WO2024101962A1 - Cellules génétiquement modifiées et leur utilisation - Google Patents

Cellules génétiquement modifiées et leur utilisation Download PDF

Info

Publication number
WO2024101962A1
WO2024101962A1 PCT/KR2023/018102 KR2023018102W WO2024101962A1 WO 2024101962 A1 WO2024101962 A1 WO 2024101962A1 KR 2023018102 W KR2023018102 W KR 2023018102W WO 2024101962 A1 WO2024101962 A1 WO 2024101962A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
present
cell
amino acid
acid sequence
Prior art date
Application number
PCT/KR2023/018102
Other languages
English (en)
Korean (ko)
Inventor
정수영
서민구
오민석
장미희
Original Assignee
주식회사 유씨아이테라퓨틱스
한국과학기술연구원
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 KR1020220150906A external-priority patent/KR20240081499A/ko
Application filed by 주식회사 유씨아이테라퓨틱스, 한국과학기술연구원 filed Critical 주식회사 유씨아이테라퓨틱스
Publication of WO2024101962A1 publication Critical patent/WO2024101962A1/fr

Links

Images

Definitions

  • the present invention relates to genetically engineered immune effector cells and uses thereof to increase the therapeutic effect for diseases such as cancer by immunotherapy.
  • Cellular immunotherapy is a very promising treatment method for cancer treatment.
  • most immunotherapeutic approaches have the following limitations in their therapeutic effectiveness against most malignant tumors, including solid tumors: (1) Reduced expression of tumor antigens on the surface of tumor cells (which reduces detection of antigens by the immune system) Sikkim); (2) expression of ligands for inhibitory receptors such as PD1, NKG2A, and TIGIT; (3) upregulation of cellular checkpoints, such as CISH, leading to immune cell deactivation; and (4) induction of a microscopic environment that releases substances such as transforming growth factor- ⁇ (TGF- ⁇ ) and adenosine, which suppress immune responses and promote tumor cell proliferation and survival. Therefore, there is a need for an improved method of cellular immunotherapy that can solve at least one of the above-mentioned challenges.
  • TGF- ⁇ transforming growth factor- ⁇
  • TGF- ⁇ signaling plays an important role in cancer progression.
  • Most cancer cells inactivate epithelial antiproliferative responses and benefit from increased TGF- ⁇ expression and autocrine TGF- ⁇ signaling through effects on gene expression, release of immunosuppressive cytokines, and epithelial plasticity.
  • TGF- ⁇ plays a role in increasing invasion and metastasis of cancer cells, stem cell properties, and drug resistance.
  • TGF- ⁇ released from cancer cells, stromal fibroblasts and other cells in the tumor microenvironment further promotes cancer progression by shaping the structure of the tumor and suppressing the anti-tumor activity of immune cells, thus creating an immunosuppressive environment and anti-cancer. Prevents or attenuates the effectiveness of immunotherapy. Therefore, inhibition of TGF- ⁇ signaling is considered a prerequisite and key means to improve the efficacy of current and future immunotherapies, including for tumors containing cancer cells that are unresponsive to TGF- ⁇ .
  • MSLN Mesothelin
  • MSLN-targeted immunotherapies reported to date support a favorable safety profile.
  • MSLN includes at least oesophageal cancer, breast cancer, gastric cancer, cholangiocarcinoma, pancreatic cancer, colon cancer, lung cancer, and thymic carcinoma.
  • CAR targets in many common solid tumors, such as mesothelioma, ovarian cancer, and endometrial cancer [Morello, A. et al. (2016) Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Disco. 6(2); 133-46].
  • One object of the present invention is to provide a peptide that can inhibit the transforming growth factor- ⁇ (TGF- ⁇ ) signaling pathway along with a chimeric antigen receptor (CAR) targeting mesothelin.
  • the aim is to provide cells that have been genetically engineered to express.
  • Another object of the present invention relates to a cell therapeutic agent comprising the genetically engineered cells.
  • Another object of the present invention relates to pharmaceutical compositions for various uses containing the genetically engineered cells.
  • a chimeric antigen receptor (CAR) targeting mesothelin MSLN
  • cells genetically engineered to express a peptide or fragment thereof capable of inhibiting the transforming growth factor- ⁇ (TGF- ⁇ ) signaling pathway a chimeric antigen receptor (CAR) targeting mesothelin (MSLN)
  • TGF- ⁇ transforming growth factor- ⁇
  • “recombinant” or “manipulated” in relation to a peptide means having an amino acid sequence that has been altered as a result of the application of genetic engineering techniques to the nucleic acid encoding the peptide and to the cell or organism expressing the peptide.
  • the terms “recombinant” or “engineered” mean having a nucleic acid sequence that has been altered as a result of the application of genetic engineering techniques. Genetic engineering technologies include PCR and DNA cloning technologies; Transfection, transduction, transformation and other gene transfer techniques; homologous recombination; site-specific mutation; and gene fusions.
  • the term “genetically engineered” or “genetically engineered” refers to a cell or organism, or an ancestor thereof, whose genomic DNA sequence has been intentionally modified by recombinant technology.
  • the “genetic manipulation” includes “gene transplantation.”
  • methods for genetically manipulating the expression of the peptide or its fragment in the cell include biological methods such as vectors, specific receptors, or cell fusion methods, microinjection methods, electroporation methods, gene guns, or ultrasonic genes.
  • biological methods such as vectors, specific receptors, or cell fusion methods, microinjection methods, electroporation methods, gene guns, or ultrasonic genes.
  • a method of introducing the gene encoding the peptide or its fragment through a physical method such as the introduction method, or a chemical method such as the calcium phosphate coprecipitation method, liposome method, lipofection method, DEAE dextran method, or alkali metal method. can do.
  • the chimeric antigen receptor; and peptides or fragments thereof; Foreign genes encoding the proteins can be introduced into the cells by transfecting the immune effector cells with vectors containing genes encoding each.
  • the “chimeric antigen receptor (CAR)” is defined as a cell-surface receptor comprising an extracellular target-binding domain, a transmembrane domain, and an intracellular signaling domain, all of which are formed on a single protein. They exist together in combinations that are not found naturally. This particularly includes receptors in which the extracellular and intracellular signaling domains are not naturally found together on a single receptor protein.
  • Mesothelin is a protein also called MSLN, and is a 40 kDa protein secreted by mesothelial cells.
  • the protein was first identified by reaction with the monoclonal antibody K1, and through continued research, the mesothelin gene was linked to a glycophosphatidylinositol linkage and a 31-kDa shed fragment, megakaryocyte-potentiating factor (MPF). ) is known to encode a precursor protein that is processed to produce mesothelin, which is attached to the cell membrane. It has been suggested that mesothelin may be involved in cell adhesion, etc., but its biological function has not yet been clearly identified.
  • the mesothelin may be composed of the amino acid sequence represented by SEQ ID NO: 1, but is not limited thereto.
  • the chimeric antigen receptor includes a mesothelin binding domain, and may further include one or more selected from the group consisting of a hinge domain, a signal peptide domain, a transmembrane domain, and one or more signaling domains. .
  • the chimeric antigen receptor of the present invention includes a mesothelin (MSLN) binding domain.
  • MSLN mesothelin
  • the mesothelin (MSLN) binding domain provided in the present invention can bind to mesothelin with higher affinity than the conventionally known anti-MSLN binding domain.
  • the binding domain may be an antibody or a fragment thereof.
  • the “antibody” refers to an immunoglobulin molecule that specifically binds to an antigen.
  • the antibody may be a natural or recombinant intact immunoglobulin, or may be an immune-reactive portion of an intact immunoglobulin.
  • Antibodies are generally tetramers of immunoglobulin molecules.
  • the antibody is a tetrameric glycosylated protein composed of two light (L) chains of about 25 kDa each and two heavy (H) chains of about 50 kDa each. Two types of light chains, referred to as lambda and kappa, can be found in antibodies.
  • immunoglobulins can be classified into five main classes A, D, E, G and M, several of which are subclasses (isotypes), such as IgG1, IgG2 , can be further subdivided into IgG3, IgG4, IgA1 and IgA2.
  • Each light chain typically contains an N-terminal variable (V) domain (VL) and constant (C) domain (CL).
  • Each heavy chain typically contains an N-terminal V domain (VH), three or four C domains (CH1-3), and a hinge region. The CH domain located closest to VH is named CH1.
  • the VH and VL domains are composed of four regions of relatively conserved sequences, referred to as framework regions, which form the scaffold for three regions of hypervariable sequences (complementarity determining regions, CDRs) (FR1). , FR2, FR3 and FR4).
  • CDRs contain most of the residues responsible for the specific interaction of the antibody with the antigen.
  • the CDRs are referred to as CDR1, CDR2 and CDR3. That is, the CDR elements on the heavy chain are referred to as CDRH1, CDRH2, and CDRH3, while the CDR elements on the light chain are referred to as CDRL1, CDRL2, and CDRL3.
  • CDRs are typically described in Sequences of Proteins of Immunological Interest, US Department of Health and Human Services (1991), eds. See Kabat CDR, as described in Kabat et al.
  • Another standard for specifying the antigen binding site is by reference to the hypervariable loop described by Chothia. For example, Chothia, D. et al. (1992) J. Mol. Biol. 227:799-817; and Tomlinson et al. (1995) EMBO J. 14:4628-4638.
  • Another standard is the AbM definition used in Oxford Molecular's AbM antibody modeling software. In general, for example, Protein Sequence and Structure Analysis of Antibody Variable Domains.
  • the antibody may exist in various forms, including but not limited to polyclonal antibodies, monoclonal antibodies, Fv, Fab, F(ab)2, etc., and single chain antibodies and humanized antibodies (document.
  • antibodies against the antigen can be obtained from immunized transgenic mice using conventional hybridoma techniques.
  • Human immunoglobulin transgenes carried by transgenic mice rearrange during B cell differentiation and subsequently undergo class switching and somatic mutation. Accordingly, using this technique, it is possible to prepare therapeutically useful IgG, IgA, IgM and IgE antibodies, such as, but not limited to, IgGl (gamma 1) and IgG3. Details of these techniques for preparing human antibodies and human monoclonal antibodies and the protocols for preparing such antibodies can be found, for example, in PCT Publication Nos.
  • “Humanized” antibodies retain similar antigenic specificity as the original antibody, i.e., the ability to bind, for example, MSLN in the present invention.
  • the “antibody fragment” or “antigen-binding fragment” is shorter than the full-length antibody, but includes at least a partial variable region (e.g., one or more CDRs and/or one or more antigen-binding sites) that binds to the antigen of the antibody. Therefore, it refers to any portion of a full-length antibody that retains the binding specificity and at least partial specific binding ability of the full-length antibody. Accordingly, antigen-binding fragment refers to an antibody fragment that contains an antigen-binding portion that binds the same antigen as the antibody from which the antibody fragment was derived.
  • Antibody fragments include antibody derivatives produced by enzymatic treatment of a full-length antibody, and derivatives produced synthetically, such as those produced recombinantly. Antibodies include antibody fragments. Antibody fragments include, but are not limited to, single chain Fv (scFv), Fab, Fab', F(ab')2, Fv, dsFv, double-antibody, Fd and Fd' fragments, and other fragments, including modified fragments. It is not limited (see, e.g., Methods in Molecular Biology, Vol 207: Recombinant Antibodies for Cancer Therapy Methods and Protocols (2003); Chapter 1; p3-25, Kipriyanov).
  • Fragments may comprise multiple chains linked together, for example, through disulfide bonds and/or through peptide linkers.
  • Antibody fragments generally contain at least or about 50 amino acids, and typically contain at least or about 200 amino acids.
  • the antigen-binding fragment is an antibody (e.g., through replacement of a corresponding region) to obtain an antibody that immunospecifically binds to the antigen (i.e., exhibits a Ka of at least or at least about 107-108 M-1).
  • a “functional fragment” or “anti-MSLN antibody analog” is a fragment or analog that can prevent or substantially reduce the ability of a receptor to bind a ligand or initiate signal transduction.
  • functional fragment generally has the same meaning as "antibody fragment” and, in the case of an antibody, is a fragment capable of preventing or substantially reducing the ability of a receptor to bind a ligand or initiate signal transduction, such as Fv. , Fab, and F(ab')2.
  • the “Fv” fragment consists of a dimer (VH-VL dimer) formed by the variable domains of one heavy chain and the variable domains of one light chain by non-covalent association.
  • VH-VL dimer dimer
  • the three CDRs of each variable domain interact to determine the target-binding site on the surface of the VH-VL dimer identical to the intact antibody.
  • the six CDRs together confer the target-binding specificity of the intact antibody.
  • a single variable domain or half of an Fv containing only three target-specific CDRs
  • the “single chain Fv (scFv)” is a single chain antibody fragment having the variable regions of the heavy and light chains of the antibody linked together.
  • scFv single chain Fv
  • the scFv is desirable because it can be genetically engineered to be expressed as part of a single chain together with other components that make up the chimeric antigen receptor.
  • the antigen binding domain is typically included as part of the extracellular portion of the chimeric antigen receptor and is capable of recognizing and binding the targeted antigen, here specifically mesothelin (MSLN).
  • the scFv can be prepared according to methods known in the art (e.g., Bird et al., (1988) Science 242:423-426 and Huston et al., (1988) Proc. Natl. Acad Sci USA 85:5879-5883).
  • ScFv molecules can be made by connecting the VH and VL regions using a flexible polypeptide linker.
  • the scFv molecule includes a linker with optimized length and/or amino acid composition (e.g., Ser-Gly linker). Linker length can greatly affect how the variable region of the scFv folds and interacts. In fact, if short polypeptide linkers (e.g. 5-10 amino acids) are used, intrachain folding is prevented.
  • interchain folding is required to bring the two variable regions into proximity to form a functional epitope binding site.
  • linker orientation and size see, e.g., Hollinger et al. 1993 Proc Natl Acad. Sci. U.S.A. 90:6444-6448, U.S. Patent Application Publication Nos. 2005/0100543, 2005/0175606, 2007/0014794, and PCT Publication Nos. WO2006/020258 and WO2007/024715, the entire contents of each of which are incorporated herein by reference. Included.
  • the mesothelin (MSLN) binding domain may include an scFv capable of specifically recognizing mesothelin.
  • the mesothelin (MSLN) binding domain includes a light chain CDR1 consisting of the amino acid sequence shown in SEQ ID NO: 2; Light chain CDR2 consisting of the amino acid sequence shown in SEQ ID NO: 3; and a light chain variable region including a light chain CDR3 consisting of the amino acid sequence shown in SEQ ID NO: 4.
  • the mesothelin (MSLN) binding domain includes a heavy chain CDR1 consisting of the amino acid sequence shown in SEQ ID NO: 5; Heavy chain CDR2 consisting of the amino acid sequence shown in SEQ ID NO: 6; and a heavy chain variable region including a heavy chain CDR3 consisting of the amino acid sequence shown in SEQ ID NO: 7.
  • variable region In the present invention, the "variable region”, “variable domain”, “V region” or “V domain” is generally located at the amino-terminus of the light or heavy chain, about 120 to 130 amino acids in the heavy chain and about 100 amino acids in the light chain. Refers to the portion of the light or heavy chain of an antibody, ranging in length from 1 to 110 amino acids, and is used in the binding and specificity of each specific antibody for a specific antigen.
  • the variable region of the heavy chain may be referred to as “VH”.
  • VL variable region of the light chain
  • the term “variable” refers to the fact that certain segments of the variable region vary widely in sequence among antibodies. The V region mediates antigen binding and defines the specificity of a particular antibody for a particular antigen.
  • variable region is a frame of about 15 to 30 amino acids separated by shorter regions of greater variability (e.g., extreme variability), referred to as “hypervariable regions,” which are each about 9 to 12 amino acids long. It consists of a less variable (i.e. relatively invariant) stretch called the work region (FR).
  • the variable regions of the heavy and light chains are composed of four FRs that predominantly adopt the ⁇ -sheet configuration, each connected by three hypervariable regions that form loops connecting, and in some cases form part of, the ⁇ -sheet structure. Includes.
  • the hypervariable regions within each chain are closely held by FRs, and hypervariable regions from other chains contribute to the formation of the antigen-binding site of the antibody (see, e.g., Kabat et al., Sequences of Proteins of Immunological Interest (5th ed. 1991)].
  • the constant region is not directly involved in the binding of the antibody to the antigen, but exhibits various effector functions, such as the antibody's participation in antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • Variable regions vary widely in sequence between different antibodies.
  • the variable region is a human variable region.
  • the "heavy chain” refers to a polypeptide chain of about 50 to 70 kDa, wherein the amino-terminal portion comprises a variable region of at least about 120 to 130 amino acids, and the carboxy-terminal portion comprises a constant region.
  • Constant regions are of five distinct types, referred to as alpha ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ), gamma ( ⁇ ), and mu ( ⁇ ), based on the amino acid sequence of the heavy chain constant region, isotype).
  • the distinct heavy chains differ in size: ⁇ , ⁇ , and ⁇ contain approximately 450 amino acids, while ⁇ and ⁇ contain approximately 550 amino acids.
  • IgA immunoglobulin A
  • IgD immunoglobulin D
  • IgE immunoglobulin G
  • IgM immunoglobulin M
  • Subclasses include IgG1, IgG2, IgG3 and IgG4.
  • the “light chain” refers to a polypeptide chain of about 25 kDa, wherein the amino-terminal portion includes a variable region of about 100 to about 110 or more amino acids, and the carboxy-terminal portion includes a constant region.
  • the approximate length of the light chain is 211 to 217 amino acids. Based on the amino acid sequence of the constant domain, there are two distinct types, referred to as kappa ( ⁇ ) or lambda ( ⁇ ).
  • CDR is used interchangeably with “hypervariable region,” “HVR,” and “complementarity determining region.”
  • CDR refers to one of the three hypervariable regions (H1, H2, or H3) within the non-framework region of an immunoglobulin (Ig or antibody) VH ⁇ -sheet framework or a non-framework region of the antibody VL ⁇ -sheet framework Refers to one of three hypervariable regions (L1, L2, or L3) within the hypervariable region.
  • CDR1, CDR2 and CDR3 within the VH domain are also referred to as HCDR1, HCDR2 and HCDR3, respectively.
  • CDR1, CDR2 and CDR3 within the VL domain are also referred to as LCDR1, LCDR2 and LCDR3, respectively. Accordingly, CDRs are variable region sequences interspersed within framework region sequences.
  • CDR regions are well known to those skilled in the art and have been defined by a well-known numbering system.
  • Kabat complementarity determining regions are based on sequence variability and are the most commonly used (e.g., Kabat et al., supra; Nick Deschacht et al., J Immunol 2010; 184 :5696-5704]).
  • Chothia instead refers to the position of a structural loop (see, e.g., Chothia and Lesk, J. Mol. Biol. 196:901-17 (1987)).
  • the end of the Chotia CDR-H1 loop varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering system places insertions at H35A and H35B; 35A or If neither 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34.
  • the AbM hypervariable region represents a compromise between the Kabat CDRs and Chotia structural loops and is used by Oxford Molecular's AbM antibody modeling software. “Contact” hypervariable regions are based on analysis of available complex crystal structures.
  • IMGT ImMunoGeneTics
  • IMGT ImMunoGeneTics
  • IGs immunoglobulins
  • TCRs T-cell receptors
  • MHC major histocompatibility complexes
  • CDRs are referred to both in terms of amino acid sequence and position within the light or heavy chain. Because the "position" of the CDRs within the structure of immunoglobulin variable domains is conserved between species and exist in structures referred to as loops, a numbering system is used to align variable domain sequences according to structural features, CDRs and framework residues. is easily identified.
  • This information can be used to graft and replace CDR residues from one species of immunoglobulin into a recipient framework, typically from a human antibody.
  • An additional numbering system (AHon) is described in Honegger and Plckthun, J. Mol. Biol. 309: 657-70 (2001)].
  • the correspondence between numbering systems including, for example, the Kabat numbering and the IMGT unique numbering system, is well known to those skilled in the art.
  • CDR complementarity determining region
  • a scheme is specified for the identification of a specific CDR or CDRs, such as CDRs defined by the IMGT, Kabat, Chotia or Contact methods.
  • the specific amino acid sequence of the CDR is provided.
  • the CDR region can also be defined by a combination of various numbering systems, for example a combination of the Kabat and Chotia numbering systems or a combination of the Kabat and IMGT numbering systems. Accordingly, terms such as “CDR1 as presented in a particular VH” include, but are not limited to, any CDR1 as defined by the exemplary CDR numbering system described above. Once a variable region (e.g., VH or VL) is given, one skilled in the art will understand that the CDRs within the region may be defined by different numbering systems or combinations thereof.
  • the "constant region” or “constant domain” refers to the carboxy-terminal portions of the light and heavy chains that are not directly involved in the binding of the antibody to the antigen, but exhibit various effector functions, such as interaction with Fc receptors. .
  • the term refers to the portion of an immunoglobulin molecule that has a more conserved amino acid sequence compared to other portions of the immunoglobulin, the variable region containing the antigen binding site.
  • the constant region may contain the CH1, CH2 and CH3 regions of the heavy chain and the CL region of the light chain.
  • the “framework” or “FR” refers to the variable region residues flanking the CDR.
  • FR residues are present in, for example, chimeric, humanized, human, domain antibodies, diabodies, linear antibodies, and bispecific antibodies.
  • FR residues are variable domain residues other than hypervariable region residues or CDR residues.
  • the mesothelin (MSLN) binding domain may include a light chain variable region (VL) of an antibody represented by SEQ ID NO: 8, or a light chain variable region of an antibody encoded by the nucleotide sequence represented by SEQ ID NO: 9. (VL) may be included.
  • the mesothelin (MSLN) binding domain may include the heavy chain variable region (VH) of the antibody represented by SEQ ID NO: 10, or the heavy chain variable region of the antibody encoded by the nucleotide sequence represented by SEQ ID NO: 11 (VH) may be included.
  • VH heavy chain variable region
  • VH the heavy chain variable region of the antibody represented by SEQ ID NO: 10
  • VH the heavy chain variable region of the antibody encoded by the nucleotide sequence represented by SEQ ID NO: 11
  • the light chain variable region includes an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence shown in SEQ ID NO: 8. It can be done, and also includes those encoded by a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the nucleotide sequence shown in SEQ ID NO: 9.
  • the heavy chain variable region includes an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence shown in SEQ ID NO: 10. It can be done, and also includes those encoded by sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the base sequence shown in SEQ ID NO: 11.
  • the scFv has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 amino acid residues between its VL region and VH region. , 18, 19, 20, 25, 30, 35, 40, 45, 50 or more linkers.
  • the linker sequence may include any naturally occurring amino acid.
  • the linker sequence may include the amino acids glycine and serine.
  • the linker sequence comprises a set of glycine and serine repeats, such as (Gly 4 Ser)n, where n may be a positive number of 1 or more, preferably 3 or 4.
  • the linker may include the amino acid sequence represented by SEQ ID NO: 12, or may be a linker encoded by the nucleotide sequence represented by SEQ ID NO: 13.
  • the mesothelin (MSLN) binding domain may include an scFv capable of specifically recognizing mesothelin, and the scFv is represented by the amino acid sequences of SEQ ID NOs: 2, 3, and 4, respectively.
  • a light chain variable region (VL) comprising light chain CDR1, CDR2, and CDR3;
  • a linker comprising the amino acid sequence shown in SEQ ID NO: 12 or encoded by the nucleotide sequence shown in SEQ ID NO: 13;
  • a heavy chain variable region (VH) comprising heavy chain CDR1, CDR2, and CDR3 represented by the amino acid sequences of SEQ ID NOs: 5, 6, and 7, respectively, but is not limited thereto.
  • the mesothelin (MSLN) binding domain may include an scFv capable of specifically recognizing mesothelin, and the scFv may include the amino acid sequence shown in SEQ ID NO: 8, or Variable light chain region (VL) of the antibody encoded by the nucleotide sequence shown in SEQ ID NO: 9; A linker comprising the amino acid sequence shown in SEQ ID NO: 12 or encoded by the nucleotide sequence shown in SEQ ID NO: 13; and a variable heavy chain region (VH) of an antibody comprising the amino acid sequence represented by SEQ ID NO: 10 or encoded by the nucleotide sequence represented by SEQ ID NO: 11; but is not limited thereto.
  • the chimeric antigen receptor of the present invention can be designed with a signal peptide added to direct the translated chimeric protein to the membrane.
  • the signal peptide may be included in the amino-terminus (N-ter) of the chimeric antigen receptor.
  • this signal peptide can be selectively cleaved from the mesothelin binding domain (eg, scFv) while the chimeric antigen receptor is processed in the cell and localized to the cell membrane.
  • the signal peptide generally ranges from 15 to 30 amino acids.
  • Non-limiting examples of the signal peptide in the present invention include CD8 signal peptide (amino acids 21), CD33 signal peptide (amino acids 17), CD4 signal peptide (amino acids 25), and IL-2R (CD25) signal peptide (amino acids 21). dog), trypsinogen-2 signal peptide (15 amino acids), VEGFR1 signal peptide (26 amino acids), EGFR signal peptide (24 amino acids), GMCSFR signal peptide (22 amino acids), IgVL signal peptide, IgVK signal peptide Or there may be Ig VH signal peptide, etc.
  • the signal peptide may be a CD8 signal peptide, and preferably may include an amino acid sequence represented by SEQ ID NO: 14, or a signal peptide encoded by the nucleotide sequence represented by SEQ ID NO: 15. It can be.
  • the signal peptide may include an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence represented by SEQ ID NO: 14, , also includes those encoded by sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the base sequence shown in SEQ ID NO: 15.
  • the chimeric antigen receptor of the present invention may further include an extracellular spacer, that is, a hinge.
  • the "hinge” refers to a flexible polypeptide connector region (also referred to herein as a "hinge region”) that provides structural flexibility and spacing to the flanking polypeptide regions and is used in natural or synthetic polypeptides. It can be composed of:
  • the chimeric antigen receptor of the present invention can connect the mesothelin binding domain to the transmembrane domain described below through a hinge.
  • the hinge is sufficiently flexible to allow the antigen binding domain to be oriented in different directions to facilitate antigen binding.
  • the hinge may be a hinge region derived from IgG, and preferably may include the amino acid sequence of a human IgG1, IgG2, IgG3, or IgG4 hinge region. Additionally, the hinge may contain one or more amino acid substitutions and/or insertions and/or deletions compared to the wild-type (naturally occurring) hinge region. For example, His229 of the human IgG1 hinge may contain a sequence substituted with Tyr.
  • the hinge includes all or part of the CD8, CD28, 4-1BB, OX40, CD3 zeta ( ⁇ ) chain, T cell receptor ⁇ or ⁇ chain, CD28, CD3 ⁇ , CD45 commonly used in the art.
  • CD8, 4-1BB, OX40, CD3 zeta ( ⁇ ) chain, T cell receptor ⁇ or ⁇ chain, CD28, CD3 ⁇ , CD45 commonly used in the art.
  • CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, CD154 their functional derivatives, or combinations thereof.
  • CD4 CD5
  • CD8 CD9 CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS
  • the hinge may be a CD8-derived hinge region, and preferably may include an amino acid sequence represented by SEQ ID NO: 16, or a hinge encoded by the nucleotide sequence represented by SEQ ID NO: 17. You can.
  • the hinge may include an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence represented by SEQ ID NO: 16, and the sequence It also includes those encoded by sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the base sequence indicated by number 17.
  • the chimeric antigen receptor of the present invention may further include a transmembrane domain.
  • the transmembrane domain includes one or more additional amino acids adjacent to the transmembrane region, for example, one or more amino acids associated with the extracellular region of the protein from which the transmembrane domain is derived (e.g., amino acid 1 of the extracellular region, 2, 3, 4, 5, 6, 7, 8, 9, 10 and up to 15 amino acids) and/or one or more additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived (e.g. It may contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, up to 15 amino acids in the region.
  • one or more amino acids associated with the extracellular region of the protein from which the transmembrane domain is derived e.g., amino acid 1 of the extracellular region, 2, 3, 4, 5, 6, 7, 8, 9, 10 and up to 15 amino acids
  • additional amino acids associated with the intracellular region of the protein from which the transmembrane protein is derived e.g. It may contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, up to 15 amino acids in the region.
  • the transmembrane domain is selected or has amino acid substitutions to prevent binding of this domain to the transmembrane domain of the same or another surface membrane protein, for example to minimize interaction with other members of the receptor complex. It can be transformed by .
  • the transmembrane domain may be of natural origin or recombinant origin.
  • the domain may be derived from any membrane-bound or transmembrane protein.
  • the transmembrane domain can transmit a signal to the intracellular domain whenever the chimeric antigen receptor binds to the target antigen.
  • the transmembrane domain includes, for example, T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8 (e.g., CD8 alpha, CD8 beta), CD9, CD16, CD22, CD33, CD37, CD64. , CD80, CD86, CD134, CD137, and may include the transmembrane region of the alpha, beta or zeta chain of CD154, but are not limited thereto.
  • the transmembrane domain is a co-stimulatory signaling domain, such as MHC class I molecule, TNF receptor protein, immunoglobulin-like protein, cytokine receptor, integrin, signaling lymphocyte activation molecule (SLAM protein) ), activated NK cell receptor, BTLA, Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7 -H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta , IL2R gamma, IL7R alpha, ITGA4, V
  • the transmembrane domain when the transmembrane domain is a synthetic domain, it may include hydrophobic residues such as leucine and valine, or triplets consisting of phenylalanine, tryptophan and valine may be found at both ends of the synthetic transmembrane domain. .
  • the transmembrane domain may be a CD8 transmembrane domain, and preferably includes the amino acid sequence represented by SEQ ID NO: 18, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 19.
  • the transmembrane domain may include an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence represented by SEQ ID NO: 18. It also includes those encoded by a base sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the base sequence shown in SEQ ID NO: 19.
  • the chimeric antigen receptor of the present invention has a cytoplasmic domain and may further include an intracellular signaling domain.
  • the “intracellular signaling domain” generally induces activation of the normal effector function of the cell into which the chimeric antigen receptor has been introduced.
  • the “effector function” refers to a specialized function of a cell.
  • the effector function of a T cell may be, for example, cytolytic activity or helper activity, including secretion of cytokines.
  • the “intracellular signaling domain” refers to a portion of a protein that converts an effector function signal and instructs the cell to perform a specialized function. Typically the entire intracellular signaling domain can be used, but in many cases the entire domain is not required to be used.
  • intracellular signaling domain is thus meant to contain any truncated region of the intracellular signaling domain sufficient to transduce an effector function signal.
  • intracellular signaling domains in the present invention include cytoplasmic sequences of the T cell receptor (TCR) and co-receptors that act cooperatively to initiate signal transduction after binding to the antigen receptor, as well as any derivatives of these sequences. Or there may be variants, and any recombinant sequences with the same functional capacity, etc. It is known that signals generated through TCR fragments are insufficient to fully activate T cells, and secondary and/or costimulatory signals are also required.
  • TCR T cell receptor
  • the primary signaling domain controls the primary activation of the TCR complex in a stimulatory or inhibitory manner.
  • the primary intracellular cleavage domain that acts in a stimulatory manner may be a specific signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM.
  • ITAMs containing primary intracellular signaling domains specifically used in the present invention include TCR zeta, FcR gamma, FcR beta, CD3 zeta, CD3 gamma, CD3 delta, CD3 epsilon, CD5, CD22, CD79a, CD79b, CD278. (also known as “ICOS”), Fc.epsilon.RI, DAP10, DAP12, and ITAM of CD66d, but are not limited thereto.
  • the primary signaling domain may include a modified ITAM domain, for example, a mutated ITAM domain whose activity is altered (e.g., increased or decreased) compared to the original ITAM domain.
  • a modified ITAM domain for example, a mutated ITAM domain whose activity is altered (e.g., increased or decreased) compared to the original ITAM domain.
  • the intracellular signaling domain may comprise a primary signaling domain, e.g., the CD3 zeta signaling domain itself, or may be combined with any other preferred intracellular signaling domain available in the present invention. You can.
  • the intracellular signaling domain of a CAR may include one or more co-stimulatory signaling domains along with a primary signaling domain, such as a CD3 zeta chain region.
  • the “co-stimulatory signaling domain” includes the intracellular domain of a co-stimulatory molecule and is a cell surface molecule other than an antigen receptor or its ligand required for an effective response of lymphocytes to an antigen.
  • examples of such molecules in the present invention include MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activation molecules (SLAM proteins), activated NK cell receptors, BTLA, Toll ligands.
  • Receptor OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS ( CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA- 1, ITGB7,
  • the intracellular signaling domain may be designed to include one or more, or two or more, for example, 2, 3, 4, 5 or more co-stimulatory signaling domains.
  • neighboring molecules may be connected directly, but by linker molecules. They may also be placed spaced apart.
  • the linker molecule may be a glycine residue or an alanine residue, but is not limited thereto.
  • the intracellular signaling domain may be a CD3 zeta signaling domain, and preferably includes the amino acid sequence represented by SEQ ID NO: 20, or is encoded by the nucleotide sequence represented by SEQ ID NO: 21. You can.
  • the intracellular signaling domain may further include 4-1BB as a co-stimulatory signaling domain.
  • the 4-1BB preferably includes the amino acid sequence represented by SEQ ID NO: 22, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 23.
  • the intracellular signaling domain may include a CD3 zeta signaling domain as a primary signaling domain and 4-1BB as a co-stimulatory domain, preferably 4-1BB.
  • the CD3 zeta signaling domain may include the amino acid sequence represented by SEQ ID NO: 20, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 21, and the 4-1BB is the amino acid sequence represented by SEQ ID NO: 22. It may include or be encoded by the nucleotide sequence shown in SEQ ID NO: 23.
  • the intracellular signaling domain may be linked directly to the C-terminus of the transmembrane domain, but alternatively may be linked to a short oligopeptide or polypeptide linker, e.g., 2-10 amino acids in length. Can be connected by a linker.
  • the type of the linker is not limited, but a non-limiting example may be a glycine-serine doublet.
  • the chimeric antigen receptor may include the amino acid sequence represented by SEQ ID NO: 24, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 26, but is not limited thereto.
  • the chimeric antigen receptor may include the amino acid sequence represented by SEQ ID NO: 25, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 27, but is not limited thereto.
  • the cells may further include cells genetically engineered to express a peptide or fragment thereof capable of inhibiting the transforming growth factor- ⁇ (TGF- ⁇ ) signaling pathway.
  • TGF- ⁇ transforming growth factor- ⁇
  • the present invention includes cells genetically engineered to express both the chimeric antigen receptor and the peptide or fragment thereof that inhibit the transforming growth factor beta pathway, as well as cells genetically engineered to express the chimeric antigen receptor. It is also included in the purpose of the present invention to simultaneously include two types of cells and cells genetically engineered to express a peptide or fragment thereof that inhibits the transforming growth factor beta pathway.
  • the "transforming growth factor- ⁇ (TGF- ⁇ )” refers to a cytokine belonging to the TGF- ⁇ family.
  • TGF- ⁇ expressed in mammals include TGF- ⁇ 1 and TGF- ⁇ . Three are known: - ⁇ 2 and TGF- ⁇ 3.
  • Signal transduction by TGF- ⁇ plays a critical role in various biological processes and performs various functions such as cell growth inhibition, apoptosis, differentiation, and epithelial-mesenchymal transition (EMT).
  • EMT epithelial-mesenchymal transition
  • the TGF- ⁇ signaling system is tightly regulated and plays a critical role in development and organ formation as well as maintaining cellular homeostasis. Therefore, disruption of TGF- ⁇ signaling can cause life-threatening diseases such as cancer, fibrosis, and congenital malformations.
  • the peptide may include the amino acid sequence represented by SEQ ID NO: 28, preferably consisting of the amino acid sequence represented by SEQ ID NO: 28.
  • sequence encoding the peptide may include the nucleotide sequence represented by SEQ ID NO: 29, preferably consisting of the nucleotide sequence represented by SEQ ID NO: 29.
  • the peptide may bind to the TGF- ⁇ receptor (TGFBR1 and/or TGFBR2) to inhibit TGF- ⁇ signaling.
  • TGFBR1 and/or TGFBR2 TGF- ⁇ receptor
  • the peptide competes with TGF- ⁇ and binds to the TGF- ⁇ receptor. It may be that TGF- ⁇ signaling is inhibited through a mechanism that prevents TGF- ⁇ cytokines from binding to the TGF- ⁇ receptor.
  • the peptide may inhibit TGF- ⁇ signaling by suppressing the expression level of TGF- ⁇ in cells.
  • the peptide may inhibit TGF- ⁇ signaling in cells through an auto-inhibition pathway. It may be suppressing TGF- ⁇ signaling through a mechanism that reduces the expression level or reduces the extracellular emissions of TGF- ⁇ .
  • the cells may be immune effector cells.
  • the “immune effector cell” may be a lymphoid cell that participates in an immune response, such as promoting an immune effector response.
  • lymphocytes refers to cells that are commonly found in lymph and include natural killer cells (NK cells), T cells, and B cells. Those skilled in the art will understand that the immune cell types listed above can be further divided into subtypes.
  • the lymphocytes may be or include Natural Killer Cells (NK cells), but are not limited thereto.
  • NK cells Natural Killer Cells
  • the "Natural Killer Cells (NK cells)” are defined as large granular lymphocytes (LGL), which constitute three types of cells differentiated from common lymphoid progenitor cells-producing B and T lymphocytes.
  • NK cells are known to differentiate and mature in the bone marrow, lymph nodes, spleen, tonsils, and thymus and enter the circulation.
  • the NK cells may include any type of NK cell without limitation, for example, cultured NK cells, such as primary NK cells, NK cells from a cultured NK cell line, or NK cells obtained from a mammal. It may be an NK cell, but is not limited thereto.
  • NK cells When NK cells are obtained from a mammal, the NK cells can be obtained from a number of sources, including but not limited to blood, bone marrow, lymph nodes, thymus, or other tissues or body fluids. NK cells may be concentrated or purified. The NK cells may preferably be human NK cells (eg, isolated from humans). NK cell lines are available, for example, from ATCC (American Type Culture Collection) and include, for example, NK-92 cells (ATCC CRL-2407), NK92MI cells (ATCC CRL-2408) or derivatives thereof, etc. .
  • ATCC American Type Culture Collection
  • a polynucleotide (or gene construct) encoding the above-mentioned chimeric antigen receptor and a polynucleotide (or gene) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway are included in one vector. construct), or a vector containing a polynucleotide (or gene construct) encoding the chimeric antigen receptor, and a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway.
  • Encoding may include both types of vectors containing polynucleotides (or gene constructs).
  • the "vector” refers to a recombinant vector that can be transfected into a suitable host cell to express a protein of interest, and refers to a genetic construct containing essential regulatory elements operably linked to express the gene insert.
  • operably linked means that the nucleic acid expression control sequence and the nucleic acid sequence encoding the protein of interest are functionally linked to perform a general function. Operational linkage with a recombinant vector can be prepared using genetic recombination techniques well known in the art, and site-specific DNA cutting and ligation can be easily performed using enzymes generally known in the art. there is.
  • various types of vectors such as nanoparticles, plasmids, viruses, and cosmids can be used as recombinant expression vectors for inserting the foreign genes.
  • the type of recombinant vector is not particularly limited as long as it functions to express the desired gene and produce the desired protein in various host cells of prokaryotic and eukaryotic cells, but specifically, it has a highly active promoter and strong expression ability while maintaining a natural state. Vectors that can produce large quantities of foreign proteins of a similar form can be used.
  • a variety of gene delivery vehicles are known in the art and include both viral and non-viral (e.g., naked DNA, plasmid) vectors.
  • Viral vectors suitable for gene transfer are known to those skilled in the art.
  • Non-limiting examples of the viral vectors include retroviral vectors (derived from Moloney murine leukemia virus vector (MoMLV), MSCV, SFFV, MPSV, SNV, etc.), lentiviral vectors (e.g., HIV-1, HIV-2) , derived from SIV, BIV, FIV, etc.), adenovirus (Ad) vectors, including replication-competent, replication-deficient and anergic forms thereof, adeno-associated virus (AAV) vectors, simian virus 40 (SV-40) vectors , bovine papilloma virus vector, Epstein Barr virus vector, herpes virus vector, chicken pox virus vector, Harvey rat sarcoma virus vector, rat mammary tumor virus vector, Rous s
  • Non-viral vectors for gene transfer include naked DNA, plasmids, transposons, and mRNA.
  • Non-limiting examples include pKK plasmid (Clonetech), pUC plasmid, pET plasmid (Novagen, Inc., Madison, Wis.), pRSET or pREP plasmid (Invitrogen, San Diego, Calif.), pMAL plasmid (New England Biolabs, Beverly , Mass.).
  • vectors in the present invention can be introduced into many suitable host cells using methods disclosed or cited herein or otherwise known to those skilled in the art.
  • a suitable expression vector of the present invention may include a base sequence encoding a signal peptide for membrane targeting or secretion in addition to expression control elements such as a promoter, start codon, stop codon, polyadenylation signal, or enhancer.
  • the initiation codon and stop codon are generally considered to be part of the nucleotide sequence encoding the immunogenic target protein and must be functional in the subject when the genetic construct is administered and must be in frame with the coding sequence.
  • the "promoter”, as used herein, refers to any sequence that regulates the expression of a coding sequence, such as a gene. Promoters can be, for example, constitutive, inducible, repressible, or tissue-specific.
  • a promoter is a control sequence, a region of polynucleotide sequence where the initiation and rate of transcription is controlled.
  • Non-limiting examples of the promoters in the present invention include Rous sarcoma virus (RSV) LTR promoter (optionally with RSV enhancer), cytomegalovirus (CMV) promoter, SV40 promoter, dihydrofolate reductase Promoter, ⁇ -actin promoter, phosphoglycerol kinase (PGK) promoter, U6 promoter, EF1alpha short form (EFS) promoter, human polypeptide chain elongation factor (EF1a) promoter, P5 promoter, Ubc promoter, CAG promoter, TRE promoter , UAS promoter, Ac5 promoter, polyhedrin promoter, CaMKIIa promoter, Gal1 promoter, TEF1 promoter, GDS promoter, ADH1 promoter, CaMV35S promoter, ubiquitin (Ubi) promoter
  • the promoter can be coupled to an enhancer to increase transcription efficiency.
  • the enhancer may include, but are not limited to, the RSV enhancer, the CMV enhancer, or the ⁇ -fetoprotein MERII enhancer.
  • the promoter may be the SSFV promoter, preferably the SSFV promoter represented by SEQ ID NO: 30, but is not limited thereto.
  • the polynucleotide (or gene construct) encoding the chimeric antigen receptor may include the base sequence represented by SEQ ID NO: 26, but is not limited thereto.
  • the polynucleotide encoding the chimeric antigen receptor may include the base sequence represented by SEQ ID NO: 27, but is not limited thereto.
  • the vector of the present invention may further include a sequence encoding a signal peptide upstream of the polynucleotide encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta (TGF- ⁇ ) signaling pathway.
  • TGF- ⁇ transforming growth factor beta
  • the signal peptide may be an IL-2 signal peptide, and preferably includes the amino acid sequence represented by SEQ ID NO: 33, or is encoded by the nucleotide sequence represented by SEQ ID NO: 34. However, it is not limited to this.
  • the vector may include one or more additional polypeptides, for example, a polynucleotide encoding one or more markers and/or one or more effector molecules.
  • the one or more markers may include a transduction marker, a surrogate marker, and/or a selection marker.
  • the additional nucleic acid sequences introduced, encoding one or more additional polypeptides may be nucleic acid sequences that can improve the efficacy of the therapy, such as by promoting the viability and/or function of the transferred cells; Nucleic acid sequences that provide genetic markers for evaluation and/or selection of cells, such as to assess survival or localization in vivo; Lupton S. D. et al., Mol.
  • the marker may be a transduction marker or a surrogate marker.
  • the transduction marker or surrogate marker can be used to detect cells into which a polynucleotide (or gene construct) of the present invention, that is, a polynucleotide containing a sequence encoding the peptide of the present invention or a fragment thereof, has been introduced.
  • the transduction marker may indicate or confirm transformation of the cell
  • the surrogate marker may be a protein prepared to be co-expressed on the cell surface together with the peptide or fragment.
  • the surrogate marker may be a surface protein modified to have little or no activity.
  • the surrogate marker may be encoded on the same polynucleotide encoding the peptide or fragment.
  • the nucleic acid sequence encoding the peptide or fragment thereof may optionally be operably linked to a nucleic acid encoding a self-cleaving peptide or a peptide that causes ribosome skipping, such as a 2A sequence.
  • the exemplary surrogate marker is a truncated form of a cell surface polypeptide, e.g., a non-functional, full-length form of the cell surface polypeptide that cannot or will not transmit signals or signals that would normally be transmitted and/or will be internalized. It may contain truncated forms that cannot or are not internalized.
  • Exemplary truncated cell surface polypeptides include truncated forms of growth factors or other receptors, such as truncated human epidermal growth factor receptor 2 (tHER2), truncated epidermal growth factor receptor (tEGFR), or It may include prostate-specific membrane antigen (PSMA) or a modified form thereof, such as truncated PSMA (tPSMA).
  • tEGFR may contain an epitope recognized by the antibody cetuximab (Erbitux) or other therapeutic anti-EGFR antibody or binding molecule, which identifies cells engineered with the tEGFR construct and the encoded exogenous protein. or to select and/or remove or isolate cells expressing the encoded exogenous protein.
  • cetuximab an epitope recognized by the antibody cetuximab (Erbitux) or other therapeutic anti-EGFR antibody or binding molecule, which identifies cells engineered with the tEGFR construct and the encoded exogenous protein. or to select and/or remove or isolate cells expressing the encoded exogenous protein.
  • the marker e.g., a surrogate marker
  • the marker includes all or part of CD34 (e.g., a truncated form), NGFR, CD19, or truncated CD19, e.g., truncated non-human CD19.
  • the marker is a detectable protein, such as a fluorescent protein, such as green fluorescent protein (GFP), enhanced green fluorescent protein (EGFP), such as super-fold GFP. , sfGFP), red fluorescent protein (RFP), such as tdTomato, mCherry, mStrawberry, AsRed2, DsRed or DsRed2, cyan fluorescent protein (CFP), blue green fluorescent protein (BFP) ), enhanced blue fluorescent protein (EBFP) yellow fluorescent protein (YFP) and its variants, including species variants, monomer variants and codon-optimized, stabilized and/or enhanced variants of fluorescent proteins. It may be or contain a variant.
  • a fluorescent protein such as green fluorescent protein (GFP), enhanced green fluorescent protein (EGFP), such as super-fold GFP.
  • sfGFP red fluorescent protein
  • RFP red fluorescent protein
  • CFP blue green fluorescent protein
  • BFP blue green fluorescent protein
  • EBFP enhanced blue fluorescent protein
  • YFP yellow fluorescent protein
  • the marker is an enzyme such as luciferase, E. coli-derived lacZ gene, alkaline phosphatase, secreted embryonic alkaline phosphatase (SEAP), chloramphenicol acetyl transferase (CAT), and ⁇ -gal. It is or includes lactosidase or ⁇ -glucuronidase ( ⁇ -glucuronidase, GUS). Expression of the enzyme can be detected by addition of a substrate that can be detected upon expression and functional activity of the enzyme.
  • the marker may be green fluorescent protein (GFP), and preferably, the marker includes the amino acid sequence represented by SEQ ID NO: 31, or is encoded by the nucleotide sequence represented by SEQ ID NO: 32. It may be possible, but it is not limited to this.
  • GFP green fluorescent protein
  • the marker may be a selection marker.
  • the selection marker may be or include a polypeptide that confers resistance to an exogenous agent or drug.
  • the selection marker may be an antibiotic resistance gene, non-limiting examples include a puromycin resistance gene, hygromycin resistance gene, blasticidin resistance gene, neomycin resistance gene, geneticin resistance gene, or zeocin. It may be or contain a resistance gene or a modified form thereof.
  • the vector includes a polynucleotide (or gene construct) encoding the chimeric antigen receptor; A polynucleotide (or gene construct) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway; and a polynucleotide (or gene construct) encoding a marker, but is not limited thereto.
  • the vector includes a polynucleotide (or gene construct) encoding the chimeric antigen receptor in one vector; A polynucleotide (or gene construct) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway; and a polynucleotide (or gene construct) encoding a marker, but is not limited thereto.
  • the position order or combination order of each of these genes in the vector is not particularly limited.
  • the vector is a vector containing a polynucleotide (or gene construct) encoding the chimeric antigen receptor; And a vector containing a polynucleotide (or gene construct) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway; but is not limited thereto.
  • the vector is a vector containing a polynucleotide (or gene construct) encoding the chimeric antigen receptor; And a vector containing a polynucleotide (or gene construct) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway, wherein a marker is added to at least one of the two vectors.
  • Encoding polynucleotides (or gene constructs) may be additionally included, but are not limited thereto.
  • polynucleotide when two separate polynucleotides (or gene constructs) are included in one vector, for example, a polynucleotide (or gene construct) encoding the chimeric antigen receptor in one vector; A polynucleotide (or gene construct) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway; And when at least two of the polynucleotides (or gene constructs) encoding the marker are included simultaneously, they may be operably linked by one promoter, or may be operably linked by two or more promoters.
  • a self-cleavable peptide is provided so that each of them can be individually delivered or introduced into the cell for intracellular expression. It may further include a polynucleotide encoding.
  • the "self-cleavage peptide” refers to 10 to 50, 12 to 42, 14 to 34 peptides that can induce cleavage of proteins synthesized within cells. It refers to a peptide consisting of 16 to 26 or 18 to 22 amino acids.
  • the self-cleaving peptide may be derived from the 2A region of the viral gene.
  • the self-cleaving peptide may be derived from P2A, E2A, F2A or T2A.
  • the self-cleaving peptide may be derived from P2A.
  • a peptide that is cleaved by a degrading enzyme present in the cytoplasm can be used.
  • the self-cleavable peptide may be P2A or a peptide derived therefrom, and preferably includes the amino acid sequence represented by SEQ ID NO: 35, or is cleaved by the nucleotide sequence represented by SEQ ID NO: 36. It may be encrypted.
  • a cell therapeutic agent comprising the genetically engineered cells provided by the present invention as an active ingredient.
  • the “cell therapy” refers to a treatment using autologous, allogenic, or xenogenic cells to restore tissue function, and is used to suppress cancer. If the immune effector cells, for example, genetically modified natural killer cells, are included as active ingredients, they can be used as a cell therapy for the treatment and prevention of cancer.
  • the cell therapeutic agent may further include a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may be, for example, saline solution, sterilized water, Ringer's solution, buffered saline solution, dextrose solution, maltodextrin solution, glycerol, ethanol, HSA (Human serum albumin), and a mixture of one or more of these ingredients.
  • Other common additives such as antioxidants, buffers, and bacteriostatic agents can be added as needed.
  • the cell therapeutic agent may be used as a suspending agent, solubilizing agent, stabilizer, isotonic agent, preservative, anti-adsorption agent, surfactant, diluent, excipient, pH adjuster, analgesic agent, buffer, sulfur-containing agent, etc. ⁇ ) Reducing agents, antioxidants, etc. can be added appropriately.
  • the suspending agent include, but are not limited to, methylcellulose, polysorbate 80, hydroxyethylcellulose, gum arabic, traganmal, sodium carboxymethylcellulose, polyoxyethylene sorbitan monolaurate, etc. no.
  • the solution auxiliary agent includes polyoxyethylene hydrogenated castor oil, polysorbate 80, nicotinic acid amide, polyoxyethylene sorbitan monolaurate, mecrogol, castor oil fatty acid ethyl ester, etc.
  • Stabilizers include, but are not limited to, dextran 40, methylcellulose, gelatin, sodium sulfite, and sodium metasulfate.
  • the isotonic agent includes, for example, D-mannitol, sorbitol, etc., but is not limited thereto.
  • the preservative includes, for example, methyl paraoxybenzoate, ethyl paraoxybenzoate, sorbic acid, phenol, cresol, chlorocresol, etc., but is not limited thereto.
  • the anti-adsorption agent includes, for example, human serum albumin, lecithin, dextran, ethylene oxide propylene oxide copolymer, hydroxypropyl cellulose, methyl cellulose, polyoxyethylene hydrogenated castor oil, polyethylene glycol, etc. , but is not limited to this.
  • the sulfur-containing reducing agent includes, for example, N-acetylcysteine, N-acetylhomocysteine, thioctoic acid, thiodiglycol, thioethanolamine, thioglycerol, thiosorbitol, thioglycolic acid and its salts, sodium thiosulfate, Examples include those having a sulfuhydryl group such as glutathione and thioalkanoic acid having 1 to 7 carbon atoms, but are not limited thereto.
  • the antioxidants include, for example, erythorbic acid, dibutylhydroxytoluene, butylhydroxyanisole, ⁇ -tocopherol, tocopherol acetate, L-ascorbic acid and its salts, L-ascorbic acid palmitate, Chelating agents such as L-ascorbate stearate, sodium bisulfite, sodium sulfite, triamyl gallate, propyl gallate or sodium ethylenediaminetetraacetate (EDTA), sodium pyrophosphate, and sodium metaphosphate may be included, but are limited thereto. It doesn't work.
  • the cell therapeutic agent is used, for example, based on an adult patient weighing 70 kg, about 1,000 to 10,000 cells/time, 1,000 to 100,000 cells/time, 1,000 to 1,000,000 cells/time, 1,000 to 10,000,000. , 1,000 ⁇ 100,000,000 cells/time, 1,000 ⁇ 1,000,000,000 cells/time, 1,000 ⁇ 10,000,000,000 cells/circuit, can be administered in divided doses once or several times a day at regular time intervals, or can be administered multiple times at regular time intervals.
  • the injectable product according to the present invention can be manufactured in the form of a filled injection by taking the amount commonly known in the art depending on the patient's constitution and type of defect.
  • the present invention relates to a pharmaceutical composition for preventing, improving or treating cancer containing the genetically engineered cells provided by the present invention as an active ingredient.
  • cancer refers to or refers to a physiological condition typically characterized by uncontrolled cell growth in mammals.
  • the cancer subject to prevention, improvement, or treatment in the present invention may be a solid tumor consisting of a lump generated by abnormal cell growth in a solid organ, and preferably expresses mesothelin (MSLN).
  • MSLN mesothelin
  • Any cancer that has cancer can be included without limitation, and specific examples include stomach cancer, liver cancer, glioblastoma, ovarian cancer, colon cancer, head and neck cancer, bladder cancer, renal cell cancer, breast cancer, metastatic cancer, prostate cancer, pancreatic cancer, biliary tract cancer, It may be melanoma or lung cancer, but is not limited thereto.
  • prevention refers to all actions that inhibit cancer or delay its progression by administering the composition of the present invention.
  • treatment and “improvement” mean any action in which cancer symptoms are improved or beneficially changed by administration of the composition of the present invention.
  • the pharmaceutical composition of the present invention may be formulated to include one or more pharmaceutically acceptable carriers in addition to the active ingredients described above.
  • Pharmaceutically acceptable carriers may be saline solution, sterile water, Ringer's solution, buffered saline solution, dextrose solution, maltodextrin solution, glycerol, ethanol, liposome, and a mixture of one or more of these ingredients, and if necessary, antioxidants.
  • other common additives such as buffer solutions and bacteriostatic agents can be added.
  • diluents, dispersants, surfactants, binders, and lubricants can be additionally added to formulate injectable formulations such as aqueous solutions, suspensions, and emulsions, as well as pills, capsules, granules, or tablets, and can act specifically on target organs.
  • Target organ-specific antibodies or other ligands can be used in combination with the carrier.
  • it can be preferably formulated according to each disease or ingredient using an appropriate method in the art or a method disclosed in Remington's Pharmaceutical Science (recent edition), Mack Publishing Company, Easton PA). there is.
  • the pharmaceutical composition of the present invention can be a solution, suspension, dispersion, emulsion, gel, injectable solution, or sustained-release preparation of the active compound, and is preferably an injection.
  • the pH is adjusted using a buffer solution such as an aqueous acid solution or phosphate that can be used as an injection to ensure product stability according to the distribution of the injection prescription, making the injection very physically and chemically stable. It can be manufactured with
  • the injection can be prepared by dissolving it in water for injection along with a stabilizer or solubilizing agent and then sterilizing it, especially by high-temperature reduced-pressure sterilization or aseptic filtration.
  • the water for injection may be distilled water for injection or a buffer solution for injection, for example, a phosphate buffer solution with a pH in the range of 3.5 to 7.5 or a sodium dihydrogen phosphate (NaH2PO4)-citric acid buffer solution.
  • the phosphate salt used may be in the form of a sodium salt or potassium salt, an anhydrous or hydrated form, and may be in the form of citric acid or an anhydrous or hydrated form.
  • the stabilizer used in the present invention includes sodium pyrosulfite, sodium bisulfite (NaHSO 3 ), sodium metabisulfite (Na 2 S 2 O 3 ), or ethylenediaminetetraacetic acid
  • Solubilizing agents include bases such as sodium hydroxide (NaOH), sodium bicarbonate (NaHCO 3 ), sodium carbonate (NaCO 3 ) or potassium hydroxide (KOH), or acids such as hydrochloric acid (HCl) or acetic acid (CH 3 COOH).
  • the injectable agent according to the present invention can be formulated to be bioabsorbable, biodegradable, and biocompatible.
  • bioabsorbable we mean that the injectable agent can disappear from the body upon initial application without decomposition or decomposition of the dispersed injectable agent.
  • Biodegradability means that the injectable agent can be broken down or decomposed in the body by hydrolysis or enzymatic degradation.
  • Biosynthesis means that all ingredients are non-toxic to the body.
  • the injection according to the present invention can be prepared using conventional fillers, weighting agents, binders, wetting agents, diluents such as surfactants, or excipients.
  • composition or active ingredient of the present invention may be administered by intravenous, intraarterial, intraperitoneal, intramuscular, intrasternal, transdermal, intranasal, subcutaneous, intrathecal, inhalational, topical, rectal, oral, intraocular or intradermal route depending on the purpose. It can be administered in a conventional manner, and preferably intravenously.
  • the composition or active ingredient of the present invention can be administered by injection or catheter.
  • the dosage of the active ingredient is 1 x 10 1 to 1 x 10 50 cells/kg, preferably 1 x 10 transformed host cells contained in the pharmaceutical composition, based on an adult weighing 60 kg. So that it can be administered within the range of 1 to 1 x 10 30 pieces/kg, more preferably 1 x 10 5 to 1 x 10 20 pieces/kg, and most preferably 1 x 10 7 to 1 x 10 9 pieces/kg. It can be adjusted.
  • the optimal dosage to be administered can be easily determined by a person skilled in the art, and can be determined based on the type of disease, the severity of the disease, the content of the active ingredient and other ingredients contained in the composition, the type of dosage form, and the patient's age, weight, and general health. It can be adjusted according to various factors, including condition, gender and diet, administration time, administration route and secretion rate of the composition, treatment period, and concurrently used drugs.
  • the active ingredient may be contained in an amount of 0.001 to 50% by weight based on the total weight of the composition.
  • the content is not limited to this.
  • composition of the present invention may further include one or more anticancer agents.
  • the anticancer agents include nitrogen mustard, imatinib, oxaliplatin, rituximab, erlotinib, neratinib, lapatinib, gefitinib, vandetanib, nirotinib, semasanib, bosutinib, axitinib, Cediranib, lestaurtinib, trastuzumab, gefitinib, bortezomib, sunitinib, carboplatin, bevacizumab, cisplatin, cetuximab, Viscum album, asparaginase, tretinoin, hydroxycarbamide , dasatinib, estramustine, gemtuzumab ozogamycin, ibritumab tusetan, heptaplatin, methylaminolevulinic acid, amsacrine, alemtuzumab, procarbazin
  • tezolomide busulfan, ifosphamide, cyclophosphamide, melphalan, altretmin, dacarbazine, thiotepa, nimustine, chlorambucil, mitolactol, leucovorin, tretonin, exemestane. , aminoglutethimide, anagrelide, navelvin, fadrazole, taciphen, toremifene, testolactone, anastrozole, letrozole, borozole, bicalutamide, lomustine and carmustine.
  • One or more types selected from the group may be used, but are not limited thereto.
  • a method for preventing, improving, or treating cancer which includes administering the cell therapy or pharmaceutical composition provided by the present invention to a subject.
  • the subject may include, without limitation, mammals, birds, reptiles, farmed fish, etc., including rats, livestock, humans, etc., that develop or are at risk of developing cancer due to a TGF- ⁇ -related disease.
  • the composition can be administered singly or multiple times in a pharmaceutically effective amount.
  • the composition can be formulated and administered in the form of a solution, powder, aerosol, injection, infusion solution (injection), capsule, pill, tablet, suppository, or patch.
  • the pharmaceutical composition for preventing or treating cancer may be administered through any general route as long as it can reach the target tissue.
  • the composition is not particularly limited thereto, but depending on the purpose, intraperitoneal administration, intravenous administration, intramuscular administration, subcutaneous administration, intradermal administration, transdermal patch administration, oral administration, intranasal administration, intrapulmonary administration, and intrarectal administration It can be administered through routes such as: However, when administered orally, it can be administered in an unformulated form, and since the active ingredients of the pharmaceutical composition may be denatured or decomposed by stomach acid, the oral composition must be coated with the active agent or protected from decomposition in the stomach. It can also be administered orally in formulated form or in the form of an oral patch. Additionally, the composition can be administered by any device that allows the active substance to move to target cells.
  • the genetically engineered immune effector cells can target only target tumor cells by a chimeric antigen receptor targeting mesothelin (MSLN) expressed in the cells,
  • MSLN chimeric antigen receptor targeting mesothelin
  • TGF- ⁇ transforming growth factor beta
  • TGF- ⁇ transforming growth factor beta
  • Figure 2 shows an NK cell line genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway in the HCC-1806 breast cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 3 shows the MD-AMB-231 breast cancer cell line into which the luciferase reporter gene was introduced in Experimental Example 1 of the present invention, which was genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway.
  • the results of comparing luciferase activity after treating NK cell lines are shown graphically.
  • Figure 4 shows an NK cell line genetically engineered to express a peptide that inhibits the chimeric antigen receptor and TGF- ⁇ signaling pathway according to the present invention in the NCI-N87 gastric cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 5 shows that in Experimental Example 1 of the present invention, the AGS gastric cancer cell line into which the luciferase reporter gene was introduced was treated with an NK cell line genetically engineered to express a peptide that inhibits the chimeric antigen receptor and the TGF- ⁇ signaling pathway according to the present invention. The results of comparing luciferase activity are shown in a graph.
  • Figure 6 shows a NK cell line genetically engineered to express a peptide that inhibits the chimeric antigen receptor and TGF- ⁇ signaling pathway according to the present invention in the NCI-H292 lung cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 7 shows NK genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway in the SW-620 colon cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention.
  • the results of comparing luciferase activity after treating cell lines are shown graphically.
  • Figure 8 shows NK genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway in the SNU-1544 colon cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after treating cell lines are shown graphically.
  • Figure 9 shows the SK-HEP-1 liver cancer cell line into which the luciferase reporter gene was introduced in Experimental Example 1 of the present invention, which was genetically engineered to express a peptide that inhibits the chimeric antigen receptor and TGF- ⁇ signaling pathway according to the present invention.
  • the results of comparing luciferase activity after treating NK cell lines are shown graphically.
  • Figure 10 shows a NK cell line genetically engineered to express a peptide that inhibits the chimeric antigen receptor and TGF- ⁇ signaling pathway according to the present invention in the HEP-G2 liver cancer cell line into which the luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 11 shows an NK cell line genetically engineered to express a peptide that inhibits the chimeric antigen receptor and TGF- ⁇ signaling pathway according to the present invention in the AsPC-1 pancreatic cancer cell line into which the luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 12 shows a NK cell line genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway in the Capan-2 pancreatic cancer cell line into which a luciferase reporter gene was introduced in Experimental Example 1 of the present invention. The results of comparing luciferase activity after processing are shown in a graph.
  • Figure 13 shows the change in pSmad2/3 expression level in Aspc1, a pancreatic cancer cell line, according to treatment with the peptide of the present invention in Reference Example 1 of the present invention.
  • Figure 14 is a diagram showing the change in pSmad2/3 expression level in Bxpc3, a pancreatic cancer cell line, according to treatment with the peptide of the present invention in Reference Example 1 of the present invention.
  • Figure 15 is a diagram showing the change in pSmad2/3 expression level in Panc1, a pancreatic cancer cell line, according to treatment with the peptide of the present invention in Reference Example 1 of the present invention.
  • a chimeric antigen receptor (CAR) targeting mesothelin MSLN
  • cells genetically engineered to express a peptide or fragment thereof capable of inhibiting the transforming growth factor- ⁇ (TGF- ⁇ ) signaling pathway a chimeric antigen receptor (CAR) targeting mesothelin (MSLN)
  • TGF- ⁇ transforming growth factor- ⁇
  • the chimeric antigen receptor includes a mesothelin binding domain, and may further include one or more selected from the group consisting of a hinge domain, a signal peptide domain, a transmembrane domain, and one or more signaling domains. .
  • the mesothelin (MSLN) binding domain may include an scFv capable of specifically recognizing mesothelin, and the scFv is represented by the amino acid sequences of SEQ ID NOs: 2, 3, and 4, respectively.
  • a light chain variable region (VL) comprising light chain CDR1, CDR2, and CDR3;
  • a linker comprising the amino acid sequence shown in SEQ ID NO: 12 or encoded by the nucleotide sequence shown in SEQ ID NO: 13;
  • a heavy chain variable region (VH) comprising heavy chain CDR1, CDR2, and CDR3 represented by the amino acid sequences of SEQ ID NOs: 5, 6, and 7, respectively, but is not limited thereto.
  • the mesothelin (MSLN) binding domain may include an scFv capable of specifically recognizing mesothelin, and the scFv may include the amino acid sequence shown in SEQ ID NO: 8, or Variable light chain region (VL) of the antibody encoded by the nucleotide sequence shown in SEQ ID NO: 9; A linker comprising the amino acid sequence shown in SEQ ID NO: 12 or encoded by the nucleotide sequence shown in SEQ ID NO: 13; and a variable heavy chain region (VH) of an antibody comprising the amino acid sequence represented by SEQ ID NO: 10 or encoded by the nucleotide sequence represented by SEQ ID NO: 11; but is not limited thereto.
  • the chimeric antigen receptor of the present invention can be designed with a signal peptide added to direct the translated chimeric protein to the membrane.
  • the signal peptide may be a CD8 signal peptide, and preferably may include an amino acid sequence represented by SEQ ID NO: 14, or a signal peptide encoded by the nucleotide sequence represented by SEQ ID NO: 15. It can be.
  • the chimeric antigen receptor of the present invention may further include an extracellular spacer, that is, a hinge.
  • the hinge may be a CD8-derived hinge region, and preferably may include an amino acid sequence represented by SEQ ID NO: 16, or a hinge encoded by the nucleotide sequence represented by SEQ ID NO: 17. You can.
  • the chimeric antigen receptor of the present invention may further include a transmembrane domain.
  • the transmembrane domain may be a CD8 transmembrane domain, and preferably includes the amino acid sequence represented by SEQ ID NO: 18, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 19.
  • the chimeric antigen receptor of the present invention has a cytoplasmic domain and may further include an intracellular signaling domain.
  • the intracellular signaling domain may be a CD3 zeta signaling domain, and preferably includes the amino acid sequence represented by SEQ ID NO: 20, or is encoded by the nucleotide sequence represented by SEQ ID NO: 21. You can.
  • the intracellular signaling domain may further include 4-1BB as a co-stimulatory signaling domain.
  • the 4-1BB preferably includes the amino acid sequence represented by SEQ ID NO: 22, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 23.
  • the intracellular signaling domain may include a CD3 zeta signaling domain as a primary signaling domain and 4-1BB as a co-stimulatory domain, preferably 4-1BB.
  • the CD3 zeta signaling domain may include the amino acid sequence represented by SEQ ID NO: 20, or may be encoded by the nucleotide sequence represented by SEQ ID NO: 21, and the 4-1BB is the amino acid sequence represented by SEQ ID NO: 22. It may include or be encoded by the nucleotide sequence shown in SEQ ID NO: 23.
  • the cells may further include cells genetically engineered to express a peptide or fragment thereof capable of inhibiting the transforming growth factor- ⁇ (TGF- ⁇ ) signaling pathway.
  • TGF- ⁇ transforming growth factor- ⁇
  • the peptide may include the amino acid sequence represented by SEQ ID NO: 28, preferably consisting of the amino acid sequence represented by SEQ ID NO: 28.
  • sequence encoding the peptide may include the nucleotide sequence represented by SEQ ID NO: 29, preferably consisting of the nucleotide sequence represented by SEQ ID NO: 29.
  • the peptide may bind to the TGF- ⁇ receptor (TGFBR1 and/or TGFBR2) to inhibit TGF- ⁇ signaling.
  • TGFBR1 and/or TGFBR2 TGF- ⁇ receptor
  • the peptide competes with TGF- ⁇ and binds to the TGF- ⁇ receptor. It may be that TGF- ⁇ signaling is inhibited through a mechanism that prevents TGF- ⁇ cytokines from binding to the TGF- ⁇ receptor.
  • the cells may be immune effector cells.
  • the immune effector cells are lymphoid cells that participate in an immune response, such as promoting an immune effector response, and may be or include Natural Killer Cells (NK cells), but are not limited thereto.
  • NK cells Natural Killer Cells
  • a vector containing a gene encoding the peptide or its fragment can be transfected into the immune effector cell to introduce a foreign gene encoding the peptide or its fragment into the cell.
  • a polynucleotide (or gene construct) encoding the above-mentioned chimeric antigen receptor and a polynucleotide (or gene) encoding a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway are included in one vector. construct), or a vector containing a polynucleotide (or gene construct) encoding the chimeric antigen receptor, and a peptide or fragment thereof capable of inhibiting the transforming growth factor beta signaling pathway.
  • Encoding may include both types of vectors containing polynucleotides (or gene constructs).
  • a cell therapeutic agent comprising the genetically engineered cells provided by the present invention as an active ingredient.
  • the present invention relates to a pharmaceutical composition for preventing, improving or treating cancer containing the genetically engineered cells provided by the present invention as an active ingredient.
  • a peptide (P6) consisting of the amino acid sequence shown in SEQ ID NO: 28 targeting the TGF- ⁇ receptor was designed and produced.
  • pancreatic cancer cell lines Aspc1, Bxpc3, and Panc1 cell lines were seeded in a 6-well plate at a concentration of 1 (Control group: 15% ACN - candidate peptide solvent, DMSO - P144 peptide solvent, P144: TSLDASIWAMMQNA (SEQ ID NO: 37)) and the expression levels of pSmad2/3 and total Smad2/3 were analyzed by Western blotting method.
  • Example 1 Construction of a vector containing a chimeric antigen receptor and a TGF- ⁇ signaling pathway inhibitory gene
  • the chimeric antigen receptor expression cassette includes CD8 signal peptide, mesothelin binding domain (light chain variable region; linker; and heavy chain variable region), CD8 hinge, CD8 transmembrane domain, 4-1BB, and CD3 zeta signaling domain.
  • the recombinant gene was placed under the control of the SFFV single promoter.
  • the base sequence of the SFFV promoter used in the experiment and the base sequences of each other gene are shown in Table 1, and the amino acid sequences encoded by these base sequences are shown in Table 2 below.
  • Natural killer cells were used as immune effector cells to be genetically manipulated to express the chimeric antigen receptor and TGF- ⁇ signaling pathway inhibitory peptide.
  • the NK-92 cell line was purchased from ATCC.
  • the 293T cell line which is the cell line to be used to transduce the lentivirus, was also purchased from ATCC.
  • Both the NK-92 cell line and the lentivirus-transduced NK-92 cell line described below were treated with 100 ⁇ g/mL of streptomycin, 100 units/mL of penicillin, and 20% fetal bovine serum. serum; FBS), 10% MEM vitamin solution, 1 streptomycin), 100 units/mL of penicillin, and 10% fetal bovine serum (FBS). All cells were cultured at 37°C with 5% CO2 (95% CO2). Cultured in an environment maintained with air.
  • lentivirus In order to obtain a lentivirus of 5 , 5.5 ⁇ g of the lentiviral transfer vector (UCI-VD9 or UCI-VD35) prepared in Example 1, 3.7 ⁇ g of the packaging vector (UCI-VD6), and 1.8 ⁇ g of the envelope vector (UCI-VD11). Transfection was performed with lipofectamine 3000 transfection reagent. After transfection, the lentivirus produced in 293T cells was ejected out of the cell and existed in the cell culture medium in the form of virus particles. Accordingly, 48 or 72 hours after transfection, only the cell culture fluid was removed from the culture plate and concentrated 100 times with lenti-X concentration reagent (concentrator) to obtain lentivirus particles in the form of a pellet. Afterwards, 1 To select cell lines in which transduction was successful, cells expressing GFP were selected using a cell sorter (SH800S).
  • SH800S cell sorter
  • Pancreatic cancer cell lines (AsPC-1, Capan-2), breast cancer cell lines (HCC-1806, MDA-MB-231), stomach cancer cell lines (NCI-N87, AGS), lung cancer cell lines (NCI-H292), colon cancer cell lines (SNU- 1544, SW-620) and liver cancer cell lines (SK-HEP-1, Hep-G2) were purchased from the Korean Cell Line Bank (KCLB), and cell lines into which a luciferase reporter gene was introduced were used.
  • KCLB Korean Cell Line Bank
  • AsPC-1_luc_puro, Capan-2_luc_puro, HCC-1806_luc_puro, MDA-MB-231_luc_puro, NCI-N87_luc_puro, AGS_luc_puro, NCI-H292_luc_puro, SNU-1544_luc_puro and SW-620_luc_puro cell lines were cultured using RPMI-1640 medium.
  • EP- 1_luc_puro and Hep-G2_luc_puro were cultured using high-glucose Dulbecco's modified Eagle medium.
  • All media used for culturing cancer cell lines into which a luciferase reporter gene was introduced contained 100 ⁇ g/mL of streptomycin, 100 units/mL of penicillin, and 10% fetal bovine serum (FBS). ) and 4 ⁇ g/mL of puromycin were included. All cells were cultured at 37°C in an environment where CO 2 was maintained at 5% (95% air).
  • NK-92 cells MSLN CAR+p6 peptide
  • liver cancer cells were treated with an NK cell line genetically engineered to express a chimeric antigen receptor according to the present invention and a peptide that inhibits the TGF- ⁇ signaling pathway, the untreated group or the mock vector It was confirmed that the number of luciferase-expressing liver cancer cells was significantly reduced compared to the case of treating NK cells transfected with .
  • pancreatic cancer cells when pancreatic cancer cells are treated with NK cells transduced with a mock vector, the killing effect on gastric cancer cells is minimal compared to the untreated group, whereas the chimeric antigen receptor and TGF- according to the present invention
  • NK cell lines genetically engineered to express a peptide that inhibits the ⁇ signaling pathway were treated, it was confirmed that the death rate of pancreatic cancer cells was very high.
  • the NK cell line genetically engineered according to the present invention can target cancer cells expressing mesothelin due to the chimeric antigen receptor expressed in the cells, and can target cancer cells expressing mesothelin by the peptide additionally expressed in the cells. It can be seen that through inhibition of the TGF- ⁇ signaling pathway, the ability of cells to invade into microtumors is improved, and the killing effect on cancer cells is also significantly increased.
  • the present invention relates to immune effector cells genetically engineered to increase the therapeutic effect of diseases such as cancer by immunotherapy, and cell therapy products using the same.

Landscapes

  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des cellules ayant été génétiquement modifiées pour exprimer un peptide ou un fragment de celui-ci pouvant inhiber la voie de signalisation du facteur de croissance transformant-β (TGF-β), en conjonction avec un récepteur antigénique chimérique (CAR) ciblant la mésothéline (MSLN).
PCT/KR2023/018102 2022-11-11 2023-11-10 Cellules génétiquement modifiées et leur utilisation WO2024101962A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2022-0150906 2022-11-11
KR1020220150906A KR20240081499A (ko) 2022-11-11 유전자 조작된 세포 및 이의 용도

Publications (1)

Publication Number Publication Date
WO2024101962A1 true WO2024101962A1 (fr) 2024-05-16

Family

ID=91033328

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/018102 WO2024101962A1 (fr) 2022-11-11 2023-11-10 Cellules génétiquement modifiées et leur utilisation

Country Status (1)

Country Link
WO (1) WO2024101962A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180021137A (ko) * 2015-06-25 2018-02-28 아이셀 진 테라퓨틱스 엘엘씨 키메라 항원 수용체 (car), 조성물 및 이의 사용 방법
KR20180055824A (ko) * 2015-08-21 2018-05-25 카르스젠 테라퓨틱스 리미티드 항 메소텔린 완전 인간 항체 및 메소텔린을 타겟팅하는 면역효과 세포
KR20180118175A (ko) * 2016-03-04 2018-10-30 노파르티스 아게 다중 키메라 항원 수용체 (car) 분자를 발현하는 세포 및 그에 따른 용도
KR20210108290A (ko) * 2020-02-25 2021-09-02 (주)녹십자셀 메소텔린 특이적인 키메라 항원 수용체 및 이의 용도
KR20220091406A (ko) * 2020-12-23 2022-06-30 한국과학기술연구원 TGF-β 신호전달을 억제할 수 있는 신규한 펩타이드 및 이의 용도

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20180021137A (ko) * 2015-06-25 2018-02-28 아이셀 진 테라퓨틱스 엘엘씨 키메라 항원 수용체 (car), 조성물 및 이의 사용 방법
KR20180055824A (ko) * 2015-08-21 2018-05-25 카르스젠 테라퓨틱스 리미티드 항 메소텔린 완전 인간 항체 및 메소텔린을 타겟팅하는 면역효과 세포
KR20180118175A (ko) * 2016-03-04 2018-10-30 노파르티스 아게 다중 키메라 항원 수용체 (car) 분자를 발현하는 세포 및 그에 따른 용도
KR20210108290A (ko) * 2020-02-25 2021-09-02 (주)녹십자셀 메소텔린 특이적인 키메라 항원 수용체 및 이의 용도
KR20220091406A (ko) * 2020-12-23 2022-06-30 한국과학기술연구원 TGF-β 신호전달을 억제할 수 있는 신규한 펩타이드 및 이의 용도

Similar Documents

Publication Publication Date Title
RU2761377C2 (ru) Иммуноконъюгаты антитела к pd-1 с мутантом il-2 или с il-15
KR102572091B1 (ko) 항 gprc5d 항체, gprc5d 및 cd3에 결합하는 이중특이성 항원 결합 분자, 및 이들의 용도
WO2019225777A1 (fr) Anticorps anti-ror1 et son utilisation
WO2019098682A1 (fr) Anticorps anti-her2 ou fragment de liaison à l'antigène de celui-ci, et récepteur antigénique chimérique le comprenant
KR102527160B1 (ko) 예정 사멸 1(pd-1)에 대한 신규한 단일클론성 항체
WO2019225787A1 (fr) Anticorps anti-b7-h3 et son utilisation
WO2019112347A2 (fr) Anticorps ou son fragment de liaison à l'antigène pour reconnaître spécifiquement une malignité de cellule b, récepteur d'antigène chimérique le comprenant, et son utilisation
WO2018222019A1 (fr) Nouveaux anticorps anti-cd40 et leur utilisation
WO2021235696A1 (fr) Anticorps spécifique de cd22 et son utilisation
WO2020005003A1 (fr) Anticorps monoclonal se liant de manière spécifique à lag-3 et son utilisation
WO2019203600A1 (fr) Molécule de commutation et récepteur antigénique chimérique commutable
WO2023277361A1 (fr) Anticorps spécifiques de la mésothéline et leur utilisation
WO2021210939A1 (fr) Afficorps anti-her2 et récepteur antigénique chimérique commutable utilisant celui-ci en tant que molécule de commutation
KR102115236B1 (ko) 췌장 또는 담관계암 치료를 위한 키메라 항원 수용체
WO2019125070A1 (fr) Anticorps ou fragment de liaison à l'antigène de celui-ci reconnaissant spécifiquement des tumeurs malignes de lymphocytes b, récepteur antigénique chimérique le comprenant, et utilisations associées
WO2021060914A1 (fr) Récepteur antigénique chimérique anti-claudine-3
WO2024049161A1 (fr) Nouveau récepteur antigénique chimérique anti-pd-l1 et cellules immunitaires l'exprimant
WO2024101962A1 (fr) Cellules génétiquement modifiées et leur utilisation
WO2024101942A1 (fr) Cellules génétiquement modifiées et leur utilisation
WO2021235697A1 (fr) Anticorps spécifique de cd22 et son utilisation
WO2022124866A1 (fr) Anticorps anti-pd-1 et ses utilisations
WO2022124864A1 (fr) Anticorps anti-tigit et son utilisation
WO2021182929A1 (fr) Anticorps spécifique à bcma et récepteur antigénique chimérique
WO2021225399A1 (fr) Récepteurs antigéniques chimériques pour le traitement du cancer
WO2022170033A1 (fr) Car à base de ligands naturels modifiés : évolution dirigée du récepteur du stress nkp30