ANTI-GPC3 SINGLE-CHAIN ANTIBODY-CONTAINING CAR

Abstract
The present invention aims to provide a CAR that contains a single-chain antibody capable of specifically binding to GPC3 localized in the cell membrane and is useful for cancer immunotherapy using a CAR-T cell; and an immunocompetent cell expressing such a CAR. A gene encoding a CAR containing: a single-chain antibody that contains specific heavy-chain CDR1 to CDR3 and specific light-chain CDR1 to CDR3, as defined in claim 1, and specifically binds to a human-derived GPC3 polypeptide; a transmembrane region fused to a carboxyl terminus of the single-chain antibody; and an immunocompetent cell activation signaling transduction region fused to a carboxyl terminus of the transmembrane region is produced, and the gene is introduced into an immunocompetent cell. Thereby, a CAR-expressing immunocompetent cell having excellent cytotoxic activity on cancer cells and interferon gamma (IFN-γ) producibility can be produced.
Description
TECHNICAL FIELD

The present invention relates to a chimeric antigen receptor (which will be hereinafter referred to also as “CAR”) containing a single-chain antibody that specifically binds to a human GPC3 (Glypican-3)-derived polypeptide consisting of the amino acid sequence shown by SEQ ID No: 11, a transmembrane region fused to the carboxyl (C) terminus of the single-chain antibody, and an immunocompetent cell activation signaling transduction region fused to the C-terminus of the transmembrane region; an immunocompetent cell expressing the CAR; an anticancer agent containing the immunocompetent cell; a CAR gene encoding the CAR; and a vector containing the CAR gene.


BACKGROUND ART

A CAR is an artificial chimeric protein obtained by fusing a single-chain antibody that recognizes a cell surface antigen of a cancer cell to a signaling transduction region that induces activation of T cells. A large amount of CAR-expressing T cells (which will be hereinafter referred to also as “CAR-T cells”) that exhibit tumor reactivity can be produced by introducing a gene encoding the CAR into peripheral blood T cells (peripheral blood T lymphocytes) that do not exhibit tumor reactivity. The CAR-T cells that exhibit tumor reactivity have cytotoxic activity on cancer cells, independently of their interaction with major histocompatibility complex (MHC).


Clinical studies on cancer immunotherapy by administration of CAR-T cells, more specifically, therapy (Non Patent Document 1) in which a T cell is collected from a patient, a gene encoding a CAR is introduced into the T cell, and the cell is amplified and transferred into the patient again have been currently in progress all over the world. The results showing efficacy in hematopoietic malignancies such as leukemia and lymphoma have been obtained.


In recent years, various CAR-T cells have been studied. For example, there have been proposed: a pharmaceutical composition that contains a modified autologous human T cell containing a nucleic acid encoding a CAR consisting of a CD19 antigen-binding region, a transmembrane region, a 4-1BB co-stimulation signal region, and a CD3ζ signal region (Patent Document 1); an anti-tag chimeric antigen receptor (AT-CAR)-expressing T-cell population that is administered to a subject simultaneously with or separately from a formulation containing one or a plurality of tagged proteins configured to bind to cancer cells and is effective for one or a plurality of therapies by binding to the tagged proteins to induce cancer cell death (Patent Document 2); a cell that contains a nucleic acid encoding a chimeric antigen receptor containing an antigen-binding domain of human antibody 139, an extracellular hinge domain, a transmembrane domain, and an intracellular T cell-signaling domain (Patent Document 3); a cell that contains a nucleic acid sequence encoding a chimeric antigen receptor, wherein the chimeric antigen receptor contains an antigen-binding domain, a transmembrane domain, a co-stimulation signaling transduction region, and a CD3ζ signaling domain containing a specific amino acid sequence (Patent Document 4); a genetically engineered CD19-specific T cell that expresses and has a CD19-specific chimeric receptor on its cell surface membrane, wherein the chimeric receptor is composed of an intracellular signaling domain for the effector function of immune cells, at least one transmembrane domain, and at least one extracellular domain, and the extracellular domain contains the CD19-specific receptor (Patent Document 5); and a chimeric antigen receptor-expressing cell in which a nucleic acid encoding a chimeric antigen receptor containing the intracellular domain of a glucocorticoid-induced tumor necrosis factor receptor (GITR) as an intracellular domain is introduced (Patent Document 6). Further, the inventors have recently reported that immune-inducing effects and antitumor activities superior to conventional CAR-T cells are exerted by a CAR-T cell expressing interleukin 7 (IL-7) or chemokine ligand 19 (CCL19) (Patent Document 7).


Meanwhile, GPC3 is an extracellular matrix protein expressed in fetal tissues, especially in the liver and kidneys, and involved in organ formation. Although GPC3 is not expressed in adult tissues other than the placenta, it is expressed in various cancer tissues such as hepatocellular carcinoma, melanoma, clear ovarian cell carcinoma, and squamous cell lung cancer. As described above, GPC3 is a protein expressed in fetal tissues like proteins, such as α-fetoprotein (AFP) and carcinoembryonic antigen (CEA), and is therefore categorized as a fetal cancer antigen. That is, GPC3 is not expressed in normal tissue cells, but it has a characteristic of being specifically expressed in cancer cells and is therefore useful as a target molecule for cancer treatment, a tumor marker, and a diagnostic marker.


As a representative of existing anti-GPC3 antibodies, there is a monoclonal antibody 1G12, available from BioMosaics Inc. This antibody is an antibody obtained by immunizing a Balb/c mouse with an antigen (polypeptide of 70 residues on the C-terminal side of GPC3) designed to avoid the complex structure and localization of GPC3 to produce a hybridoma, followed by screening with the antigen. Further, antibodies GC33 and GC199 developed by a domestic pharmaceutical manufacturer are also monoclonal antibodies established based on the same concept and are antibodies obtained by using the C-terminal partial fragment of GPC3 as an antigen (Patent Document 8).


Recently, the inventors have found an anti-GPC3 antibody that recognizes an epitope different from those recognized by existing antibodies (such as GC33 and GC199) and can specifically bind to GPC3 localized in the cell membrane even in the state of a single-chain antibody (PCT/JP2018/257).


PRIOR ART DOCUMENTS
Patent Documents



  • Patent Document 1: U.S. Patent Application Publication No. 2014/0106449

  • Patent Document 2: Japanese unexamined Patent Application Publication (Translation of PCT Application) No. 2014-504294

  • Patent Document 3: Japanese unexamined Patent Application Publication (Translation of PCT Application) No. 2014-516510

  • Patent Document 4: Japanese unexamined Patent Application Publication (Translation of PCT Application) No. 2014-507118

  • Patent Document 5: Japanese unexamined Patent Application Publication No. 2011-004749

  • Patent Document 6: International Publication No. 2013/051718 Pamphlet

  • Patent Document 7: International Publication No. 2016/056228 Pamphlet

  • Patent Document 8: Japanese Patent No. 4011100



Non-Patent Documents



  • [Non-patent Document 1] Yozo Nakazawa, The Shinshu Medical Journal 61 (4): 197 to 203 (2013)



SUMMARY OF THE INVENTION
Object to be Solved by the Invention

It is an object of the present invention to provide: a CAR that contains a single-chain antibody capable of specifically binding to GPC3 localized in the cell membrane and is useful for cancer immunotherapy using CAR-T cells; and an immunocompetent cell expressing such a CAR.


Means to Solve the Object

The inventors are continuing diligent research to achieve the above object. In the process, they have found that a CAR-T cell with excellent cytotoxic activity on cancer cells and interferon gamma (IFN-γ) producibility can be produced by producing an anti-GPC3 single-chain antibody based on an anti-GPC3 antibody (GC33 antibody) developed by a domestic pharmaceutical manufacturer and introducing a vector that expresses a CAR to which the anti-GPC3 single-chain antibody is fused into a T cell, thereby accomplishing the present invention.


That is, the present invention is as follows.


[1] A chimeric antigen receptor (CAR) (which may be hereinafter referred to as “the CAR of the present invention”) comprising: a single-chain antibody that specifically binds to a human GPC3 (Glypican-3)-derived polypeptide consisting of the amino acid sequence shown by SEQ ID No: 11; a transmembrane region fused to a carboxyl terminus of the single-chain antibody; and an immunocompetent cell activation signaling transduction region fused to the carboxyl terminus of the transmembrane region, wherein the single-chain antibody comprises: a heavy chain complementarity determining region (CDR) 1 consisting of the amino acid sequence shown by SEQ ID No: 1, a heavy-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 2, and a heavy-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 3; and a light-chain CDR1 consisting of the amino acid sequence shown by SEQ ID No: 4, a light-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 5, and a light-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 6.


[2] The CAR according to [1] above, wherein the single-chain antibody comprises: a heavy-chain variable region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 7; and a light-chain variable region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 8.


[3] The CAR according to [1] or [2] above, wherein the single-chain antibody comprises: an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 12; or an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 13.


[4] The CAR according to any one of [1] to [3] above, wherein the transmembrane region and the immunocompetent cell activation signaling transduction region fused to the carboxyl terminus of the transmembrane region comprise an amino acid sequence having at least 80% or more sequence identity with an amino acid sequence shown by any one of SEQ ID Nos: 14 to 16.


[5] An immunocompetent cell (which may be hereinafter referred to as “the immunocompetent cell of the present invention”) expressing the CAR according to any one of [1] to [4] above.


[6] The immunocompetent cell according to [5] above, further expressing interleukin 7 (IL-7) and chemokine ligand 19 (CCL19).


[7] An anticancer agent (which may be hereinafter referred to as “the anticancer agent of the present invention”) comprising: the immunocompetent cell according to [5] or [6] above; and a pharmaceutically acceptable additive.


[8] A CAR gene (which may be hereinafter referred to as “the CAR gene of the present invention”) encoding the CAR according to any one of [1] to [4] above.


[9] A vector (which may be hereinafter referred to as “the CAR-expressing vector of the present invention”) comprising: a promoter; and the CAR gene encoding the CAR according to [8] above operably linked downstream of the promoter.


Further, examples of other embodiments of the present invention can include a method for producing an immunocompetent cell expressing the CAR of the present invention, the method including a step of introducing the CAR-expressing vector of the present invention into an immunocompetent cell; an immunocompetent cell expressing the CAR of the present invention produced by introducing the CAR-expressing vector of the present invention into an immunocompetent cell; and an immunocompetent cell containing the CAR gene of the present invention and expressing the CAR of the present invention (preferably, an immunocompetent cell further containing IL-7 gene and CCL19 gene and expressing the CAR of the present invention, IL-7, and CCL19).


Effect of the Invention

According to the present invention, when the CAR of the present invention is expressed on the cell membrane of an immunocompetent cell such as a T cell, the immunocompetent cell exhibits excellent cytotoxic activity on cancer cells and IFN-γ producibility. Therefore, the CAR of the present invention is useful for cancer immunotherapy using an immunocompetent cell.





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 includes graphs showing the analysis results for expression of CAR (horizontal axis) and CD8 (vertical axis) in T cells (FIG. 1A) and CAR-T cells (FIG. 1B) with a flow cytometer. The regions “Q1”, “Q2”, “Q3”, and “Q4” surrounded by rectangles in the figures respectively represent a “CAR-negative CD8-positive T-cell group”, a “CAR-positive CD8-positive T-cell group”, a “CAR-positive CD8-negative T-cell group”, and a “CAR-negative CD8-negative T-cell group”, and the ratio (%) of the number of cells in each of the four cell groups with respect to the total number of cells is shown in the each region.



FIG. 2 shows the analysis results of the number of CD45-negative cells (corresponding to the remaining Sk-HEP-1 GPC3 cells) (FIG. 2A), and the ratio of the CD45-negative cells with respect to the total cells (FIG. 2B), when Sk-HEP-1 cells (“mock” in the figures) and Sk-HEP-1 GPC3 cells (“GPC3” in the figures) are cultured in the presence of a T-cell group (“T cells” in the figures) or a T-cell group containing anti-GPC3 scFv CAR-T cells (“CAR-T cells” in the figures).



FIG. 3 is a graph showing the analysis results for the concentration of IFN-γ produced in a culture medium when Sk-HEP-1 cells (“mock” in the figures) and Sk-HEP-1 GPC3 cells (“GPC3” in the figures) are cultured in the presence of a T-cell group (“T cells” in the figures) or a T-cell group containing anti-GPC3 scFv CAR-T cells (“CAR-T cells” in the figures).



FIG. 4 includes graphs showing the analysis results for the concentration of IL-7 (FIG. 4A) and CCL19 (FIG. 4B) produced in a culture medium, when Sk-HEP-1 cells (“mock” in the figures) and Sk-HEP-1 GPC3 cells (“GPC3” in the figures) are cultured in the presence of a T-cell group (“T cells” in the figures) or a T-cell group containing anti-GPC3 scFv CAR-T cells (“CAR-T cells” in the figures).





MODE OF CARRYING OUT THE INVENTION

The CAR of the present invention is a polypeptide containing at least i)-iii) below: i) a single-chain antibody (single-chain Fv; scFv) (which may be hereinafter referred to as “the single-chain antibody of the present invention”) that contains heavy (H)-chain complementarity determining region (CDR) 1 consisting of the amino acid sequence shown by SEQ ID No: 1, H-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 2, and H-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 3, and light (L)-chain CDR1 consisting of the amino acid sequence shown by SEQ ID No: 4, L-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 5, and L-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 6, and specifically binds to a human GPC3-derived polypeptide consisting of the amino acid sequence shown by SEQ ID No: 11 (corresponding to the polypeptide consisting of glycine at position 537 to lysine at position 563 of human GPC3); ii) a transmembrane region fused to the C-terminus of the single-chain antibody of the present invention; and iii) an immunocompetent cell activation signaling transduction region fused to the C-terminus of the transmembrane region, wherein the single-chain antibody of the present invention and the transmembrane region, or the transmembrane region and the immunocompetent cell activation signaling transduction region may be fused to each other via a peptide linker or IgG4 hinge region. Further, the single-chain antibody of the present invention generally contains H-chain variable (V) regions containing the H-chain CDR1 to CDR3, L-chain V-regions containing the L-chain CDR1 to CDR3, and the H-chain V-regions and the L-chain V-regions are generally bound to each other via a peptide linker.


Examples of the length of the peptide linker can include 1 to 100 amino acid residues, preferably 10 to 50 amino acid residues. Further, specific examples of the peptide linker in the antibody of the present invention can include three continuous amino acid sequences each consisting of one to four glycines (G) and one serine (S).


In the V-region of the single-chain antibody of the present invention, framework regions (FR) are included as regions other than CDR1 to CDR3. Among such FRs, examples of H-chain FRs can include H-chain FR1 linked to the N-terminus of the H-chain CDR1, H-chain FR2 linked to the C-terminus of the H-chain CDR1 (N-terminus of the H-chain CDR2), H-chain FR3 linked to the C-terminus of the H-chain CDR2 (N-terminus of the H-chain CDR3), and H-chain FR4 linked to the C-terminus of the H-chain CDR3. Among the FRs, examples of L-chain FRs can include L-chain FR1 linked to the N-terminus of the L-chain CDR1, L-chain FR2 linked to the C-terminus of the L-chain CDR1 (N-terminus of the L-chain CDR2), L-chain FR3 linked to the C-terminus of the L-chain CDR2 (N-terminus of the L-chain CDR3), and L-chain FR4 linked to the C-terminus of the L-chain CDR3.


Examples of the H-chain FR1 can specifically include a polypeptide consisting of the amino acid residues at positions 1 to 30 in the amino acid sequence shown by SEQ ID No: 7, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the H-chain FR2 can specifically include a polypeptide consisting of the amino acid residues at positions 36 to 49 in the amino acid sequence shown by SEQ ID No: 7, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the H-chain FR3 can specifically include a polypeptide consisting of the amino acid residues at positions 67 to 98 in the amino acid sequence shown by SEQ ID No: 7, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the H-chain FR4 can specifically include a polypeptide consisting of the amino acid residues at positions 105 to 114 in the amino acid sequence shown by SEQ ID No: 7, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the L-chain FR1 can specifically include a polypeptide consisting of the amino acid residues at positions 1 to 23 in the amino acid sequence shown by SEQ ID No: 8, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the L-chain FR2 can specifically include a polypeptide consisting of the amino acid residues at positions 40 to 54 in the amino acid sequence shown by SEQ ID No: 8, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the L-chain FR3 can specifically include a polypeptide consisting of the amino acid residues at positions 62 to 93 in the amino acid sequence shown by SEQ ID No: 8, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


Examples of the L-chain FR4 can specifically include a polypeptide consisting of the amino acid residues at positions 103 to 113 in the amino acid sequence shown by SEQ ID No: 8, or a polypeptide consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of the polypeptide.


As the FRs of the single-chain antibody of the present invention, FRs of known human antibodies are preferable. Examples of the “FRs of known human antibodies” can include FRs of human antibodies registered in known sequence databases such as GenBank, and FRs selected from consensus sequences derived from each subgroup of human antibodies (Human Most Homologous Consensus Sequence; Kabat, E. A. et al., Sequences of Proteins of Immunological Interest, US Dept. Health and Human Services, 1991).


The H-chain CDR1 in the single-chain antibody of the present invention is generally located at positions H31 to 35 according to the numbering by Kabat (see Literature “Kabat, E. A. et al., (1991) NIH Publication No. 91-3242, sequences of proteins of immunological interest”). Further, the H-chain CDR2 in the single-chain antibody of the present invention is generally located at positions H50 to 52, H52A, and H53 to 65 according to the numbering by Kabat. Further, the H-chain CDR3 in the single-chain antibody of the present invention is generally located at positions H95 to 100, H100A, H100B, H101, and H102 according to the numbering by Kabat. Further, the L-chain CDR1 in the single-chain antibody of the present invention is generally located at positions L24 to 34 according to the numbering by Kabat. Further, the L-chain CDR2 in the single-chain antibody of the present invention is generally located at positions L50 to 56 according to the numbering by Kabat. Further, the L-chain CDR3 in the single-chain antibody of the present invention is generally located at positions L89 to 97 according to the numbering by Kabat.


Examples of the H-chain V-region in the single-chain antibody of the present invention can specifically include a H-chain V-region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 7. Examples of the L-chain V-region in the single-chain antibody of the present invention can specifically include a L-chain V-region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 8.


Examples of the single-chain antibody of the present invention can specifically include those having the effects demonstrated in Examples described below, that is, a single-chain antibody containing an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 12, and a single-chain antibody containing an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 13.


The transmembrane region may be any peptide capable of penetrating through cell membranes, and examples thereof can include all or a part of a transmembrane region derived from CD8, α or β chain of a T-cell receptor, CD3, CD28, CD3E, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, CD154, EGFR (Epidermal Growth Factor Receptor), or GITR. Examples thereof can specifically include a human CD8 transmembrane region consisting of the amino acid residues at positions 1 to 83 in the amino acid sequence shown by SEQ ID No: 14. Further, the transmembrane region may be those in which 1 to 10 amino acid residues, preferably 6 to 7 amino acid residues, on the C-terminal side of the peptide capable of penetrating through cell membranes are deleted. Examples thereof can include variant 1 of the human CD8 transmembrane region consisting of the amino acid residues at positions 1 to 77 in the amino acid sequence shown by SEQ ID No: 14, and variant 2 of the human CD8 transmembrane region consisting of the amino acid residues at positions 1 to 76 in the amino acid sequence shown by SEQ ID No: 14.


The immunocompetent cell activation signaling transduction region may be any region capable of transmitting signals into an immunocompetent cell when the single-chain antibody of the present invention is bound to human GPC3. Those containing at least one or more selected from polypeptides in the intracellular regions of CD28, 4-1BB (CD137), GITR, CD27, OX40, HVEM, CD3ζ, and Fc Receptor-associated γ-chain are preferable, and those containing three polypeptides in the intracellular regions of CD28, 4-1BB, and CD3ζ are more preferable. Examples of the polypeptide in the intracellular region of CD28 can specifically include a polypeptide in the intracellular region of human CD28 consisting of the amino acid residues at positions 85 to 124 in the amino acid sequence shown by SEQ ID No: 14. Examples of the “polypeptide in the intracellular region of 4-1BB” can specifically include a polypeptide in the intracellular region of human 4-1BB consisting of the amino acid residues at positions 125 to 170 in the amino acid sequence shown by SEQ ID No: 14. Examples of the polypeptide in the intracellular region of CD3 ζ can specifically include a polypeptide in the intracellular region of human CD3 ζ consisting of the amino acid residues at positions 172 to 283 in the amino acid sequence shown by SEQ ID No: 14.


Here, arginine (Arg) at position 84 in the amino acid sequence shown by SEQ ID No: 14, arginine at position 78 in the amino acid sequence shown by SEQ ID No: 15, and arginine at position 77 in the amino acid sequence shown by SEQ ID No: 16 are consensus sequences of the polypeptides in the transmembrane region derived from human CD8 and the polypeptides in the intracellular region of human CD28. Further, leucine (Leu) at position 171 in the amino acid sequence shown by SEQ ID No: 14, leucine at position 165 in the amino acid sequence shown by SEQ ID No: 15, and leucine at position 164 in the amino acid sequence shown by SEQ ID No: 16 are consensus sequences of the polypeptides in the intracellular region of human 4-1BB and the polypeptides in the intracellular region of human CD3.


In this description, the “immunocompetent cell” means a cell responsible for the immune function in a living body (preferably, a cell separated from the living body). Examples of the immunocompetent cell can include a lymphocytic cell such as a T cell, a natural killer cell (NK cell), and a B cell; an antigen-presenting cell such as a monocyte, a macrophage, and a dendritic cell; and a granulocyte such as a neutrophil, an eosinophil, a basophil, and a mast cell. Examples thereof can specifically include a T cell derived from a mammal such as a human, a dog, a cat, a pig, and a mouse. Among them, a human-derived T cell can be mentioned as a suitable example.


Further, the T cell can be obtained, for example, by separating a cell population containing an immunocompetent cell from a body fluid such as blood and bone marrow fluid, a tissue such as spleen, thymus, and lymph nodes, or a cancer tissue such as a primary tumor, a metastatic tumor, and cancer ascites.


For increasing the proportion of the T cell contained in the cell population, the T cell can be obtained, for example, also by further isolating or purifying the cell population separated above, as required, according to a conventional method. Further, a T cell produced from an ES cell (embryonic stem cell) or an iPS cell (induced pluripotent stem cell) may be used. Examples of the T cell can include an alpha/beta T cell, a gamma/delta T cell, a CD8+ T cell, a CD4+ T cell, a tumor-infiltrating T cell, a memory T cell, a naive T cell, and an NK T cell. The origin of the immunocompetent cell and the administration target is the same or different. Further, in the case where the administration target is a human, the immunocompetent cell may be an autologous cell collected from the patient who is the administration target or an allogeneic cell collected from another person. That is, the donor and recipient may or may not match, but they preferably match.


Examples of the administration target can suitably include a mammal or a mammalian cell. Among such mammals, examples thereof can include more suitably a human, a mouse, a dog, a rat, a guinea pig, a rabbit, a sheep, a pig, a cow, a horse, a cat, a monkey, and a chimpanzee, particularly suitably a human.


The CAR of the present invention is preferably used in cancer treatment to express ex vivo on the cell surface of an immunocompetent cell collected from a cancer patient. In the case of using a T cell as the immunocompetent cell, examples of the peptide consisting of the transmembrane region in the CAR of the present invention and the immunocompetent cell activation signaling transduction region fused to the C-terminus of the transmembrane region can specifically include a peptide containing an amino acid sequence having at least 80% or more sequence identity with an amino acid sequence shown by any one of SEQ ID Nos: 14 to 16. Further, examples of the CAR of the present invention can specifically include those containing the single-chain antibody of the present invention and a peptide, linked to the C-terminal side of the single-chain antibody, containing an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of SEQ ID No: 14, those containing the single-chain antibody of the present invention and a peptide, linked to the C-terminal side of the single-chain antibody, containing an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of SEQ ID No: 15, and those containing the single-chain antibody of the present invention and a peptide, linked to the C-terminal side of the single-chain antibody, containing an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence of SEQ ID No: 16.


The immunocompetent cell of the present invention may be any immunocompetent cell that expresses the CAR of the present invention, and since the CAR of the present invention usually does not exist naturally, it is an immunocompetent cell that expresses an exogenous CAR rather than an endogenous CAR. The immunocompetent cell of the present invention further preferably express IL-7 and/or CCL19. In the case where the immunocompetent cell is a cell that does not express IL-7 and/or CCL19 (such as a T cell), or the immunocompetent cell is a cell other than the T cell and exhibits a low expression level of IL-7 and/or CCL19, the immunocompetent cell of the present invention preferably expresses exogenous IL-7 and/or CCL19. Here, the immunocompetent cell expressing exogenous IL-7 and/or CCL19 can be obtained by introducing exogenous IL-7 gene and/or CCL19 gene (preferably, exogenous IL-7 gene and/or CCL19 gene operably linked downstream of the promoter) into an immunocompetent cell. The immunocompetent cell obtained by introducing the exogenous IL-7 gene and/or CCL19 gene contains the exogenous IL-7 gene and/or CCL19 gene incorporated into the genome or not incorporated into the genome (for example, in an episomal state) within the immunocompetent cell. The immunocompetent cell of the present invention also includes a cell culture that is obtained by culturing the immunocompetent cell of the present invention and expresses the CAR of the present invention.


The immunocompetent cell of the present invention can be produced by introducing the CAR-expressing vector of the present invention into the immunocompetent cell. The introduction method may be any method for introducing DNA into an immunocompetent cell. Examples thereof can include methods such as the electroporation method (Cytotechnology, 3, 133 (1990)), the calcium phosphate method (Japanese unexamined Patent Application Publication No. 2-227075), the lipofection method (Proc. Natl. Acad. Sci. U.S.A, 84, 7413 (1987)), and a virus infection method. Examples of the virus infection method can include a method in which a CAR-expressing vector (International Publication No. 2016/056228 Pamphlet) and a packaging plasmid are transfected into a packaging cell such as a GP2-293 cell (available from Takara Bio Inc.), a Plat-GP cell (available from Cosmo Bio Co., Ltd.), a PG13 cell (ATCC CRL-10686), and a PA317 cell (ATCC CRL-9078) to produce a recombinant virus, and a T cell is infected with the recombinant virus. The immunocompetent cell (the immunocompetent cell of the present invention) obtained by introducing the CAR-expressing vector of the present invention contains the CAR gene of the present invention incorporated into the genome or not incorporated into the genome within the immunocompetent cell.


Further, the immunocompetent cell of the present invention may be produced also by incorporating a nucleotide encoding the CAR of the present invention into the genome of the cell using a known gene-editing technique so as to be able to express under the control of an appropriate promoter. Examples of the known gene-editing technique can include a technique using an endonuclease such as zinc finger nuclease, TALEN (transcriptional activation-like effector nuclease), and CRISPR (Clustered Regularly Interspaced Short Palindromic Repeat)-Cas system.


The anticancer agent of the present invention may be any composition the application of which is specified “for anti-cancer (that is, for suppressing the growth of cancer)” and which contains the immunocompetent cell of the present invention and a pharmaceutically acceptable additive. Examples of the pharmaceutically acceptable additive can include normal saline, buffer normal saline, cell culture medium, dextrose, water for injection, glycerol, ethanol, a stabilizer, a solubilizer, a surfactant, a buffer, a preservative, an isotonic agent, a filler, and a lubricant. Further, the additive may be a simple substance consisting of one additive or a mixture containing a plurality (at least two) of additives.


The anticancer agent of the present invention can be administered to a test subject (cancer patient) in need of suppression of cancer growth (in other words, cancer treatment) using a method known to those skilled in the art. Examples of the administration method can include a method of intravenous, intratumor, intradermal, subcutaneous, intramuscular, intraperitoneal, intraarterial, intramedullary, intracardiac, intraarticular, intrasynovial, intracranial, intrathecal, or subarachnoid (spinal fluid) injection and an administration method to a tumor site using a catheter.


The number of immunocompetent cells of the present invention to be contained in the anticancer agent of the present invention can be appropriately selected depending on the conditions such as the type, location, and severity of cancer, and the age and body weight and condition of the subject to be treated. For example, the number of cells is 1×104 to 1×1010, preferably 1×105 to 1×109, more preferably 5×106 to 5×108 per administration.


The number of times of administration of the anticancer agent of the present invention is, for example, 4 times, 3 times, twice, or once per day. Further, the administration interval of the anticancer agent of the present invention is, for example, every other day, every 3 days, every 4 days, every 5 days, every 6 days, every 7 days, every 8 days, every 9 days, every 10 days, or every other month.


Examples of the cancer for which the anticancer agent of the present invention is administered can include colorectal cancer (colon cancer or rectal cancer); gastric cancer; liver cancer; brain tumor; lung cancer (adenocarcinoma, squamous-cell carcinoma, adenosquamous carcinoma, undifferentiated cancer, large cell carcinoma, or small cell carcinoma); esophageal cancer; duodenal cancer; small intestine cancer; skin cancer; breast cancer; prostate cancer; bladder cancer; vaginal cancer; cervical cancer; endometrial cancer; renal cancer; pancreatic cancer; spleen cancer; tracheal cancer; bronchial cancer; head and neck cancer; gallbladder cancer; cholangiocarcinoma; testicular cancer; ovarian cancer; cancer in bone tissues, cartilage tissues, adipose tissues, muscle tissues, nerve tissues, vascular tissues, or hematopoietic tissues (specifically, a sarcoma such as chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, or soft tissue sarcoma; a blastoma such as hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, pleuropulmonary blastoma, or retinoblastoma; embryonic cell tumor; lymphoma; or leukemia).


The anticancer agent of the present invention can be used in combination with another anticancer agent. Examples of the other anticancer agent can include an alkylating drug such as cyclophosphamide, bendamustine, iosfamide, or dacarbazine, an antimetabolite such as pentostatin, fludarabine, cladribine, methotrexate, 5-fluorouracil, 6-mercaptopurine, or enocitabin, a molecular targeted drug such as rituximab, cetuximab, or trastuzumab, a kinase inhibitor such as imatinib, gefitinib, erlotinib, afatinib, dasatinib, sunitinib, or trametinib, a proteasome inhibitor such as bortezomib, a calcineurin inhibitor such as cyclosporine or tacrolimus, an antitumor antibiotic such as idarubicin or doxorubicin mitomycin C, a plant alkaloid such as irinotecan or etoposide, a platinum preparation such as cisplatin, oxaliplatin, or carboplatin, a hormone therapeutic drug such as tamoxifen or bicalutamide, or an immunoregulatory drug such as interferon, nivolumab, or pembrolizumab.


Examples of the method of “using the anticancer agent of the present invention in combination with another anticancer agent” can include a treatment method using another anticancer agent and then the anticancer agent of the present invention, a method using the anticancer agent of the present invention and another anticancer agent at the same time, or a treatment method using the anticancer agent of the present invention and then another anticancer agent. Further, in the case of using the anticancer agent of the present invention in combination with another anticancer agent, the therapeutic effect on cancer is further improved, and each side effect can be reduced by reducing the number of doses or dose.


The CAR gene of the present invention is not specifically limited as long as it is an antibody gene (nucleotide) encoding a polypeptide containing at least i)-iii) below: i) the single-chain antibody of the present invention, ii) a transmembrane region fused to the C-terminus of the single-chain antibody of the present invention, and iii) an immunocompetent cell activation signaling transduction region fused to the C-terminus of the transmembrane region. Examples of the antibody gene encoding the single-chain antibody of the present invention can specifically include a H-chain V-region gene consisting of a nucleotide sequence having at least 80% or more sequence identity with the nucleotide sequence shown by SEQ ID No: 9 (gene encoding a H-chain V-region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 7), and a L-chain V-region gene consisting of a nucleotide sequence having at least 80% or more sequence identity with the nucleotide sequence shown by SEQ ID No: 10 (gene encoding a L-chain V-region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 8).


In this description, the term “identity” means the degree of approximation of the polypeptide or polynucleotide sequence (which is determined depending on matching between the query sequence and another sequence, preferably, of the same type (nucleic acid or protein sequence)). Examples of a preferable method of computer program for calculating and determining the “identity” can include GCG BLAST (Basic Local Alignment Search Tool) (Altschul et al., J. Mol. Biol. 1990, 215: 403-410; Altschul et al., Nucleic Acids Res. 1997, 25: 3389-3402; Devereux et al., Nucleic Acid Res. 1984, 12: 387), and BLASTN 2.0 (Gish W., http://blast.Wustl.edu, 1996-2002), and FASTA (Pearson and Lipman, Proc. Natl. Acad. Sci. USA 1988, 85: 2444-2448), and GCG GelMerge to determine and align the longest overlapping pair of contigs (Wibur and Lipman, SIAM J. Appl. Math. 1984, 44: 557-567; Needleman and Wunsch, J. Mol. Biol. 1970, 48: 443-453).


In this description, the “at least 80% or more identity” means that the identity is 80% or more, preferably 85% or more, more preferably 88% or more, further preferably 90% or more, furthermore more preferably 93% or more, particularly preferably 95% or more, still more preferably 98% or more, most preferably 100%.


In this description, the “amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: X” is, in other words, “an amino acid sequence in which zero, one, or some amino acid residues are deleted, substituted, inserted, and/or added in the amino acid sequence shown by SEQ ID No: X” and has a function equivalent to the amino acid sequence shown by SEQ ID No: X. Here, the “amino acid sequence in which one or some amino acid residues are deleted, substituted, inserted, and/or added”, for example, means an amino acid sequence in which the number of the amino acid residues deleted, substituted, inserted, and/or added is in the range of 1 to 30, preferably in the range of 1 to 20, more preferably in the range of 1 to 15, further preferably in the range of 1 to 10, further preferably in the range of 1 to 5, further preferably in the range of 1 to 3, further preferably in the range of 1 to 2. Mutation treatment of these amino acid residues can be performed by any method known to those skilled in the art, such as chemical synthesis, genetic engineering, and mutagenesis.


In this description, the “gene operably linked downstream of the promoter” means that the promoter DNA and the gene DNA are functionally linked so that the promoter can initiate gene transcription.


The promoter in the CAR-expressing vector of the present invention may be any region that initiates transcription of mRNA encoded by the CAR gene of the present invention located downstream of the promoter, and the promoter generally includes the transcription start site (TSS).


The type of the promoter or vector in the CAR-expressing vector of the present invention can be appropriately selected, corresponding to the type of the host cell (immunocompetent cell) to be introduced. Here, examples of the promoter can include cytomegalovirus (CMV) IE (immediate early) gene promoter, SV40 early promoter, retrovirus promoter, metallothionein promoter, heat shock promoter, SRα promoter, NFAT promoter, and HIF promoter. Further, examples of the vector can include a retroviral vector such as pMSGV vector (Tamada k et al., Clin Cancer Res 18: 6436-6445 (2002)) or pMSCV vector (available from Takara Bio Inc.), or those derived from such a vector.


The CAR-expressing vector of the present invention may further contain a nucleotide sequence of an enhancer region or ribosome-binding site (RBS), for further enhancing the gene expression efficiency, drug-resistant gene (such as spectinomycin-resistant gene, chloramphenicol-resistant gene, tetracycline-resistant gene, kanamycin-resistant gene, ampicillin-resistant gene, puromycin-resistant gene, hygromycin-resistant gene, blasticidin-resistant gene, and genetisin-resistant gene) corresponding to the type of the immunocompetent cell of the present invention, for screening the immunocompetent cell of the present invention. Such an enhancer region is generally arranged upstream of the promoter, and RBS is generally arranged between the promoter and the CAR gene of the present invention. In the nucleotide sequence of the CAR gene of the present invention in the CAR-expressing vector of the present invention, the codon sequence may be optimized for the expressing immunocompetent cell of the present invention. The CAR of the present invention, the CAR-expressing vector of the present invention, and the CAR gene of the present invention can be produced by known methods using the gene recombination technology.


The CAR-expressing vector of the present invention may further contain a gene other than the CAR gene of the present invention, such as IL-7 gene, CCL19 gene, or a gene encoding a polypeptide with apoptotic action (suicide gene) (preferably, IL-7 gene, CCL19 gene, or a suicide gene operably linked downstream of the promoter). Examples of the suicide gene can include a gene encoding herpes simplex virus thymidine kinase [HSV-TK] or a gene encoding inducible caspase 9. For example, in the case where cancer cells disappear in the course of cancer treatment, the CAR-expressing vector of the present invention containing a suicide gene enables the immunocompetent cells of the present invention in the body that are no longer needed to be killed by administering a drug that activates the functions of the polypeptide encoded by the suicide gene (which is ganciclovir in the case of HSV-TK or AP1903 that is chemical induction of dimerization: CID in the case of inducible caspase 9, for example) (Cooper LJ., et. al. Cytotherapy. 2006; 8 (2): 105-17, Jensen M. C. et. al. Biol Blood Marrow Transplant. 2010 September; 16 (9): 1245-56, Jones BS. Front Pharmacol. 2014 Nov. 27; 5: 254, Minagawa K., Pharmaceuticals (Basel). 2015 May 8; 8(2): 230-49, and Bole-Richard E., Front Pharmacol. 2015 Aug. 25; 6: 174).


References such as academic literatures, patents, and patent applications cited herein are hereby incorporated by reference in their entirety to the same extent as they are specifically described in this description.


Hereinafter, the present invention will be described more specifically according to Examples, but the technical range of the present invention is not limited to these examples. In Examples below, GP2-293 packaging cell line (available from Takara Bio Inc.) was cultured in a CO2 incubator (under the condition of 37° C.) in the presence of DMEM (Dulbecco's Modified Eagle's Medium) culture medium containing 10% fetal bovine serum (FBS), penicillin G, and streptomycin sulfate. Further, the human T cells were cultured in a CO2 incubator (under the condition of 37° C.) in the presence of GT-T551 (available from Takara Bio Inc.) culture medium containing penicillin G (100 Units/mL) and others.


EXAMPLES
1. Preparation of Materials

[Production of CAR, hIL-7, hCCL19, and HSV-TK-Expressing Retroviral Vector]


Based on the amino acid sequences of a H-chain V-region (VH) and a L-chain V-region (VL) of an anti-GPC3 antibody (GC33 antibody) developed by a domestic pharmaceutical manufacturer, two types of anti-GPC3 human scFvs (VH-linker-VL [polypeptide consisting of the amino acid sequence of SEQ ID No: 12] and VL-linker-VH [polypeptide consisting of the amino acid sequence of SEQ ID No: 13]) were designed (see Table 1). A linker consisting of 15 amino acid residues obtained by repeating a polypeptide “GGGGS” three times was used therefor.









TABLE 1







SEQ ID No: 12: Anti-GPC3 human scFvs



QVQLQQSGAELVRPGASVKLSCKASGYTFTDYEMHWVKQTPVHGLKWI







GALDPKTGDTAYSQKFKGKATLTADKSSSTAYMELRSLTSEDSAVYYC








embedded image





DQASISCRSSQSLVHSNGNTYLHWYLQKPGQSPKLLIYKVSNRFSGVP







DRFSGSGSGTDFTLKISRVEAEDLGVYFCSQNTHVPPTFGSGTKLEIK







R






SEQ ID No: 13: Anti-GPC3 human scFvs (Ver. 2)



DVVMTQTPLSLPVSLGDQASISCRSSQSLVHSNGNTYLHWYLQKPGQS







PKLLIYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVYFCSQN








embedded image





SVKLSCKASGYTFTDYEMHWVKQTPVHGLKWIGALDPKTGDTAYSQKF







KGKATLTADKSSSTAYMELRSLTSEDSAVYYCTRFYSYTYWGQGTLVT







VS










In the table, the double squares each represent a linker, the single underlines each represent VH, and the double underlines each represent VL.


Of the two types of anti-GPC3 human scFvs designed, VH-linker-VL (polypeptide consisting of the amino acid sequence of SEQ ID No: 12) is selected as an example, and a fusion peptide consisting of a human CD8-derived transmembrane region and a human CD28/4-1BB/CD3zeta-derived immunocompetent cell activation signaling transduction region (the amino acid residues at positions 243 to 525 in the amino acid sequence of SEQ ID No: 17) was linked downstream thereof, to design an anti-GPC3 scFv CAR consisting of the amino acid sequence of SEQ ID No: 17 (see Table 2).









TABLE 2







SEQ ID No: 17: CAR of scFvs human antibody of


GC33 antibody




embedded image






embedded image






embedded image






embedded image






embedded image





TRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNRSKRSRLLHSD







YMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRFSVVKRGRKKLLYZFKQ







PFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQ







LYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKM







AEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR










In the table, the single underline represents an anti-GPC3 human scFv, the double underline represents a fusion peptide consisting of a human CD8-derived transmembrane region and a human CD28/4-1BB/CD3zeta-derived immunocompetent cell activation signaling transduction region.


Further, a human immunoglobulin H-chain-derived signal sequence (amino acid residues at positions 1 to 19 in the amino acid sequence of SEQ ID No: 18) was linked upstream of the anti-GPC3 scFv CAR designed, and a 2A self-cleaving peptide (amino acid residues at positions 551 to 575 in the amino acid sequence of SEQ ID No: 18), human (h)IL-7 (amino acid residues at positions 577 to 753 in the amino acid sequence of SEQ ID No: 18), a 2A self-cleaving peptide (amino acid residues at positions 754 to 778 in the amino acid sequence of SEQ ID No: 18), hCCL19 (amino acid residues at positions 779 to 876 in the amino acid sequence of SEQ ID No: 18), a 2A self-cleaving peptide (amino acid residues at positions 877 to 901 in the amino acid sequence of SEQ ID No: 18), and herpes simplex virus thymidine kinase (HSV-TK) (amino acid residues at positions 902 to 1277 in the amino acid sequence of SEQ ID No: 18) were sequentially linked downstream thereof, to design a fusion polypeptide consisting of the amino acid sequence of SEQ ID No: 18 (anti-GPC3 scFv CAR+hIL-7+hCCL19+HSV-TK) (see Table 3).









TABLE 3







SEQ ID No: 18: CAR+hIL-7+hCCL19+HSV-TK




embedded image





FTDYEMHWVKQTPVHGLKWIGALDPKTGDTAYSQKFKGKATLTADKSS







STAYMELRSLTSEDSAVYYCTRFYSYTYWGQGTLVTVSGGGGSGGGGS







GGGGSDVVMTQTPLSLPVSLGDQASISGRSSQSLVHSNGNTYLHWYLQ







KPGQSPKLLIYKVSNRFSGVPDRFSGSGSGTDFTLKISRVEAEDLGVY







FCSQNTHVPPTFGSGTKLEIKRAAAFVPVFLPAKPTTTPAPRPPTPAP







TIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYZWAPLAGTCGVLLL







SLVITLYCNHRNRSKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDF







AAYRSRFSVVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEG







GCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPE







MGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQG








embedded image





MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKDGKQYESVLMVSIDQL







LDSMKEIGSNCLNNEFNFFKRHICDANKEGMFLFRAARKLRQFLKMNS







TGDFDLHLLKVSEGTTILLNCTGQVKGRKPAALGEAQPTKSLEENKSL








embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image











In the table, the single square represents a human immunoglobulin H-chain-derived signal sequence, the double squares each represent a 2A self-cleaving peptide, the single underline represents a CAR, the double underline represents hIL-7, the broken underline represents hCCL19, and the wavy underline represents HSV-TK.


Then, a gene DNA consisting of a nucleotide sequence encoding the fusion polypeptide (CAR+hIL-7+hCCL19+HSV-TK) and optimized for human codon (polynucleotide consisting of the nucleotide sequence of SEQ ID No: 19, see Table 4) was incorporated into a pMSGV retroviral vector (Tamada k et al., Clin Cancer Res 18: 6436-6445 (2002)), to produce a retroviral vector for expression of CAR, hIL-7, hCCL19, and HSV-TK (which will be hereinafter referred to as “retroviral vector for expression of anti-GPC3 scFv CAR, etc.”) (see International Publication No. 2016/056228 Pamphlet).









TABLE 4







SEQ ID No: 19: ″CAR+hIL-7+hCCL19+HSV-TK″ gene




embedded image






embedded image





AGACCTGGCGCCAGCGTGAAACTGAGCTGCAAGGCCAGCGGCTACACC







TTCACCGACTACGAGATGCACTGGGTGAAGCAGACACCCGTGCACGGC







CTGAAGTGGATCGGAGCCCTGGACCCCAAGACCGGAGACACCGCCTAC







AGCCAGAAGTTTAAGGGCAAGGCCACCCTCACCGCCGATAAGAGCTCC







AGCACCGCCTACATGGAGCTGAGGAGCCTGACCTCCGAGGACAGCGCC







GTGTACTATTGCACCAGGTTCTACAGCTACACCTACTGGGGCCAGGGA







ACACTGGTGACCGTCTCCGGCGGCGGCGGCAGCGGCGGCGGAGGCAGC






GGCGGCGGCGGCAGCGATGTGGTGATGACACAGACCCCTCTGAGCCTG






CCTGTGAGCCTGGGAGACCAGGCCAGCATCAGCTGCAGGAGCAGCCAG







AGCCTGGTGCACAGCAACGGCAACACATACCTCCACTGGTACCTGCAG







AAGCCTGGCCAGAGCCCCAAGCTGCTGATCTACAAGGTGAGCAACAGG







TTCTCCGGCGTGCCCGATAGATTCAGCGGCTCCGGCAGCGGCACCGAC







TTTACCCTGAAGATCTCCAGAGTGGAGGCCGAGGACCTGGGCGTGTAC







TTTTGCAGCCAGAACACCCACGTGCCTCCTACCTTCGGCTCCGGCACC







AAGCTCGAAATCAAGAGGGCGGCCGCATTCGTGCCGGTCTTCCTGCCA







GCGAAGCCCACCACGACGCCAGCGCCGCGACCACCAACACCGGCGCCC







ACCATCGCGTCGCAGCCCCTGTCCCTGCGCCCAGAGGCGTGCCGGCCA







GCGGCGGGGGGCGCAGTGCACACGAGGGGGCTGGACTTCGCCTGTGAT







ATCTACATCTGGGCGCCCTTGGCCGGGACTTGTGGGGTCCTTCTCCTG







TCACTGGTTATCACCCTTTACTGCAACCACAGGAACAGGAGTAAGAGG







AGCAGGCTCCTGCACAGTGACTACATGAACATGACTCCCCGCCGCCCC







GGGCCCACCCGCAAGCATTACCAGCCCTATGCCCCACCACGCGACTTC







GCAGCCTATCGCTCCCGTTTCTCTGTTGTTAAACGGGGCAGAAAGAAG







CTCCTGTATATATTCAAACAACCATTTATGAGACGAGTACAAACTAGT







CAAGAGGAAGATGGCTGTAGCTGCCGATTTCCAGAAGAAGAAGAAGGA







GGATGTGAACTGAGAGTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCG







TACCAGCAGGGCCAGAACCAGCTCTATAACGAGCTCAATCTAGGACGA







AGAGAGGAGTACGATGTTTTGGACAAGAGACGTGGCCGGGACCCTGAG







ATGGGGGGAAAGCCGAGAAGGAAGAACCCTCAGGAAGGCCTGTACAAT







GAACTGCAGAAAGATAAGATGGCGGAGGCCTACAGTGAGATTGGGATG







AAAGGCGAGCGCCGGAGGGGCAAGGGGCACGATGGCCTTTACCAGGGT







CTCAGTACAGCCACCAAGGACACCTACGACGCCCTTCACATGCAGGCC








embedded image






embedded image





ATGTTCCATGTGAGCTTCAGGTACATCTTCGGACTGCCTCCTCTCATC







CTGGTCCTCCTCCCCGTGGCCAGCTCCGACTGTGACATCGAAGGAAAG







GATGGCAAGCAGTACGAAAGCGTGCTGATGGTGAGCATCGATCAGCTC







CTGGATTCCATGAAGGAAATCGGCTCCAACTGCCTCAACAATGAGTTC







AACTTTTTTAAGAGGCATATCTGCGACGCCAACAAGGAGGGCATGTTT







CTGTTCAGGGCCGCCAGGAAGCTGAGACAGTTCCTCAAGATGAATAGC







ACCGGCGACTTCGACCTCCATCTGCTGAAGGTGTCCGAGGGAACCACC







ATCCTGCTGAACTGCACCGGCCAAGTGAAGGGAAGAAAACCTGCTGCC







CTGGGCGAGGCTCAGCCTACCAAGAGCCTCGAGGAGAACAAAAGCCTG







AAGGAGCAGAAGAAGCTGAACGACCTGTGCTTCCTCAAGAGGCTCCTG







CAGGAGATTAAGACCTGTTGGAACAAGATCCTGATGGGCACAAAGGAG








embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image






embedded image











In the table, the single square represents a gene encoding a human immunoglobulin H-chain-derived signal sequence, the double squares each represent a gene encoding a 2A self-cleaving peptide, the single underline represents a gene encoding a CAR, the double underline represents a gene encoding hIL-7, the broken underline represents a gene encoding hCCL19, and the wavy underline represents a gene encoding HSV-TK.


[Production of Recombinant Retrovirus]

The retroviral vector for expression of anti-GPC3 scFv CAR, etc. produced was transfected into a GP2-293 packaging cell line (available from Takara Bio Inc.) using Lipofectamine 3000 (available from Life Technologies Corporation). 48 hours after the transfection, a culture supernatant containing the recombinant retrovirus (retrovirus for expression of anti-GPC3 scFv CAR, etc.) produced was collected.


[Infection of T Cells with Recombinant Retrovirus]


In order to produce anti-GPC3 scFv CAR-expressing T cells, T cells were infected with the recombinant retrovirus (retrovirus for expression of anti-GPC3 scFv CAR, etc.), to introduce the retroviral vector for expression of anti-GPC3 scFv CAR, etc. into the T cells. Specifically, peripheral blood mononuclear cells were isolated from the peripheral blood of a healthy person according to a conventional method and then activated and cultured for 48 hours in the presence of immobilized anti-CD3 monoclonal antibody clone (OKT3) (5 μg/mL) and Retronectin® available from Takara Bio Inc. (25 μg/mL), and IL-2. A virus adsorption plate was produced by adding a culture supernatant containing a recombinant retrovirus for expression of anti-GPC3 scFv CAR, etc. onto a plate coated with retronectin, followed by centrifugation at 4° C. and 2000 g for two hours. The human T cells activated and cultured were seeded at 1×105 cells/mL onto the virus adsorption plate produced and cultured overnight in the presence of IL-2. On the next day, the T cells were collected and seeded onto a new virus adsorption plate to perform the second viral infection. After culturing for 4 hours, the cells were collected, and 3 times the amount of the culture solution was added thereto. The cells were seeded on a cell culture plate and then cultured for 3 days.


2. Method
[Flow Cytometry Analysis]

Flow cytometry analysis was performed to confirm that the human T cells infected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. express an anti-GPC3 scFv CAR. Specifically, the human T-cell group (T-cell group containing the anti-GPC3 scFv CAR-T cells) infected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. was incubated on ice for 30 minutes in the presence of a biotinylated recombinant GPC3 protein (available from R&D Systems). A FACS Buffer (1% BSA/PBS solution) was added thereto, followed by centrifugation, to remove the supernatant. Thereafter, an APC-labeled anti-CD8 antibody (available from BioLegend, Inc.) and BV421-labeled streptavidin (available from BioLegend, Inc.) were added thereto, and the mixture was incubated on ice for 30 minutes. A flow cytometer (BD LSR Fortessa) (available from BD Biosciences) was used for detecting APC and BV421 and measuring the fluorescence levels thereof. As a negative control, a human T-cell group (T-cell group) uninfected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. was subjected to flow cytometry analysis in the same manner.


[Injury Activity on GPC3-Expressing Cancer Cells]

In order to confirm the cytotoxic activity of the anti-GPC3 scFv CAR-T cell on cancer cells, GPC3-expressing cancer cells were cultured in the presence of the T-cell group containing the anti-GPC3 scFv CAR-T cells. Specifically, the T-cell group containing the anti-GPC3 scFv CAR-T cells was mixed with a hepatocellular carcinoma-derived cell line (Sk-HEP-1 cells) (purchased from ECACC) or GPC3-expressing Sk-HEP-1 cells (Sk-HEP-1 GPC3 cells) at 1:1 (1×103/well), and the mixture was cultured in a 24-well plate. 48 hours thereafter, the cells were collected and incubated on ice for 30 minutes in the presence of a Phycoerythrin-labeled anti-CD45 antibody (available from BioLegend, Inc). A FACS Buffer (1% BSA/PBS solution) was added thereto, followed by centrifugation, to remove the supernatant. Thereafter, a flow cytometer (BD LSR Fortessa) (available from BD Biosciences) was used for detecting Phycoerythrin and analyzing the fluorescence level, and the numbers of CD45-positive cells (corresponding to the T cells) and CD45-negative cells (corresponding to remaining Sk-HEP-1 GPC3 cells) and the ratio thereof were measured.


[IFN-γ Producibility of Anti-GPC3 scFv CAR-T Cells]


Whether or not the anti-GPC3 scFv CAR-T cells have interferon gamma (IFN-γ) producibility was analyzed. Specifically, the T-cell group containing the anti-GPC3 scFv CAR-T cells (“CAR-T cells” in FIG. 3) or a T-cell group without gene introduction (“T cells” in FIG. 3) was mixed with Sk-HEP-1 cells or Sk-HEP-1 GPC3 cells at 1:1 (1×105/well each), the mixture was cultured in a 24-well plate for 48 hours, and the concentration of IFN-γ produced in the culture supernatant was measured by ELISA (available from BioLegend, Inc).


[IL-7 and CCL19 Producibility of Anti-GPC3 scFv CAR-T Cells]


Whether or not the anti-GPC3 scFv CAR-T cells have IL-7 and CCL19 producibility was analyzed. Specifically, the T-cell group containing the anti-GPC3 scFv CAR-T cells (“CAR-T cells” in FIG. 4) or a T-cell group without gene introduction (“T cells” in FIG. 4) was mixed with Sk-HEP-1 cells or Sk-HEP-1 GPC3 cells at 1:1 (1×105/well each), the mixture was cultured in a 24-well plate for 48 hours, and the concentrations of IL-7 and CCL19 produced in the culture supernatant were measured by ELISA (available from R&D Systems). As a negative control, a CAR T-cell group without gene introduction (T cells) with Sk-HEP-1 cells or Sk-HEP-1 GPC3 cells was cultured under the same conditions.


3. Results
[Flow Cytometry Analysis]

It was shown that 48.1% of cells in the human T-cell group infected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. were CAR positive in which BV421 was detected (see FIG. 1B). Meanwhile, almost no CAR-positive cells were observed in the retrovirus-uninfected human T-cell group (see Figure 1A). These results demonstrated that about 50% of cells in the human T-cell group infected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. were T cells expressing the anti-GPC3 scFv CAR (anti-GPC3 scFv CAR-T cells).


[Injury Activity on GPC3-Expressing Cancer Cells]

As compared with the case of using the T-cell group uninfected with the retrovirus for expression of anti-GPC3 scFv CAR, etc., the T-cell group containing the anti-GPC3 scFv CAR-T cells exhibited very high injury activity on Sk-HEP-1 GPC3 cells (“GPC3” in FIG. 2) and killed most of the Sk-HEP-1 GPC3 cells (see FIG. 2). Meanwhile, the T-cell group containing the anti-GPC3 scFv CAR-T cells exhibited almost no injury activity on Sk-HEP-1 cells (“mock” in FIG. 2), like the T-cell group (see FIG. 2). These results demonstrated that the anti-GPC3 scFv CAR-T cells exert cytotoxic activity specific for cancer cells expressing GPC3.


[IFN-γ Producibility of Anti-GPC3 scFv CAR-T Cells]


The T-cell group containing the anti-GPC3 scFv CAR-T cells exhibited very high IFN-γ producibility in the presence of Sk-HEP-1 GPC3 cells, as compared with the case of using the T-cell group uninfected with the retrovirus for expression of anti-GPC3 scFv CAR, etc. (see FIG. 3). Meanwhile, the T-cell group containing the anti-GPC3 scFv CAR-T cells exhibited almost no IFN-γ producibility in the presence of Sk-HEP-1 cells, like the T-cell group (see FIG. 3). These results demonstrated that the anti-GPC3 scFv in the anti-GPC3 scFv CAR-T cells were bound to GPC3 surface antigens in the cancer cells and activated, as a result of which IFN-γ was produced.


[IL-7 and CCL19 Producibility of Anti-GPC3 scFv CAR-T Cells]


The T-cell group containing the anti-GPC3 scFv CAR-T cells produced a low value of IL-7 and CCL19 in the presence of Sk-HEP-1 cells but produced a further high level of IL-7 and CCL19 in the presence of Sk-HEP-1 GPC3 cells (see FIG. 4). In the T-cell group uninfected with the retrovirus for expression of anti-GPC3 scFv CAR, etc., IL-7 production and CCL19 production were hardly observed in the presence of both Sk-HEP-1 cells and Sk-HEP-1 GPC3 cells. These results demonstrated that the anti-GPC3 scFv CAR-T cells produced IL-7 and CCL19.


INDUSTRIAL APPLICABILITY

The present invention contributes to the cancer immunotherapy field using immunocompetent cells.

Claims
  • 1. A chimeric antigen receptor (CAR) comprising: a single-chain antibody that specifically binds to a human GPC3 (Glypican-3)-derived polypeptide consisting of the amino acid sequence shown by SEQ ID No: 11;a transmembrane region fused to a carboxyl terminus of the single-chain antibody; andan immunocompetent cell activation signaling transduction region fused to the carboxyl terminus of the transmembrane region, whereinthe single-chain antibody comprises:a heavy chain complementarity determining region (CDR) 1 consisting of the amino acid sequence shown by SEQ ID No: 1, a heavy-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 2, and a heavy-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 3; anda light-chain CDR1 consisting of the amino acid sequence shown by SEQ ID No: 4, a light-chain CDR2 consisting of the amino acid sequence shown by SEQ ID No: 5, and a light-chain CDR3 consisting of the amino acid sequence shown by SEQ ID No: 6.
  • 2. The CAR according to claim 1, wherein the single-chain antibody comprises:a heavy-chain variable region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 7; anda light-chain variable region consisting of an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 8.
  • 3. The CAR according to claim 1, wherein the single-chain antibody comprises:an amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 12; oran amino acid sequence having at least 80% or more sequence identity with the amino acid sequence shown by SEQ ID No: 13.
  • 4. The CAR according to claim 1, wherein the transmembrane region and the immunocompetent cell activation signaling transduction region fused to the carboxyl terminus of the transmembrane region comprise an amino acid sequence having at least 80% or more sequence identity with an amino acid sequence shown by any one of SEQ ID Nos: 14 to 16.
  • 5. An isolated immunocompetent cell expressing the CAR according to claim 1.
  • 6. The isolated immunocompetent cell according to claim 5, further expressing interleukin 7 (IL-7) and chemokine ligand 19 (CCL19).
  • 7. An anticancer agent comprising: the isolated immunocompetent cell according to claim 5; anda pharmaceutically acceptable additive.
  • 8. A CAR gene encoding the CAR according to claim 1.
  • 9. A vector comprising: a promoter; andthe CAR gene encoding the CAR according to claim 8 operably linked downstream of the promoter.
  • 10. An isolated immunocompetent cell expressing the CAR according to claim 2.
  • 11. An isolated immunocompetent cell expressing the CAR according to claim 3.
  • 12. An isolated immunocompetent cell expressing the CAR according to claim 4.
  • 13. The isolated immunocompetent cell according to claim 10, further expressing IL-7 and CCL19.
  • 14. The isolated immunocompetent cell according to claim 11, further expressing IL-7 and CCL19.
  • 15. The isolated immunocompetent cell according to claim 12, further expressing IL-7 and CCL19.
  • 16. An anticancer agent comprising: the isolated immunocompetent cell according to claim 10; anda pharmaceutically acceptable additive.
  • 17. An anticancer agent comprising: the isolated immunocompetent cell according to claim 11; anda pharmaceutically acceptable additive.
  • 18. An anticancer agent comprising: the isolated immunocompetent cell according to claim 12; anda pharmaceutically acceptable additive.
  • 19. An anticancer agent comprising: the isolated immunocompetent cell according to claim 13; anda pharmaceutically acceptable additive.
  • 20. An anticancer agent comprising: the isolated immunocompetent cell according to claim 14; anda pharmaceutically acceptable additive.
Priority Claims (1)
Number Date Country Kind
2018-133970 Jul 2018 JP national
PCT Information
Filing Document Filing Date Country Kind
PCT/JP2019/027835 7/16/2019 WO