정도의 세포독성을 보인 반면, HL-60 세포(HLA class II positive cell)는 22% 이상의 높은 세포독성을 보임을 확인하였다(p<0.01). 반면, 대조군인 공벡터를 도입한 세포독성 T 세포의 경우 Jurkat 세포(HLA class II negative cell) 또는 HL-60 세포(HLA class II positive cell)에 대하여, 세포독성을 갖지 않는 것으로 확인되었다. As a result, as shown in Figure 7a, the prepared chimeric antigen receptor-expressing cytotoxic T cells (LAG-3 CAR-T cells) showed about 0.5% cytotoxicity to Jurkat cells (HLA class II negative cells), whereas , It was confirmed that HL-60 cells (HLA class II positive cell) showed high cytotoxicity of 22% or more (p<0.01). On the other hand, in the case of cytotoxic T cells introduced with the empty vector as a control, it was confirmed not to have cytotoxicity against Jurkat cells (HLA class II negative cells) or HL-60 cells (HLA class II positive cells).
상기 결과를 통해 LAG-3 세포외 도메인을 포함하고 있는 키메릭 항원 인지 수용체 발현 세포독성 T 세포가 MHC class II 단백질에 특이적인 세포독성을 보임으로써, 상기 CAR-T T세포를 이용하여 관련된 암 세포 치료가 가능함을 확인할 수 있었다.Through the above results, the chimeric antigen recognition receptor-expressing cytotoxic T cells containing the LAG-3 extracellular domain showed cytotoxicity specific to the MHC class II protein, and thus the related cancer cells using the CAR-T T cells. It was confirmed that treatment was possible.
실시예 9. CAR-NK 세포의 MHC class II+ cell 특이적 세포독성 검증Example 9. MHC class II+ cell-specific cytotoxicity verification of CAR-NK cells
제작한 키메릭 항원 수용체 발현 NK 세포가 MHC class II 세포를 특이적으로 인지하여 독성을 나타내는지 확인하기 위해, 실시예 3에서 선정한 타겟 세포인 HL-60 세포(HLA class II positive cell)와 Jurkat 세포(HLA class II negative cell)를 키메릭 항원 수용체 발현 NK 세포(LAG-3 CAR-NK cell) 혹은 공벡터를 도입한 NK 세포(Mock NK cell)와 공배양하였다. In order to confirm whether the prepared chimeric antigen receptor-expressing NK cells specifically recognize MHC class II cells and show toxicity, HL-60 cells (HLA class II positive cells) and Jurkat cells selected in Example 3 (HLA class II negative cells) were co-cultured with chimeric antigen receptor-expressing NK cells (LAG-3 CAR-NK cells) or NK cells introduced with an empty vector (Mock NK cells).
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. 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 after co-culture.
먼저, 96 well 세포 배양 접시의 well에 자연살해세포(1x105 cell)와 표적 세포(1x104 cell)를 각각 넣어 effector: target ratio를 10:1로 맞추고, well당 부피는 100μl가 되도록 접종 후 원심분리기를 이용해 250g의 조건에서 4분 동안 원심분리하여 세포 간 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 cells) and target cells (1x10 4 cells) into the wells of a 96-well cell culture dish, set the effector: target ratio to 10:1, and inoculate it so that the volume per well becomes 100μl and centrifuge Centrifuge for 4 minutes at 250 g using a separator to close the intercellular space. After culturing for 6 hours, 50 μl of the supernatant from each well is removed and transferred to a transparent 96-well dish for absorbance measurement. After stopping the enzymatic reaction, the absorbance of the 490 nm wavelength band was measured and numerically converted using a fluorescence/luminescence/absorption meter (multi-detection plate reader) to quantify the degree of cytotoxicity of NK cells in each well.
그 결과 도 7b에서 나타낸 바와 같이, 제작한 키메릭 항원 수용체 발현 NK 세포(LAG-3 CAR-NK cell)는 Jurkat 세포(HLA class II negative cell)에 대해 거의 세포독성을 안보인 반면, HL-60 세포(HLA class II positive cell)는 47% 이상의 매우 높은 세포독성을 보임을 확인하였다(p<0.01). 반면, 대조군인 공벡터를 도입한 세포독성 T 세포의 경우 Jurkat 세포(HLA class II negative cell) 또는 HL-60 세포(HLA class II positive cell)에 대하여, 세포독성을 갖지 않는 것으로 확인되었다.As a result, as shown in FIG. 7b, the prepared chimeric antigen receptor-expressing NK cells (LAG-3 CAR-NK cells) showed almost no cytotoxicity to Jurkat cells (HLA class II negative cells), whereas HL-60 It was confirmed that the cells (HLA class II positive cell) showed a very high cytotoxicity of more than 47% (p<0.01). On the other hand, in the case of cytotoxic T cells introduced with the empty vector as a control, it was confirmed not to have cytotoxicity against Jurkat cells (HLA class II negative cells) or HL-60 cells (HLA class II positive cells).
상기 결과를 통해 LAG-3 세포외 도메인을 포함하고 있는 키메릭 항원 인지 수용체 발현 NK 세포가 MHC class II 단백질에 특이적인 세포독성을 보임으로써, 상기 CAR-NK 세포를 이용하여 관련된 암 세포 치료가 가능함을 확인할 수 있었다.Through the above results, the chimeric antigen-recognizing receptor-expressing NK cells containing the LAG-3 extracellular domain showed cytotoxicity specific to the MHC class II protein, so that it is possible to treat the related cancer cells using the CAR-NK cells. was able to confirm
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, with respect to the present invention, the preferred embodiments have been looked at. Those of ordinary skill in the art to which the present invention pertains will understand that the present invention may be implemented in a modified form without departing from the essential characteristics of the present invention. Therefore, the disclosed embodiments are to be considered in an illustrative rather than a restrictive sense. The scope of the present invention is indicated in the claims rather than the foregoing description, and all differences within the scope equivalent thereto should 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, A chimeric antigen receptor (CAR) comprising an intracellular signaling domain comprising:
    상기 항원 결합 도메인은 HLA class II에 특이적으로 결합하는 LAG-3의 세포외 도메인(1번부터 4번)을 포함하는 것인, 키메릭 항원 수용체.The antigen-binding domain is a chimeric antigen receptor comprising an extracellular domain (No. 1 to No. 4) of LAG-3 that specifically binds to HLA class II.
  2. 제1항에 있어서,According to claim 1,
    상기 항원 결합 도메인은 LAG-3의 세포외 도메인(1번부터 4번) 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 characterized in that a pair of extracellular domains (No. 1 to No. 4) of LAG-3 are connected to each human immunoglobulin G 1 heavy chain constant region in the form of a dimer (IgG 1 heavy chain constant region) A chimeric antigen receptor.
  3. 제1항에 있어서,According to claim 1,
    상기 LAG-3의 세포외 도메인(1번부터 4번)은 서열번호 1로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The extracellular domain of the LAG-3 (No. 1 to No. 4) will include the amino acid sequence shown in SEQ ID NO: 1, a chimeric antigen receptor.
  4. 제2항에 있어서,3. The method of claim 2,
    인간 이뮤노글로불린 G1 중쇄 불변 영역(IgG1 heavy chain constant region)은 서열번호 2로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.Human immunoglobulin G 1 heavy chain constant region (IgG 1 heavy chain constant region) is a chimeric antigen receptor comprising the amino acid sequence represented by SEQ ID NO: 2.
  5. 제1항에 있어서,According to claim 1,
    상기 막관통 도메인은 CD8이며, 서열번호 3으로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The transmembrane domain is CD8, the chimeric antigen receptor comprising the amino acid sequence shown in SEQ ID NO: 3.
  6. 제1항에 있어서,The method of 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 a chimeric antigen receptor comprising one consisting of CD28, 4-1BB and CD3-zeta or a combination thereof.
  7. 제6항에 있어서,7. The method of claim 6,
    상기 CD28은 서열번호 4; 4-1BB은 서열번호 5; 및 CD3-zeta은 서열번호 6로 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 shown in SEQ ID NO: 6.
  8. 제1항에 있어서, According to claim 1,
    상기 항원 결합 도메인은 LAG-3 신호 펩타이드를 포함하며, 서열번호 7로 표시되는 아미노산 서열을 포함하는 것인, 키메릭 항원 수용체.The antigen binding domain comprises a LAG-3 signal peptide, and the amino acid sequence shown in SEQ ID NO: 7, the 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,12. The method of claim 11,
    상기 세포는 세포독성 T 세포 또는 NK 세포인 것을 특징으로 하는 세포.The cell, characterized in that the cell is a cytotoxic T cell or NK cell.
  13. 제12항의 세포를 유효성분으로 포함하는 HLA class II를 발현하는 암의 치료용 약학적 조성물.A pharmaceutical composition for the treatment of cancer expressing HLA class II comprising the cell of claim 12 as an active ingredient.
  14. 제13항에 있어서,14. The method of claim 13,
    상기 HLA class II를 발현하는 암은 머리 및 목 편평 세포 암종(Head and neck squamous cell carcinoma), 흉선종(thymoma), 식도 편평 세포 암종(Esophageal squamous cell carcinoma), 흑색종(melanoma), 유방암(breast cancer), 결장암(colorectal cancer), 난소암(ovarian cancer), 전립선암(prostate cancer), 신경아 세포종(neuroblstoma), 신경교종(glioma), 비소세포형 폐암(non-small cell lung cancer), 클래식 호지킨 림프종(classic Hodgkin lymphoma), T 세포 백혈병/림프종(T-cell leukemia/lymphoma), B세포 림프종 (B cell lymphoma), 만성골수성 백혈병(chronic myeloid leukemia), 만성림프성 백혈병(chronic lymphocytic leukemia) , 급성 골수성 백혈병(acute myeloid leukemia) 및 급성 림프구성 백혈병(acute lymphoid leukemia)으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 조성물.The cancer expressing the HLA class II is head and neck squamous cell carcinoma, thymoma, esophageal squamous cell carcinoma, melanoma, breast cancer ), colorectal cancer, ovarian cancer, prostate cancer, neuroblstoma, glioma, non-small cell lung cancer, classic Hodgkin Lymphoma (classic Hodgkin lymphoma), T-cell leukemia/lymphoma, B-cell lymphoma, chronic myeloid leukemia, chronic lymphocytic leukemia, acute A composition selected from the group consisting of acute myeloid leukemia and acute lymphoid leukemia.
  15. 제11항의 세포를 포함하는 세포 치료제.A cell therapeutic agent comprising the cell of claim 11 .
PCT/KR2022/002352 2021-02-26 2022-02-17 Chimeric antigen receptor specifically binding to hla class ii and use thereof WO2022182059A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020210026090A KR102671408B1 (en) 2021-02-26 Chimeric antigen receptor specifically binding to HLA class II and use thereof
KR10-2021-0026090 2021-02-26

Publications (1)

Publication Number Publication Date
WO2022182059A1 true WO2022182059A1 (en) 2022-09-01

Family

ID=83049440

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2022/002352 WO2022182059A1 (en) 2021-02-26 2022-02-17 Chimeric antigen receptor specifically binding to hla class ii and use thereof

Country Status (1)

Country Link
WO (1) WO2022182059A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020032784A1 (en) * 2018-08-10 2020-02-13 주식회사 유틸렉스 Chimeric antigen receptor binding to hla-dr, and car-t cell
KR20200063215A (en) * 2017-10-06 2020-06-04 오슬로 유니버시테시케후스 에이치에프 Chimeric antigen receptor

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200063215A (en) * 2017-10-06 2020-06-04 오슬로 유니버시테시케후스 에이치에프 Chimeric antigen receptor
WO2020032784A1 (en) * 2018-08-10 2020-02-13 주식회사 유틸렉스 Chimeric antigen receptor binding to hla-dr, and car-t cell

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE PROTEIN 23 March 2015 (2015-03-23), ANONYMOUS : "lymphocyte-activation gene 3, isoform CRA_b [Homo sapiens] ", XP055961409, retrieved from NCBI Database accession no. EAW88741 *
DATABASE PROTEIN 3 October 2016 (2016-10-03), ANONYMOUS : "IgG1-CD3z [Cloning vector pCDH-EF1a-GaussiaSP-MCS-myc-IgG1-CD3z-IRES-copGFP] ", XP055961410, retrieved from NCBI Database accession no. AOS59250 *
MACLACHLAN BRUCE J., MASON GEORGINA H., GREENSHIELDS‐WATSON ALEXANDER, TRIEBEL FREDERIC, GALLIMORE AWEN, COLE DAVID K., GODKIN AND: "Molecular characterization of HLA class II binding to the LAG‐3 T cell co‐inhibitory receptor", EUROPEAN JOURNAL OF IMMUNOLOGY, WILEY-VCH, HOBOKEN, USA, vol. 51, no. 2, 1 February 2021 (2021-02-01), Hoboken, USA, pages 331 - 341, XP055961403, ISSN: 0014-2980, DOI: 10.1002/eji.202048753 *

Also Published As

Publication number Publication date
KR20220122831A (en) 2022-09-05

Similar Documents

Publication Publication Date Title
US20220211756A1 (en) T cell which expresses a gamma-delta t cell receptor (tcr) and a chimeric antigen receptor (car)
US11883494B2 (en) Combination of immune effector cells specific for a target antigen and hematopoietic calls that express the target antigen in an altered form
CA2188422C (en) Bifunctional protein, preparation and use
KR20220029757A (en) Chimeric antigen receptors targeting b-cell maturation antigen
WO2021256724A1 (en) Chimeric antigen receptor targeting bcma and use thereof
WO2021066420A1 (en) Chimeric antigen receptor specifically binding to cd138, immune cell expressing same, and anticancer use thereof
CN113195535A (en) Bispecific polypeptides for linking CAR-expressing immune cells to antigen presenting cells and uses thereof
WO2022203226A1 (en) Transformed antigen-specific professional antigen presenting cell including chimeric antigen receptor (car) and use thereof
WO2022182059A1 (en) Chimeric antigen receptor specifically binding to hla class ii and use thereof
WO2023003404A1 (en) Novel chimeric antigen receptor and immune cells expressing same
WO2022215919A1 (en) Chimeric antigen receptor specifically binding to cd47 and use thereof
WO2022220433A1 (en) Chimeric antigen receptor binding specifically to programmed death-ligand 1 (pd-l1) and use thereof
WO2022186682A1 (en) Chimeric antigen receptor specifically binding to rank ligand, and use thereof
WO2023287049A1 (en) Chimeric antigen receptor binding specifically to cd30 and use thereof
WO2023075177A1 (en) Chimeric antigen receptor specifically binding to cadm1 and uses thereof
CN111542547A (en) Chimeric antigen receptor specific for BDCA2 antigen
WO2022239986A1 (en) Chimeric antigen receptor comprising extracellular domain of baff and use thereof
WO2023068584A1 (en) Chimeric antigen receptor specifically binding to cd138 and uses thereof
KR102231285B1 (en) Chimeric antigen receptor specifically binding to VLA-4 and use thereof
KR102231284B1 (en) Chimeric antigen receptor specifically binding to CD38 and use thereof
WO2023113292A1 (en) Transformed antigen-specific professional antigen-presenting cell comprising chimeric antigen receptor (car) and use thereof
WO2022211376A1 (en) Transformed antigen-specific professional antigen presenting cells comprising chimeric antigen receptor (car) and use thereof
WO2023113293A1 (en) Transformed antigen-specific professional antigen-presenting cell containing chimeric antigen receptor (car) and use thereof
WO2022234976A1 (en) Transformed antigen-specific professional antigen-presenting cell containing chimeric antigen receptor (car) and use thereof
WO2022203227A1 (en) Transformed antigen-specific professional antigen-presenting cell 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: 22759977

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22759977

Country of ref document: EP

Kind code of ref document: A1