1
|
Liu M, Akahori Y, Imai N, Wang L, Negishi K, Kato T, Fujiwara H, Miwa H, Shiku H, Miyahara Y. MAGE-A4 pMHC-targeted CAR-T cells exploiting TCR machinery exhibit significantly improved in vivo function while retaining antigen specificity. J Immunother Cancer 2024; 12:e010248. [PMID: 39572159 PMCID: PMC11580264 DOI: 10.1136/jitc-2024-010248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/27/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The development of chimeric antigen receptor (CAR)-T cell therapies for solid tumors has attracted considerable attention, yet their clinical efficacy remains limited. Therefore, various efforts have been made to improve the efficacy of CAR-T cell therapy. As one promising strategy, incorporating the T-cell receptor (TCR) machinery into CAR structures has been reported to improve the efficacy of CAR-T cells in studies using conventional CARs targeting such as EGFR. However, in the case of peptide/major histocompatibility complex (pMHC)-targeted CARs, the advantages of exploiting TCR machinery have not been fully elucidated. We recently developed MAGE-A4-derived pMHC (MAGE-A4 pMHC)-targeted CAR-T cells (MA-CAR-T cells) using a highly specific human scFv antibody against MAGE-A4p230-239/HLA-A*02:01. We aimed to determine whether MAGE-A4 pMHC-targeted CAR-T cells using the TCR machinery (Hybrid MA-TCR-T cells) exhibit superior functionality without compromising antigen specificity. METHODS We constructed a retroviral vector expressing Hybrid MA-TCR where MAGE-A4 pMHC-specific scFv are fused to human TCR constant chains. RESULTS Hybrid MA-TCR-T cells demonstrated superior in vitro functions compared with MA-CAR-T cells, while maintaining strict antigen specificity. In addition, functional superiority of Hybrid MA-TCR-T cells to MA-CAR-T cells became more pronounced on repetitive antigen stimulation. In particular, Hybrid MA-TCR-T cells significantly inhibited tumor growth in an immunodeficient mouse model more effectively than MA-CAR-T cells. Ex vivo analyses indicated that their enhanced therapeutic efficacy might result from higher infiltration of functionally active, less differentiated Hybrid MA-TCR-T cells in tumor tissues. CONCLUSIONS These findings suggest that leveraging the TCR machinery is a promising strategy for enhancing pMHC-targeted CAR-T cell therapy for solid tumors, potentially leading to more effective treatments.
Collapse
Affiliation(s)
- Meiou Liu
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yasushi Akahori
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Naoko Imai
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Linan Wang
- Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kohei Negishi
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Takuma Kato
- Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Fujiwara
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroshi Miwa
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroshi Shiku
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yoshihiro Miyahara
- Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
2
|
Kawamoto H, Masuda K. Trends in cell medicine: from autologous cells to allogeneic universal-use cells for adoptive T-cell therapies. Int Immunol 2024; 36:65-73. [PMID: 38189591 PMCID: PMC10872703 DOI: 10.1093/intimm/dxad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/04/2024] [Indexed: 01/09/2024] Open
Abstract
In currently ongoing adoptive T-cell therapies, T cells collected from patients are given back to them after ex vivo activation and expansion. In some cases, T cells are transduced with chimeric antigen receptor (CAR) or T-cell receptor (TCR) genes during the ex vivo culture period in order to endow T cells with the desired antigen specificity. Although such strategies are effective in some types of cancer, there remain issues to be solved: (i) the limited number of cells, (ii) it is time-consuming, (iii) it is costly, and (iv) the quality can be unstable. Points (ii) and (iv) can be solved by preparing allogeneic T cells and cryopreserving them in advance and methods are being developed using healthy donor-derived T cells or pluripotent stem cells as materials. Whereas it is difficult to solve (i) and (iii) in the former case, all the issues can be cleared in the latter case. However, in either case, a new problem arises: rejection by the patient's immune system. Deletion of human leukocyte antigen (HLA) avoids rejection by recipient T cells, but causes rejection by NK cells, which can recognize loss of HLA class I. Various countermeasures have been developed, but no definitive solution is yet available. Therefore, further research and development are necessary.
Collapse
Affiliation(s)
- Hiroshi Kawamoto
- Laboratory of Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Laboratory of Regenerative Immunology, International Center for Cell and Gene Therapy, Fujita Health University, Toyoake 470-1192, Japan
| | - Kyoko Masuda
- Laboratory of Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
3
|
Takayanagi SI, Wang B, Hasegawa S, Nishikawa S, Fukumoto K, Nakano K, Chuganji S, Kato Y, Kamibayashi S, Minagawa A, Kunisato A, Nozawa H, Kaneko S. Mini-TCRs: Truncated T cell receptors to generate T cells from induced pluripotent stem cells. Mol Ther Methods Clin Dev 2023; 31:101109. [PMID: 37822720 PMCID: PMC10562677 DOI: 10.1016/j.omtm.2023.101109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/13/2023] [Indexed: 10/13/2023]
Abstract
Allogeneic T cell platforms utilizing induced pluripotent stem cell (iPSC) technology exhibit significant promise for the facilitation of adoptive immunotherapies. While mature T cell receptor (TCR) signaling plays a crucial role in generating T cells from iPSCs, the introduction of exogenous mature TCR genes carries a potential risk of causing graft-versus-host disease (GvHD). In this study, we present the development of truncated TCRα and TCRβ chains, termed mini-TCRs, which lack variable domains responsible for recognizing human leukocyte antigen (HLA)-peptide complexes. We successfully induced cytotoxic T lymphocytes (CTLs) from iPSCs by employing mini-TCRs. Combinations of TCRα and TCRβ fragments were screened from mini-TCR libraries based on the surface localization of CD3 proteins and their ability to transduce T cell signaling. Consequently, mini-TCR-expressing iPSCs underwent physiological T cell development, progressing from the CD4 and CD8 double-positive stage to the CD8 single-positive stage. The resulting iPSC-derived CTLs exhibited comparable cytokine production and cytotoxicity in comparison to that of full-length TCR-expressing T lymphocytes when chimeric antigen receptors (CARs) were expressed. These findings demonstrate the potential of mini-TCR-carrying iPSCs as a versatile platform for CAR T cell therapy, offering a promising avenue for advancing adoptive immunotherapies.
Collapse
Affiliation(s)
- Shin-ichiro Takayanagi
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Bo Wang
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Shinobi Therapeutics, Inc., 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Saki Hasegawa
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoshi Nishikawa
- R&D Division, Kyowa Kirin Co. Ltd, 3-6-6 Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Ken Fukumoto
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kohei Nakano
- Shinobi Therapeutics, Inc., 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Sayaka Chuganji
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuya Kato
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Sanae Kamibayashi
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsutaka Minagawa
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsushi Kunisato
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Hajime Nozawa
- Kirin Central Research Institute, Kirin Holdings Company, Ltd., 26-1, Muraoka-Higashi 2, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Shinobi Therapeutics, Inc., 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
4
|
Okada S, Muraoka D, Yasui K, Tawara I, Kawamura A, Okamoto S, Mineno J, Seo N, Shiku H, Eguchi S, Ikeda H. T cell receptor gene-modified allogeneic T cells with siRNA for endogenous T cell receptor induce efficient tumor regression without graft-versus-host disease. Cancer Sci 2023; 114:4172-4183. [PMID: 37675556 PMCID: PMC10637063 DOI: 10.1111/cas.15954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023] Open
Abstract
Adoptive immunotherapy using genetically engineered patient-derived lymphocytes to express tumor-reactive receptors is a promising treatment for malignancy. However, utilization of autologous T cells in this therapy limits the quality of gene-engineered T cells, thereby inhibiting the timely infusion of the cells into patients. In this study, we evaluated the anti-tumor efficacy and the potential to induce graft-versus-host disease (GVHD) in T cell receptor (TCR) gene-engineered allogeneic T cells that downregulate the endogenous TCR and HLA class I molecules with the aim of developing an "off-the-shelf" cell product with expanded application of genetically engineered T cells. We transduced human lymphocytes with a high-affinity TCR specific to the cancer/testis antigen NY-ESO-1 using a novel retrovirus vector with siRNAs specific to the endogenous TCR (siTCR vector). These T cells showed reduced expression of endogenous TCR and minimized reactivity to allogeneic cells in vitro. In non-obese diabetic/SCID/γcnull mice, TCR gene-transduced T cells induced tumor regression without development of GVHD. A lentivirus-based CRISPR/Cas9 system targeting β-2 microglobulin in TCR gene-modified T cells silenced the HLA class I expression and prevented allogeneic CD8+ T cell stimulation without disrupting their anti-tumor capacity. This report is the first demonstration that siTCR technology is effective in preventing GVHD. Adoptive cell therapy with allogeneic T cells engineered with siTCR vector may be useful in developing an "off-the-shelf" therapy for patients with malignancy.
Collapse
Affiliation(s)
- Satomi Okada
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Department of SurgeryNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Daisuke Muraoka
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Division of Translational OncoimmunologyAichi Cancer Center Research InstituteNagoyaJapan
| | - Kiyoshi Yasui
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Isao Tawara
- Department of Hematology and OncologyMie University Graduate School of MedicineMieJapan
| | | | | | | | - Naohiro Seo
- Department of Personalized Cancer ImmunotherapyMie University Graduate School of MedicineMieJapan
- Department of Bioengineering, School of EngineeringThe University of TokyoTokyoJapan
| | - Hiroshi Shiku
- Department of Personalized Cancer ImmunotherapyMie University Graduate School of MedicineMieJapan
| | - Susumu Eguchi
- Department of SurgeryNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| | - Hiroaki Ikeda
- Department of OncologyNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
- Leading Medical Research Core UnitNagasaki University Graduate School of Biomedical SciencesNagasakiJapan
| |
Collapse
|
5
|
Li J, Xiao Z, Wang D, Jia L, Nie S, Zeng X, Hu W. The screening, identification, design and clinical application of tumor-specific neoantigens for TCR-T cells. Mol Cancer 2023; 22:141. [PMID: 37649123 PMCID: PMC10466891 DOI: 10.1186/s12943-023-01844-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Recent advances in neoantigen research have accelerated the development of tumor immunotherapies, including adoptive cell therapies (ACTs), cancer vaccines and antibody-based therapies, particularly for solid tumors. With the development of next-generation sequencing and bioinformatics technology, the rapid identification and prediction of tumor-specific antigens (TSAs) has become possible. Compared with tumor-associated antigens (TAAs), highly immunogenic TSAs provide new targets for personalized tumor immunotherapy and can be used as prospective indicators for predicting tumor patient survival, prognosis, and immune checkpoint blockade response. Here, the identification and characterization of neoantigens and the clinical application of neoantigen-based TCR-T immunotherapy strategies are summarized, and the current status, inherent challenges, and clinical translational potential of these strategies are discussed.
Collapse
Affiliation(s)
- Jiangping Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Zhiwen Xiao
- Department of Otolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, People's Republic of China
| | - Donghui Wang
- Department of Radiation Oncology, The Third Affiliated Hospital Sun Yat-Sen University, Guangzhou, 510630, People's Republic of China
| | - Lei Jia
- International Health Medicine Innovation Center, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Shihong Nie
- Department of Radiation Oncology, West China Hospital, Sichuan University, Cancer Center, Chengdu, 610041, People's Republic of China
| | - Xingda Zeng
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Wei Hu
- Division of Vascular Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| |
Collapse
|
6
|
Barakat C, Inagaki Y, Mizuno S, Nishio N, Katsuyama N, Sato Y, Kobayashi M, Ozeki K, Iida H, Tomita A, Sawa M, Demachi-Okamura A, Takahashi Y, Nishikawa H, Akatsuka Y. Development of TCR-T cell therapy targeting mismatched HLA-DPB1 for relapsed leukemia after allogeneic transplantation. Int J Hematol 2023:10.1007/s12185-023-03621-y. [PMID: 37310580 DOI: 10.1007/s12185-023-03621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2023]
Abstract
Relapsed leukemia after allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains a significant challenge, with the re-emergence of the primary disease being the most frequent cause of death. Human leukocyte antigen (HLA)-DPB1 mismatch occurs in approximately 70% of unrelated allo-HSCT cases, and targeting mismatched HLA-DPB1 is considered reasonable for treating relapsed leukemia following allo-HSCT if performed under proper conditions. In this study, we established several clones restricted to HLA-DPB1*02:01, -DPB1*04:02, and -DPB1*09:01 from three patients who underwent HLA-DPB1 mismatched allo-HSCT using donor-derived alloreactive T cells primed to mismatched HLA-DPB1 in the recipient's body after transplantation. A detailed analysis of the DPB1*09:01-restricted clone 2A9 showed reactivity against various leukemia cell lines and primary myeloid leukemia blasts, even with low HLA-DP expression. T cell receptor (TCR)-T cells derived from clone 2A9 retained the ability to trigger HLA-DPB1*09:01-restricted recognition and lysis of various leukemia cell lines in vitro. Our study demonstrated that the induction of mismatched HLA-DPB1 specific T cell clones from physiologically primed post-allo-HSCT alloreactive CD4+ T cells and the redirection of T cells with cloned TCR cDNA by gene transfer are feasible as techniques for future adoptive immunotherapy.
Collapse
Affiliation(s)
- Carolyne Barakat
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
| | - Yuichiro Inagaki
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Shohei Mizuno
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Nobuhiro Nishio
- Center for Advanced Medicine and Clinical Research, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Katsuyama
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
| | - Yoshie Sato
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
| | - Miki Kobayashi
- Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Kazutaka Ozeki
- Department of Hematology and Oncology, JA Aichi Konan Kosei Hospital, Konan, Japan
| | - Hiroatsu Iida
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan
| | - Yoshiki Akatsuka
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8560, Japan.
| |
Collapse
|
7
|
Karahan ZS, Aras M, Sütlü T. TCR-NK Cells: A Novel Source for Adoptive Immunotherapy of Cancer. Turk J Haematol 2023; 40:1-10. [PMID: 36719099 PMCID: PMC9979742 DOI: 10.4274/tjh.galenos.2022.2022.0534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Antigen-specific retargeting of cytotoxic lymphocytes against tumor-associated antigens has thus far remained largely dependent on chimeric antigen receptors (CARs) that can be constructed by the fusion of an extracellular targeting domain (classically a single-chain variable fragment from an antibody) fused with intracellular signaling domains to trigger activation of T or natural killer (NK) cells. A major limitation of CAR-based therapies is that this technology only allows for the targeting of antigens that would be located on the surface of target cells while non-surface antigens, which affect approximately three-fourths of all human genes, remain out of reach. The targeting of non-surface antigens is only possible using inherent T cell receptor (TCR) mechanisms. However, introducing a second TCR into T cells via genetic modification is problematic due to the heterodimeric nature of the TCR ligand-binding domain, which is composed of TCR α and β chains. It has been observed that the delivery of a second TCR α/β pair may lead to the mispairing of new TCR chains with the endogenously expressed ones and create mixed TCR dimers, and this has negatively affected the advancement of TCR-based T cell therapies. Recently, NK cells have been put forward as possible effectors for TCR gene therapy. Since NK cells do not endogenously express TCR chains, this seems to be an infallible approach to circumventing the problem of mispairing. Moreover, the similarity of intracellular signaling pathways and mechanisms of cytotoxicity between NK and T cells ensures that the triggering of antigen-specific responses by the TCR/CD3 complex can be used to induce antigen-specific cytotoxicity by TCR-modified NK (TCR-NK) cells. This review provides an overview of the initial studies of TCR-NK cells, identifies open questions in the field, and defines the place of this approach within the spectrum of adoptive immunotherapy techniques that rely on cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Zeynep Sena Karahan
- Boğaziçi University Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, İstanbul, Türkiye
| | - Mertkaya Aras
- Boğaziçi University Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, İstanbul, Türkiye,Sabancı University Faculty of Engineering and Natural Sciences, Department of Molecular Biology, Genetics, and Bioengineering, İstanbul, Türkiye
| | - Tolga Sütlü
- Boğaziçi University Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, İstanbul, Türkiye,* Address for Correspondence: Boğaziçi University Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, İstanbul, Türkiye E-mail:
| |
Collapse
|
8
|
Sato O, Tsuchikawa T, Kato T, Amaishi Y, Okamoto S, Mineno J, Takeuchi Y, Sasaki K, Nakamura T, Umemoto K, Suzuki T, Wang L, Wang Y, Hatanaka KC, Mitsuhashi T, Hatanaka Y, Shiku H, Hirano S. Tumor Growth Suppression of Pancreatic Cancer Orthotopic Xenograft Model by CEA-Targeting CAR-T Cells. Cancers (Basel) 2023; 15:601. [PMID: 36765558 PMCID: PMC9913141 DOI: 10.3390/cancers15030601] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Chimeric antigen receptor engineered T cell (CAR-T) therapy has high therapeutic efficacy against blood cancers, but it has not shown satisfactory results in solid tumors. Therefore, we examined the therapeutic effect of CAR-T therapy targeting carcinoembryonic antigen (CEA) in pancreatic adenocarcinoma (PDAC). CEA expression levels on the cell membranes of various PDAC cell lines were evaluated using flow cytometry and the cells were divided into high, medium, and low expression groups. The relationship between CEA expression level and the antitumor effect of anti-CEA-CAR-T was evaluated using a functional assay for various PDAC cell lines; a significant correlation was observed between CEA expression level and the antitumor effect. We created orthotopic PDAC xenograft mouse models and injected with anti-CEA-CAR-T; only the cell line with high CEA expression exhibited a significant therapeutic effect. Thus, the therapeutic effect of CAR-T therapy was related to the target antigen expression level, and the further retrospective analysis of pathological findings from PDAC patients showed a correlation between the intensity of CEA immunostaining and tumor heterogeneity. Therefore, CEA expression levels in biopsies or surgical specimens can be clinically used as biomarkers to select PDAC patients for anti-CAR-T therapy.
Collapse
Affiliation(s)
- Osamu Sato
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Takuma Kato
- Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
- Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu 514-8507, Mie, Japan
| | | | | | | | - Yuta Takeuchi
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Katsunori Sasaki
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Kazufumi Umemoto
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Tomohiro Suzuki
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| | - Linan Wang
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Yizheng Wang
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Kanako C. Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo 060-8648, Hokkaido, Japan
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo 060-8648, Hokkaido, Japan
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo 060-8648, Hokkaido, Japan
| | - Hiroshi Shiku
- Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu 514-8507, Mie, Japan
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Hokkaido, Japan
| |
Collapse
|
9
|
Ishihara M, Kitano S, Kageyama S, Miyahara Y, Yamamoto N, Kato H, Mishima H, Hattori H, Funakoshi T, Kojima T, Sasada T, Sato E, Okamoto S, Tomura D, Nukaya I, Chono H, Mineno J, Kairi MF, Diem Hoang Nguyen P, Simoni Y, Nardin A, Newell E, Fehlings M, Ikeda H, Watanabe T, Shiku H. NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome. J Immunother Cancer 2022; 10:e003811. [PMID: 35768164 PMCID: PMC9244667 DOI: 10.1136/jitc-2021-003811] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Because of the shortage of ideal cell surface antigens, the development of T-cell receptor (TCR)-engineered T cells (TCR-T) that target intracellular antigens such as NY-ESO-1 is a promising approach for treating patients with solid tumors. However, endogenous TCRs in vector-transduced T cells have been suggested to impair cell-surface expression of transduced TCR while generating mispaired TCRs that can become self-reactive. METHODS We conducted a first-in-human phase I clinical trial with the TCR-transduced T-cell product (TBI-1301) in patients with NY-ESO-1-expressing solid tumors. In manufacturing TCR-T cells, we used a novel affinity-enhanced NY-ESO-1-specific TCR that was transduced by a retroviral vector that enables siRNA (small interfering RNA)-mediated silencing of endogenous TCR. The patients were divided into two cohorts. Cohort 1 was given a dose of 5×108 cells (whole cells including TCR-T cells) preconditioned with 1500 mg/m2 cyclophosphamide. Cohort 2 was given 5× 109 cells preconditioned with 1500 mg/m2 cyclophosphamide. RESULTS In vitro study showed that both the CD8+ and CD4+ T fractions of TCR-T cells exhibited cytotoxic effects against NY-ESO-1-expressing tumor cells. Three patients and six patients were allocated to cohort 1 and cohort 2, respectively. Three of the six patients who received 5×109 cells showed tumor response, while three patients developed early-onset cytokine release syndrome (CRS). One of the patients developed a grade 3 lung injury associated with the infiltration of the TCR-T cells. No siRNA-related adverse events other than CRS were observed. Cytokines including interleukin 6 I and monocyte chemotactic protein-1/chemokine (C-C motif) ligand (CCL2)increased in the sera of patients with CRS. In vitro analysis showed these cytokines were not secreted from the T cells infused. A significant fraction of the manufactured T cells in patients with CRS was found to express either CD244, CD39, or both at high levels. CONCLUSIONS The trial showed that endogenous TCR-silenced and affinity-enhanced NY-ESO-1 TCR-T cells were safely administered except for grade 3 lung injury. The TCR-T cell infusion exhibited significant tumor response and early-onset CRS in patients with tumors that express NY-ESO-1 at high levels. The differentiation properties of the manufactured T cells may be prognostic for TCR-T-related CRS. TRIAL REGISTRATION NUMBER NCT02366546.
Collapse
Affiliation(s)
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy Development, Advanced Medical Development Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Experimental Therapeutics, National Cancer Institue Hospital, Tokyo, Japan
| | - Shinichi Kageyama
- Departments of Immuno-Gene Therapy and Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yoshihiro Miyahara
- Departments of Immuno-Gene Therapy and Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Institue Hospital, Tokyo, Japan
| | - Hidefumi Kato
- Department of Transfusion Medicine, Aichi Medical University, Nagakute, Japan
| | | | - Hiroyoshi Hattori
- Laboratory of Advanced Therapy, Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Eiichi Sato
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takashi Watanabe
- Departments of Immuno-Gene Therapy and Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hiroshi Shiku
- Departments of Immuno-Gene Therapy and Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
10
|
Zhang X, Jin X, Sun R, Zhang M, Lu W, Zhao M. Gene knockout in cellular immunotherapy: Application and limitations. Cancer Lett 2022; 540:215736. [DOI: 10.1016/j.canlet.2022.215736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/30/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022]
|
11
|
Jones HF, Molvi Z, Klatt MG, Dao T, Scheinberg DA. Empirical and Rational Design of T Cell Receptor-Based Immunotherapies. Front Immunol 2021; 11:585385. [PMID: 33569049 PMCID: PMC7868419 DOI: 10.3389/fimmu.2020.585385] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/04/2020] [Indexed: 01/04/2023] Open
Abstract
The use of T cells reactive with intracellular tumor-associated or tumor-specific antigens has been a promising strategy for cancer immunotherapies in the past three decades, but the approach has been constrained by a limited understanding of the T cell receptor's (TCR) complex functions and specificities. Newer TCR and T cell-based approaches are in development, including engineered adoptive T cells with enhanced TCR affinities, TCR mimic antibodies, and T cell-redirecting bispecific agents. These new therapeutic modalities are exciting opportunities by which TCR recognition can be further exploited for therapeutic benefit. In this review we summarize the development of TCR-based therapeutic strategies and focus on balancing efficacy and potency versus specificity, and hence, possible toxicity, of these powerful therapeutic modalities.
Collapse
Affiliation(s)
- Heather F. Jones
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, New York, NY, United States
| | - Zaki Molvi
- Weill Cornell Medicine, New York, NY, United States
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Martin G. Klatt
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
12
|
Gaissmaier L, Elshiaty M, Christopoulos P. Breaking Bottlenecks for the TCR Therapy of Cancer. Cells 2020; 9:E2095. [PMID: 32937956 PMCID: PMC7564186 DOI: 10.3390/cells9092095] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors have redefined the treatment of cancer, but their efficacy depends critically on the presence of sufficient tumor-specific lymphocytes, and cellular immunotherapies develop rapidly to fill this gap. The paucity of suitable extracellular and tumor-associated antigens in solid cancers necessitates the use of neoantigen-directed T-cell-receptor (TCR)-engineered cells, while prevention of tumor evasion requires combined targeting of multiple neoepitopes. These can be currently identified within 2 weeks by combining cutting-edge next-generation sequencing with bioinformatic pipelines and used to select tumor-reactive TCRs in a high-throughput manner for expeditious scalable non-viral gene editing of autologous or allogeneic lymphocytes. "Young" cells with a naive, memory stem or central memory phenotype can be additionally armored with "next-generation" features against exhaustion and the immunosuppressive tumor microenvironment, where they wander after reinfusion to attack heavily pretreated and hitherto hopeless neoplasms. Facilitated by major technological breakthroughs in critical manufacturing steps, based on a solid preclinical rationale, and backed by rapidly accumulating evidence, TCR therapies break one bottleneck after the other and hold the promise to become the next immuno-oncological revolution.
Collapse
Affiliation(s)
- Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Mariam Elshiaty
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany; (L.G.); (M.E.)
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| |
Collapse
|
13
|
A bicistronic vector backbone for rapid seamless cloning and chimerization of αβT-cell receptor sequences. PLoS One 2020; 15:e0238875. [PMID: 32903281 PMCID: PMC7480877 DOI: 10.1371/journal.pone.0238875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/25/2020] [Indexed: 11/19/2022] Open
Abstract
To facilitate preclinical testing of T-cell receptors (TCRs) derived from tumor-reactive T-cell clones it is necessary to develop convenient and rapid cloning strategies for the generation of TCR expression constructs. Herein, we describe a pDONR™221 vector backbone allowing to generate Gateway™ compatible entry clones encoding optimized bicistronic αβTCR constructs. It harbors P2A-linked TCR constant regions and head-to-head-oriented recognition sites of the Type IIS restriction enzymes BsmBI and BsaI for seamless cloning of the TCRα and TCRβ V(D)J regions, respectively. Additional well-established TCR optimizations were incorporated to enhance TCR functionality. This included replacing of the human αβTCR constant regions with their codon-optimized murine counterparts for chimerization, addition of a second interchain disulfide bond and arrangement of the TCR chains in the order β-P2A-α. We exemplified the utility of our vector backbone by cloning and functional testing of three melanoma-reactive TCRs in primary human T cells.
Collapse
|
14
|
Sakai T, Terakura S, Miyao K, Okuno S, Adachi Y, Umemura K, Julamanee J, Watanabe K, Hamana H, Kishi H, Leitner J, Steinberger P, Nishida T, Murata M, Kiyoi H. Artificial T Cell Adaptor Molecule-Transduced TCR-T Cells Demonstrated Improved Proliferation Only When Transduced in a Higher Intensity. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:613-622. [PMID: 33005728 PMCID: PMC7509457 DOI: 10.1016/j.omto.2020.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
An artificial T cell adaptor molecule (ATAM) was generated to improve persistence of T cell receptor (TCR) gene-transduced T (TCR-T) cells compared to such persistence in a preceding study. ATAMs are gene-modified CD3ζ with the intracellular domain of 4-1BB inserted in the middle of CD3ζ. NY-ESO-1 TCR-T cells transduced with an ATAM with two separated virus vectors demonstrated superior proliferation upon antigen stimulation. To further develop clinically applicable ATAM-transduced TCR-T cells, we attempted to make a single virus vector to transduce the TCR and ATAM simultaneously. Because we failed to observe improved proliferation capacity upon stimulation after one virus vector (1vv) transduction, we compared TCR-T cells transduced with 1vv and two virus vector (2vv) methods to elucidate the reason. In Jurkat reporter cells, an ATAM transduced by the 2vv method demonstrated a higher intensity than by the 1vv method, and the ATAM intensity was associated with increased nuclear factor κB (NF-κB) signals upon stimulation. In ATAM-transduced primary T cells, a transduced ATAM by the 2vv method showed higher intensity and better proliferation. ATAM-transduced TCR-T cells demonstrated improved proliferation only when the ATAM was transduced at a higher intensity. To create a simpler transduction method, we need to develop a strategy to make a higher ATAM expression to prove the efficacy of ATAM transduction in TCR-T therapy.
Collapse
Affiliation(s)
- Toshiyasu Sakai
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seitaro Terakura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kotaro Miyao
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shingo Okuno
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitaka Adachi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Umemura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jakrawadee Julamanee
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Clinical Hematology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Keisuke Watanabe
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cancer Immunology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroshi Hamana
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Judith Leitner
- Division for Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division for Immune Receptors and T Cell Activation, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Tetsuya Nishida
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Murata
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
15
|
Strategies for Optimizing the Production of Proteins and Peptides with Multiple Disulfide Bonds. Antibiotics (Basel) 2020; 9:antibiotics9090541. [PMID: 32858882 PMCID: PMC7558204 DOI: 10.3390/antibiotics9090541] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Bacteria can produce recombinant proteins quickly and cost effectively. However, their physiological properties limit their use for the production of proteins in their native form, especially polypeptides that are subjected to major post-translational modifications. Proteins that rely on disulfide bridges for their stability are difficult to produce in Escherichia coli. The bacterium offers the least costly, simplest, and fastest method for protein production. However, it is difficult to produce proteins with a very large size. Saccharomyces cerevisiae and Pichia pastoris are the most commonly used yeast species for protein production. At a low expense, yeasts can offer high protein yields, generate proteins with a molecular weight greater than 50 kDa, extract signal sequences, and glycosylate proteins. Both eukaryotic and prokaryotic species maintain reducing conditions in the cytoplasm. Hence, the formation of disulfide bonds is inhibited. These bonds are formed in eukaryotic cells during the export cycle, under the oxidizing conditions of the endoplasmic reticulum. Bacteria do not have an advanced subcellular space, but in the oxidizing periplasm, they exhibit both export systems and enzymatic activities directed at the formation and quality of disulfide bonds. Here, we discuss current techniques used to target eukaryotic and prokaryotic species for the generation of correctly folded proteins with disulfide bonds.
Collapse
|
16
|
The Quest for the Best: How TCR Affinity, Avidity, and Functional Avidity Affect TCR-Engineered T-Cell Antitumor Responses. Cells 2020; 9:cells9071720. [PMID: 32708366 PMCID: PMC7408146 DOI: 10.3390/cells9071720] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Over the past decades, adoptive transfer of T cells has revolutionized cancer immunotherapy. In particular, T-cell receptor (TCR) engineering of T cells has marked important milestones in developing more precise and personalized cancer immunotherapies. However, to get the most benefit out of this approach, understanding the role that TCR affinity, avidity, and functional avidity play on how TCRs and T cells function in the context of tumor-associated antigen (TAA) recognition is vital to keep generating improved adoptive T-cell therapies. Aside from TCR-related parameters, other critical factors that govern T-cell activation are the effect of TCR co-receptors on TCR–peptide-major histocompatibility complex (pMHC) stabilization and TCR signaling, tumor epitope density, and TCR expression levels in TCR-engineered T cells. In this review, we describe the key aspects governing TCR specificity, T-cell activation, and how these concepts can be applied to cancer-specific TCR redirection of T cells.
Collapse
|
17
|
Kawamura K, Tanaka Y, Nakasone H, Ishihara Y, Kako S, Kobayashi S, Tanaka Y, Ohmori T, Uchimaru K, Okamoto S, Mineno J, Shiku H, Nishimura S, Kanda Y. Development of a Unique T Cell Receptor Gene-Transferred Tax-Redirected T Cell Immunotherapy for Adult T Cell Leukemia. Biol Blood Marrow Transplant 2020; 26:1377-1385. [PMID: 32311478 DOI: 10.1016/j.bbmt.2020.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 03/24/2020] [Accepted: 04/05/2020] [Indexed: 11/28/2022]
Abstract
Adult T cell leukemia/lymphoma (ATL) is an aggressive peripheral T cell neoplasm caused by infection with human T cell lymphotropic virus type-1 (HTLV-1). Its prognosis remains extremely poor. Tax, the most important regulatory protein for HTLV-1, is associated with the aggressive proliferation of host cells and is also a major target antigen for CD8+ cytotoxic T cells (CTLs). Based on our previous findings that Tax-specific CTLs with a T cell receptor (TCR) containing a unique amino-acid sequence motif exhibit strong HLA-A*24:02-restricted, Tax301-309-specific activity against HTLV-1, we aimed to develop a Tax-redirected T cell immunotherapy for ATL. TCR-ɑ/β genes were cloned from a previously established CTL clone and transduced into peripheral blood mononuclear cells (PBMCs) of healthy volunteers using a retroviral siTCR vector. Then the cytotoxic efficacy against HTLV-1-infected T cells or primary ATL cells was assessed both in vitro and in vivo. The redirected CTLs (Tax-siCTLs) produced a large amount of cytokines and showed strong killing activity against ATL/HTLV-1-infected T cells in vitro, although they did not have universal activity against ATL cells. Next, in a xenograft mouse model using an HTLV-1-infected T cell line (MT-2), in all mice treated with Tax-siCTLs, the tumor rapidly diminished and finally disappeared without normal tissue damage, although all mice that were untreated or treated with non-gene-modified PBMCs died because of tumor progression. Our findings confirm that Tax-siCTLs can exert strong anti-ATL/HTLV-1 effects without a significant reaction against normal cells and have the potential to be a novel immunotherapy for ATL patients.
Collapse
Affiliation(s)
- Koji Kawamura
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yukie Tanaka
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan; Division of Molecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Research Core, Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideki Nakasone
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yuko Ishihara
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Shinichi Kako
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Seiichiro Kobayashi
- Division of Molecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuetsu Tanaka
- Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University, Tochigi, Japan
| | - Kaoru Uchimaru
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | - Hiroshi Shiku
- Department for Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Saitama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan; Division of Hematology, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| |
Collapse
|
18
|
Clauss J, Obenaus M, Miskey C, Ivics Z, Izsvák Z, Uckert W, Bunse M. Efficient Non-Viral T-Cell Engineering by Sleeping Beauty Minicircles Diminishing DNA Toxicity and miRNAs Silencing the Endogenous T-Cell Receptors. Hum Gene Ther 2019; 29:569-584. [PMID: 29562762 DOI: 10.1089/hum.2017.136] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transposon-based vectors have entered clinical trials as an alternative to viral vectors for genetic engineering of T cells. However, transposon vectors require DNA transfection into T cells, which were found to cause adverse effects. T-cell viability was decreased in a dose-dependent manner, and DNA-transfected T cells showed a delayed response upon T-cell receptor (TCR) stimulation with regard to blast formation, proliferation, and surface expression of CD25 and CD28. Gene expression analysis demonstrated a DNA-dependent induction of a type I interferon response and interferon-β upregulation. By combining Sleeping Beauty transposon minicircle vectors with SB100X transposase-encoding RNA, it was possible to reduce the amount of total DNA required, and stable expression of therapeutic TCRs was achieved in >50% of human T cells without enrichment. The TCR-engineered T cells mediated effective tumor cell killing and cytokine secretion upon antigen-specific stimulation. Additionally, the Sleeping Beauty transposon system was further improved by miRNAs silencing the endogenous TCR chains. These miRNAs increased the surface expression of the transgenic TCR, diminished mispairing with endogenous TCR chains, and enhanced antigen-specific T-cell functionality. This approach facilitates the rapid non-viral generation of highly functional, engineered T cells for immunotherapy.
Collapse
Affiliation(s)
- Julian Clauss
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany
| | - Matthias Obenaus
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,2 Charité Universitätsmedizin Berlin , Campus Virchow-Klinikum, Berlin, Germany
| | - Csaba Miskey
- 3 Division of Medical Biotechnology, Paul Ehrlich-Institut , Langen, Germany
| | - Zoltán Ivics
- 3 Division of Medical Biotechnology, Paul Ehrlich-Institut , Langen, Germany
| | - Zsuzsanna Izsvák
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,4 Berlin Institute of Health , Berlin, Germany
| | - Wolfgang Uckert
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany .,4 Berlin Institute of Health , Berlin, Germany .,5 Institute of Biology, Humboldt-Universität zu Berlin , Berlin, Germany
| | - Mario Bunse
- 1 Max Delbrück Center for Molecular Medicine in the Helmholtz Association , Berlin, Germany
| |
Collapse
|
19
|
Ohta R, Demachi-Okamura A, Akatsuka Y, Fujiwara H, Kuzushima K. Improving TCR affinity on 293T cells. J Immunol Methods 2018; 466:1-8. [PMID: 30468736 DOI: 10.1016/j.jim.2018.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022]
Abstract
This study presents an efficient method to improve TCR affinity, comprising 1) CDR-directed saturation mutation of TCR cDNA, 2) transient TCR display on CD3-expressing HEK293T (CD3-293T) cells by simple plasmid transfection, 3) staining with HLA-tetramers, and 4) multi-round sorting of cells with CD8-independent tetramer binding on a flow cytometer. Using these procedures, we successfully identified mutant TCRs with enhanced binding from an HLA-A*24:02-restricted, human telomerase reverse transcriptase (hTERT)-specific TCR. Two such clones, 2A7A and 2D162, harboring mutations in CDR1 and CDR2 of TCRβ, respectively, were isolated with both showing sequential four amino acid substitutions. When expressed on CD3-293T cells along with wild-type TCRα, the TCR molecules of these mutants as well as their combinatory mutation, bound to HLA-A24/hTERT-tetramers more strongly than the wild-type TCRs, without binding to control tetramers. Besides, in order to facilitate a functional study of TCR, we established an artificial T cell line, designated as CD8I-J2, which expresses a human CD8 and IFN-γ producing cassette by modifying Jurkat-derived J.RT3-T3.5 cells. CD8I-J2 cells expressing wild-type or affinity-enhanced hTERT-specific TCRs were analyzed for their recognition of serially diluted cognate peptide on HLA-A*24:02-transduced T2 cells. CD8I-J2 cells expressing each mutant TCR recognized the hTERT peptide at lower concentrations than wild-type TCR. The hierarchy of peptide recognition is concordant with tetramer binding on CD3-293T cells and none of these mutant TCRs were cross-reactive with irrelevant peptides reported to be present on HLA-A*24:02 molecules as far as tested. These methods might thus be useful for obtaining high affinity mutants from other TCRs of interest.
Collapse
Affiliation(s)
- Rieko Ohta
- Division of Immune Response, Aichi, Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Ayako Demachi-Okamura
- Division of Immune Response, Aichi, Cancer Center Research Institute, Nagoya 464-8681, Japan
| | - Yoshiki Akatsuka
- Division of Immune Response, Aichi, Cancer Center Research Institute, Nagoya 464-8681, Japan; Department of Hematology, Fujita Health University, Aichi 470-1192, Japan
| | - Hiroshi Fujiwara
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, 791-0295, Japan
| | - Kiyotaka Kuzushima
- Division of Immune Response, Aichi, Cancer Center Research Institute, Nagoya 464-8681, Japan; Division of Cellular Oncology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
20
|
Campillo-Davo D, Fujiki F, Van den Bergh JMJ, De Reu H, Smits ELJM, Goossens H, Sugiyama H, Lion E, Berneman ZN, Van Tendeloo V. Efficient and Non-genotoxic RNA-Based Engineering of Human T Cells Using Tumor-Specific T Cell Receptors With Minimal TCR Mispairing. Front Immunol 2018; 9:2503. [PMID: 30464762 PMCID: PMC6234959 DOI: 10.3389/fimmu.2018.02503] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Genetic engineering of T cells with tumor specific T-cell receptors (TCR) is a promising strategy to redirect their specificity against cancer cells in adoptive T cell therapy protocols. Most studies are exploiting integrating retro- or lentiviral vectors to permanently introduce the therapeutic TCR, which can pose serious safety issues when treatment-related toxicities would occur. Therefore, we developed a versatile, non-genotoxic transfection method for human unstimulated CD8+ T cells. We describe an optimized double sequential electroporation platform whereby Dicer-substrate small interfering RNAs (DsiRNA) are first introduced to suppress endogenous TCR α and β expression, followed by electroporation with DsiRNA-resistant tumor-specific TCR mRNA. We demonstrate that double sequential electroporation of human primary unstimulated T cells with DsiRNA and TCR mRNA leads to unprecedented levels of transgene TCR expression due to a strongly reduced degree of TCR mispairing. Importantly, superior transgenic TCR expression boosts epitope-specific CD8+ T cell activation and killing activity. Altogether, DsiRNA and TCR mRNA double sequential electroporation is a rapid, non-integrating and highly efficient approach with an enhanced biosafety profile to engineer T cells with antigen-specific TCRs for use in early phase clinical trials.
Collapse
Affiliation(s)
- Diana Campillo-Davo
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Johan M J Van den Bergh
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Hans De Reu
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Evelien L J M Smits
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Faculty of Medicine and Health Sciences, Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Division of Clinical Biology, Antwerp University Hospital, Edegem, Belgium
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eva Lion
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Zwi N Berneman
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.,Division of Hematology, Antwerp University Hospital, Edegem, Belgium
| | - Viggo Van Tendeloo
- Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
21
|
Luke GA, Ryan MD. "Therapeutic applications of the 'NPGP' family of viral 2As". Rev Med Virol 2018; 28:e2001. [PMID: 30094875 DOI: 10.1002/rmv.2001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/29/2018] [Accepted: 07/01/2018] [Indexed: 12/15/2022]
Abstract
Oligopeptide "2A" and "2A-like" sequences ("2As"; 18-25aa) are found in a range of RNA virus genomes controlling protein biogenesis through "recoding" of the host-cell translational apparatus. Insertion of multiple 2As within a single open reading frame (ORF) produces multiple proteins; hence, 2As have been used in a very wide range of biotechnological and biomedical applications. During translation, these 2A peptide sequences mediate a eukaryote-specific, self-"cleaving" event, termed "ribosome skipping" with very high efficiency. A particular advantage of using 2As is the ability to simultaneously translate a number of proteins at an equal level in all eukaryotic systems although, naturally, final steady-state levels depend upon other factors-notably protein stability. By contrast, the use of internal ribosome entry site elements for co-expression results in an unbalanced expression due to the relative inefficiency of internal initiation. For example, a 1:1 ratio is of particular importance for the biosynthesis of the heavy-chain and light-chain components of antibodies: highly valuable as therapeutic proteins. Furthermore, each component of these "artificial polyprotein" systems can be independently targeted to different sub-cellular sites. The potential of this system was vividly demonstrated by concatenating multiple gene sequences, linked via 2A sequences, into a single, long, ORF-a polycistronic construct. Here, ORFs comprising the biosynthetic pathways for violacein (five gene sequences) and β-carotene (four gene sequences) were concatenated into a single cistron such that all components were co-expressed in the yeast Pichia pastoris. In this review, we provide useful information on 2As to serve as a guide for future utilities of this co-expression technology in basic research, biotechnology, and clinical applications.
Collapse
Affiliation(s)
- Garry A Luke
- Centre for Biomolecular Sciences, School of Biology, University of St Andrews, St Andrews, UK
| | - Martin D Ryan
- Centre for Biomolecular Sciences, School of Biology, University of St Andrews, St Andrews, UK
| |
Collapse
|
22
|
Chheda ZS, Kohanbash G, Okada K, Jahan N, Sidney J, Pecoraro M, Yang X, Carrera DA, Downey KM, Shrivastav S, Liu S, Lin Y, Lagisetti C, Chuntova P, Watchmaker PB, Mueller S, Pollack IF, Rajalingam R, Carcaboso AM, Mann M, Sette A, Garcia KC, Hou Y, Okada H. Novel and shared neoantigen derived from histone 3 variant H3.3K27M mutation for glioma T cell therapy. J Exp Med 2017; 215:141-157. [PMID: 29203539 PMCID: PMC5748856 DOI: 10.1084/jem.20171046] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/01/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022] Open
Abstract
The median overall survival for children with diffuse intrinsic pontine glioma (DIPG) is less than one year. The majority of diffuse midline gliomas, including more than 70% of DIPGs, harbor an amino acid substitution from lysine (K) to methionine (M) at position 27 of histone 3 variant 3 (H3.3). From a CD8+ T cell clone established by stimulation of HLA-A2+ CD8+ T cells with synthetic peptide encompassing the H3.3K27M mutation, complementary DNA for T cell receptor (TCR) α- and β-chains were cloned into a retroviral vector. TCR-transduced HLA-A2+ T cells efficiently killed HLA-A2+H3.3K27M+ glioma cells in an antigen- and HLA-specific manner. Adoptive transfer of TCR-transduced T cells significantly suppressed the progression of glioma xenografts in mice. Alanine-scanning assays suggested the absence of known human proteins sharing the key amino acid residues required for recognition by the TCR, suggesting that the TCR could be safely used in patients. These data provide us with a strong basis for developing T cell-based therapy targeting this shared neoepitope.
Collapse
Affiliation(s)
- Zinal S Chheda
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA.,Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kaori Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Naznin Jahan
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - John Sidney
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Matteo Pecoraro
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Xinbo Yang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA
| | - Diego A Carrera
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Kira M Downey
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Shruti Shrivastav
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Shuming Liu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Yi Lin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Chetana Lagisetti
- Department of Public Health, University of California, Berkeley, Berkeley, CA
| | - Pavlina Chuntova
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Payal B Watchmaker
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Raja Rajalingam
- Department of Surgery, Immunogenetics and Transplantation Laboratory, University of California, San Francisco, San Francisco, CA
| | | | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Alessandro Sette
- Center for Infectious Disease, Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA.,Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA
| | - Yafei Hou
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA .,Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, CA.,The Parker Institute for Cancer Immunotherapy, San Francisco, CA
| |
Collapse
|
23
|
Ma Y, Kim SS, Kwag DG, Kim SH, Ryu SH, Lee DH, So JH, Lee C, Nam MM, Park JS. Peptides Containing Multiple Disulfide-Bond Mosaic Expression in the Periplasm of Escherichia coli. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Yunqi Ma
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - So-Sun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Dong-Geon Kwag
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Seo-Hyun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Seung-Ho Ryu
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Dong-Hoon Lee
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Jae-Hyeong So
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| | - Chu Lee
- East Sea Fisheries Research Institute; National Fisheries Research and Development Institute; 1194 Haean-ro, Yeongko-myeon, Gangnenung-si Gangwon-do 210-861 Korea
| | - Myung-Mo Nam
- East Sea Fisheries Research Institute; National Fisheries Research and Development Institute; 1194 Haean-ro, Yeongko-myeon, Gangnenung-si Gangwon-do 210-861 Korea
| | - Jang-Su Park
- Department of Chemistry and Chemistry Institute of Functional Materials; Pusan National University; Busan 609-735 Republic of Korea
| |
Collapse
|
24
|
Wang L, Ma N, Okamoto S, Amaishi Y, Sato E, Seo N, Mineno J, Takesako K, Kato T, Shiku H. Efficient tumor regression by adoptively transferred CEA-specific CAR-T cells associated with symptoms of mild cytokine release syndrome. Oncoimmunology 2016; 5:e1211218. [PMID: 27757303 PMCID: PMC5048773 DOI: 10.1080/2162402x.2016.1211218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/29/2016] [Accepted: 07/03/2016] [Indexed: 01/01/2023] Open
Abstract
Carcinoembryonic antigen (CEA) is a cell surface antigen highly expressed in various cancer cell types and in healthy tissues. It has the potential to be a target for chimeric antigen receptor (CAR)-modified T-cell therapy; however, the safety of this approach in terms of on-target/off-tumor effects needs to be determined. To address this issue in a clinically relevant model, we used a mouse model in which the T cells expressing CEA-specific CAR were transferred into tumor-bearing CEA-transgenic (Tg) mice that physiologically expressed CEA as a self-antigen. The adoptive transfer in conjunction with lymphodepleting and myeloablative preconditioning mediated significant tumor regression but caused weight loss in CEA-Tg, but not in wild-type mice. The weight loss was not associated with overt inflammation in the CEA-expressing gastrointestinal tract but was associated with malnutrition, reflected in elevated systemic levels of cytokines linked to anorexia, which could be controlled by the administration of an anti-IL-6 receptor monoclonal antibody without compromising efficacy. The apparent relationship between lymphodepleting and myeloablative preconditioning, efficacy, and off-tumor toxicity of CAR-T cells would necessitate the development of CEA-specific CAR-T cells with improved signaling domains that require less stringent preconditioning for their efficacy. Taken together, these results suggest that CEA-specific CAR-based adoptive T-cell therapy may be effective for patients with CEA+ solid tumors. Distinguishing the fine line between therapeutic efficacy and off-tumor toxicity would involve further modifications of CAR-T cells and preconditioning regimens.
Collapse
Affiliation(s)
- Linan Wang
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine , Tsu, Mie, Japan
| | - Ning Ma
- Faculty of Nursing Science, Suzuka Medical Science University , Suzuka, Mie, Japan
| | | | | | - Eiichi Sato
- Department of Pathology, Institute of Medical Science, Medical Research Center, Tokyo Medical University , Tokyo, Japan
| | - Naohiro Seo
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine , Tsu, Mie, Japan
| | | | - Kazutoh Takesako
- Department of Pathology, Institute of Medical Science, Medical Research Center, Tokyo Medical University , Tokyo, Japan
| | - Takuma Kato
- Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Tsu, Mie, Japan; Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu, Mie, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Tsu, Mie, Japan; Center for Comprehensive Cancer Immunotherapy, Mie University, Tsu, Mie, Japan
| |
Collapse
|
25
|
Casey NP, Fujiwara H, Tanimoto K, Okamoto S, Mineno J, Kuzushima K, Shiku H, Yasukawa M. A Functionally Superior Second-Generation Vector Expressing an Aurora Kinase-A-Specific T-Cell Receptor for Anti-Leukaemia Adoptive Immunotherapy. PLoS One 2016; 11:e0156896. [PMID: 27271876 PMCID: PMC4896450 DOI: 10.1371/journal.pone.0156896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 05/21/2016] [Indexed: 11/23/2022] Open
Abstract
Aurora Kinase A is a cancer-associated protein normally involved in the regulation of mitosis. Being over-expressed in a range of cancers, it is a suitable target for cell-based immunotherapy. Gene transfer of T-cell receptor sequences cognisant of HLA-A*0201-restricted Aurora Kinase A antigen has previously been shown to transfer specific immunoreactivity against the target peptide in a Human Lymphocyte Antigen-restricted manner. While T cell receptor gene-transfer has great potential in overcoming the difficulties of isolating and expanding tumour-reactive lymphocytes from a patient’s own cells, one hurdle is potential mispairing and competition between exogenous and endogenous T cell receptor chains. We have used a retroviral vector design bearing a short-interfering RNA that downregulates endogenous T cell receptor chains, without affecting expression of the transgenic T cell receptor sequences. The T cell receptor expression cassette also includes a 2A self-cleaving peptide, resulting in equimolar expression of the T cell receptor alpha and beta chains, further enhancing formation of the desired T cell receptor. Via a simple, modular cloning method, we have cloned the alpha and beta chains of the anti-Aurora Kinase A-reactive T cell receptor into this ‘siTCR’ vector. We then compared the activity of this vector against the original, ‘conventional’ vector across a panel of assays. T cell receptors expressed from the siTCR-vector retained the cytotoxic functionality of the original vector, with evidence of reduced off-target reactivity. The rate of expression of correctly-formed T cell receptors was superior using the siTCR design, and this was achieved at lower vector copy numbers. Maintaining T cell receptor efficacy with a reduced vector copy number reduces the risk of genotoxicity. The siTCR design also reduces the risk of mispairing and cross-reactivity, while increasing the functional titre. Such improvements in the safety of T cell receptor gene-transfer will be crucial for clinical applications of this technology.
Collapse
Affiliation(s)
- Nicholas Paul Casey
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroshi Fujiwara
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazushi Tanimoto
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| | | | | | | | - Hiroshi Shiku
- Department of Cancer Vaccine and Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Masaki Yasukawa
- Department of Hematology, Clinical Immunology and Infectious Disease, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
26
|
Mancini N, Marrone L, Clementi N, Sautto GA, Clementi M, Burioni R. Adoptive T-cell therapy in the treatment of viral and opportunistic fungal infections. Future Microbiol 2016; 10:665-82. [PMID: 25865200 DOI: 10.2217/fmb.14.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viral infections and opportunistic fungal pathogens represent a major menace for immunocompromised patients. Despite the availability of antifungal and antiviral drugs, mortality in these patients remains high, underlining the need of novel therapeutic options based on completely different strategies. This review describes the potential of several T-cell-based therapeutic approaches in the prophylaxis and treatment of infectious diseases with a particular focus on persistent viral infections and opportunistic fungal infections, as these mostly affect immunocompromised patients.
Collapse
Affiliation(s)
- Nicasio Mancini
- Laboratorio di Microbiologia e Virologia, Università 'Vita-Salute' San Raffaele, DIBIT2, via Olgettina 58, 20132, Milan, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Direct tumor recognition by a human CD4(+) T-cell subset potently mediates tumor growth inhibition and orchestrates anti-tumor immune responses. Sci Rep 2015; 5:14896. [PMID: 26447332 PMCID: PMC4597193 DOI: 10.1038/srep14896] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 09/11/2015] [Indexed: 11/08/2022] Open
Abstract
Tumor antigen-specific CD4(+) T cells generally orchestrate and regulate immune cells to provide immune surveillance against malignancy. However, activation of antigen-specific CD4(+) T cells is restricted at local tumor sites where antigen-presenting cells (APCs) are frequently dysfunctional, which can cause rapid exhaustion of anti-tumor immune responses. Herein, we characterize anti-tumor effects of a unique human CD4(+) helper T-cell subset that directly recognizes the cytoplasmic tumor antigen, NY-ESO-1, presented by MHC class II on cancer cells. Upon direct recognition of cancer cells, tumor-recognizing CD4(+) T cells (TR-CD4) potently induced IFN-γ-dependent growth arrest in cancer cells. In addition, direct recognition of cancer cells triggers TR-CD4 to provide help to NY-ESO-1-specific CD8(+) T cells by enhancing cytotoxic activity, and improving viability and proliferation in the absence of APCs. Notably, the TR-CD4 either alone or in collaboration with CD8(+) T cells significantly inhibited tumor growth in vivo in a xenograft model. Finally, retroviral gene-engineering with T cell receptor (TCR) derived from TR-CD4 produced large numbers of functional TR-CD4. These observations provide mechanistic insights into the role of TR-CD4 in tumor immunity, and suggest that approaches to utilize TR-CD4 will augment anti-tumor immune responses for durable therapeutic efficacy in cancer patients.
Collapse
|
28
|
Askou AL, Aagaard L, Kostic C, Arsenijevic Y, Hollensen AK, Bek T, Jensen TG, Mikkelsen JG, Corydon TJ. Multigenic lentiviral vectors for combined and tissue-specific expression of miRNA- and protein-based antiangiogenic factors. Mol Ther Methods Clin Dev 2015; 2:14064. [PMID: 26052532 PMCID: PMC4449022 DOI: 10.1038/mtm.2014.64] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/14/2014] [Accepted: 12/10/2014] [Indexed: 01/22/2023]
Abstract
Lentivirus-based gene delivery vectors carrying multiple gene cassettes are powerful tools in gene transfer studies and gene therapy, allowing coexpression of multiple therapeutic factors and, if desired, fluorescent reporters. Current strategies to express transgenes and microRNA (miRNA) clusters from a single vector have certain limitations that affect transgene expression levels and/or vector titers. In this study, we describe a novel vector design that facilitates combined expression of therapeutic RNA- and protein-based antiangiogenic factors as well as a fluorescent reporter from back-to-back RNApolII-driven expression cassettes. This configuration allows effective production of intron-embedded miRNAs that are released upon transduction of target cells. Exploiting such multigenic lentiviral vectors, we demonstrate robust miRNA-directed downregulation of vascular endothelial growth factor (VEGF) expression, leading to reduced angiogenesis, and parallel impairment of angiogenic pathways by codelivering the gene encoding pigment epithelium-derived factor (PEDF). Notably, subretinal injections of lentiviral vectors reveal efficient retinal pigment epithelium-specific gene expression driven by the VMD2 promoter, verifying that multigenic lentiviral vectors can be produced with high titers sufficient for in vivo applications. Altogether, our results suggest the potential applicability of combined miRNA- and protein-encoding lentiviral vectors in antiangiogenic gene therapy, including new combination therapies for amelioration of age-related macular degeneration.
Collapse
Affiliation(s)
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Corinne Kostic
- Department of Ophthalmology, Unit of Gene Therapy and Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Yvan Arsenijevic
- Department of Ophthalmology, Unit of Gene Therapy and Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | | | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | |
Collapse
|
29
|
Adoptive immunotherapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors. Pharmaceuticals (Basel) 2014; 7:1049-68. [PMID: 25517545 PMCID: PMC4276906 DOI: 10.3390/ph7121049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 11/26/2014] [Accepted: 12/08/2014] [Indexed: 01/09/2023] Open
Abstract
Accumulating clinical evidence suggests that adoptive T-cell immunotherapy could be a promising option for control of cancer; evident examples include the graft-vs-leukemia effect mediated by donor lymphocyte infusion (DLI) and therapeutic infusion of ex vivo-expanded tumor-infiltrating lymphocytes (TIL) for melanoma. Currently, along with advances in synthetic immunology, gene-modified T cells retargeted to defined tumor antigens have been introduced as “cellular drugs”. As the functional properties of the adoptive immune response mediated by T lymphocytes are decisively regulated by their T-cell receptors (TCRs), transfer of genes encoding target antigen-specific receptors should enable polyclonal T cells to be uniformly redirected toward cancer cells. Clinically, anticancer adoptive immunotherapy using genetically engineered T cells has an impressive track record. Notable examples include the dramatic benefit of chimeric antigen receptor (CAR) gene-modified T cells redirected towards CD19 in patients with B-cell malignancy, and the encouraging results obtained with TCR gene-modified T cells redirected towards NY-ESO-1, a cancer-testis antigen, in patients with advanced melanoma and synovial cell sarcoma. This article overviews the current status of this treatment option, and discusses challenging issues that still restrain the full effectiveness of this strategy, especially in the context of hematological malignancy.
Collapse
|
30
|
RNAi-mediated TCR knockdown prevents autoimmunity in mice caused by mixed TCR dimers following TCR gene transfer. Mol Ther 2014; 22:1983-91. [PMID: 25048215 DOI: 10.1038/mt.2014.142] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 07/17/2014] [Indexed: 12/31/2022] Open
Abstract
Genetically modified T cells that express a transduced T cell receptor (TCR) α/β heterodimer in addition to their endogenous TCR are used in clinical studies to treat cancer. These cells express two TCR-α and two TCR-β chains that do not only compete for CD3 proteins but also form potentially self-reactive mixed TCR dimers, composed of endogenous and transferred chains. To overcome these deficits, we developed an RNAi-TCR replacement vector that simultaneously silences the endogenous TCR and expresses an RNAi-resistant TCR. Transduction of the virus-specific P14 TCR without RNAi resulted in unequal P14 TCR-α and -β chain surface levels, indicating heterodimerization with endogenous TCR chains. Such unequal expression was also observed following TCR gene optimization. Equal surface levels of the introduced TCR chains were however achieved by silencing the endogenous TCR. Importantly, all mice that received cells transduced with the native or optimized P14 TCR developed lethal TCR gene transfer-induced graft-versus-host-disease (TI-GVHD) due to formation of mixed TCR dimers. In contrast, TI-GVHD was almost completely prevented when using the RNAi-TCR replacement vector. Our data demonstrate that RNAi-assisted TCR replacement reduces the formation of mixed TCR dimers, and thereby significantly reduces the risk of TI-GVHD in TCR gene therapy.
Collapse
|
31
|
Adoptive T-cell therapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors. Int J Hematol 2013; 99:123-31. [DOI: 10.1007/s12185-013-1493-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/24/2022]
|
32
|
DcR3 regulates the growth and metastatic potential of SW480 colon cancer cells. Oncol Rep 2013; 30:2741-8. [PMID: 24101127 DOI: 10.3892/or.2013.2769] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/02/2013] [Indexed: 11/05/2022] Open
Abstract
Decoy receptor 3 (DcR3) is considered to have anti‑apoptotic and pro-metastatic functions, suggesting it might be a therapeutic target. We examined the role and mechanisms of DcR3 on growth and the metastatic ability of SW480 colon cancer cells to provide therapeutic information for targeting DcR3 by RNA interference (RNAi) technology. Growth and the metastatic ability were inhibited, apoptosis was induced and cell cycle profile was changed after decreasing DcR3 expression, with lower levels of vascular endothelial growth factors (VEGFs) and matrix metalloproteinases (MMPs) expression. Our results implied the therapeutic potential of silencing DcR3 expression by RNAi in colon cancer.
Collapse
|
33
|
Development of a novel redirected T-cell–based adoptive immunotherapy targeting human telomerase reverse transcriptase for adult T-cell leukemia. Blood 2013; 121:4894-901. [DOI: 10.1182/blood-2012-11-465971] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Key Points
The efficacy and safety of a novel redirected T-cell–based adoptive immunotherapy targeting hTERT for patients with adult T-cell leukemia. hTERT-specific T-cell receptor gene-transduced CD8+ T cells lyse ATL cells, but not normal cells, both in vitro and in vivo.
Collapse
|