1
|
Yao Z, Zeng Y, Liu C, Jin H, Wang H, Zhang Y, Ding C, Chen G, Wu D. Focusing on CD8 + T-cell phenotypes: improving solid tumor therapy. J Exp Clin Cancer Res 2024; 43:266. [PMID: 39342365 PMCID: PMC11437975 DOI: 10.1186/s13046-024-03195-5] [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: 07/31/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Vigorous CD8+ T cells play a crucial role in recognizing tumor cells and combating solid tumors. How T cells efficiently recognize and target tumor antigens, and how they maintain the activity in the "rejection" of solid tumor microenvironment, are major concerns. Recent advances in understanding of the immunological trajectory and lifespan of CD8+ T cells have provided guidance for the design of more optimal anti-tumor immunotherapy regimens. Here, we review the newly discovered methods to enhance the function of CD8+ T cells against solid tumors, focusing on optimizing T cell receptor (TCR) expression, improving antigen recognition by engineered T cells, enhancing signal transduction of the TCR-CD3 complex, inducing the homing of polyclonal functional T cells to tumors, reversing T cell exhaustion under chronic antigen stimulation, and reprogramming the energy and metabolic pathways of T cells. We also discuss how to participate in the epigenetic changes of CD8+ T cells to regulate two key indicators of anti-tumor responses, namely effectiveness and persistence.
Collapse
Affiliation(s)
- Zhouchi Yao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yayun Zeng
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Cheng Liu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huimin Jin
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hong Wang
- Department of Scientific Research, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121001, China
| | - Yue Zhang
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Chengming Ding
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Guodong Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Daichao Wu
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Laboratory of Structural Immunology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
2
|
Yang D, Duan Z, Yuan P, Ding C, Dai X, Chen G, Wu D. How does TCR-T cell therapy exhibit a superior anti-tumor efficacy. Biochem Biophys Res Commun 2023; 687:149209. [PMID: 37944471 DOI: 10.1016/j.bbrc.2023.149209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/26/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
TCR-engineered T cells have achieved great progress in solid tumor therapy, some of which have been applicated in clinical trials. Deep knowledge about the current progress of TCR-T in tumor therapy would be beneficial to understand the direction. Here, we classify tumor antigens into tumor-associated antigens, tumor-specific antigens, tumor antigens expressed by oncogenic viruses, and tumor antigens caused by abnormal protein modification; Then we detail the TCR-T cell therapy effects targeting those tumor antigens in clinical or preclinical trials, and propose that neoantigen specific TCR-T cell therapy is expected to be a promising approach for solid tumors; Furthermore, we summarize the optimization strategies, such as tumor microenvironment, TCR pairing and affinity, to improve the therapeutic effect of TCR-T. Overall, this review provides inspiration for the antigen selection and therapy strategies of TCR-T in the future.
Collapse
Affiliation(s)
- Dandan Yang
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhihui Duan
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ping Yuan
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Chengming Ding
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoming Dai
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guodong Chen
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
3
|
Zhang P, Zhang G, Wan X. Challenges and new technologies in adoptive cell therapy. J Hematol Oncol 2023; 16:97. [PMID: 37596653 PMCID: PMC10439661 DOI: 10.1186/s13045-023-01492-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
Adoptive cell therapies (ACTs) have existed for decades. From the initial infusion of tumor-infiltrating lymphocytes to the subsequent specific enhanced T cell receptor (TCR)-T and chimeric antigen receptor (CAR)-T cell therapies, many novel strategies for cancer treatment have been developed. Owing to its promising outcomes, CAR-T cell therapy has revolutionized the field of ACTs, particularly for hematologic malignancies. Despite these advances, CAR-T cell therapy still has limitations in both autologous and allogeneic settings, including practicality and toxicity issues. To overcome these challenges, researchers have focused on the application of CAR engineering technology to other types of immune cell engineering. Consequently, several new cell therapies based on CAR technology have been developed, including CAR-NK, CAR-macrophage, CAR-γδT, and CAR-NKT. In this review, we describe the development, advantages, and possible challenges of the aforementioned ACTs and discuss current strategies aimed at maximizing the therapeutic potential of ACTs. We also provide an overview of the various gene transduction strategies employed in immunotherapy given their importance in immune cell engineering. Furthermore, we discuss the possibility that strategies capable of creating a positive feedback immune circuit, as healthy immune systems do, could address the flaw of a single type of ACT, and thus serve as key players in future cancer immunotherapy.
Collapse
Affiliation(s)
- Pengchao Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China.
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
4
|
Du S, Yan J, Xue Y, Zhong Y, Dong Y. Adoptive cell therapy for cancer treatment. EXPLORATION (BEIJING, CHINA) 2023; 3:20210058. [PMID: 37933232 PMCID: PMC10624386 DOI: 10.1002/exp.20210058] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/17/2023] [Indexed: 11/08/2023]
Abstract
Adoptive cell therapy (ACT) is a rapidly growing anti-cancer strategy that has shown promise in treating various cancer types. The concept of ACT involves activating patients' own immune cells ex vivo and then transferring them back to the patients to recognize and eliminate cancer cells. Currently, the commonly used ACT includes tumor-infiltrating lymphocytes (TILs), genetically engineered immune cells, and dendritic cells (DCs) vaccines. With the advancement of cell culture and genetic engineering techniques, ACT has been used in clinics to treat malignant hematological diseases and many new ACT-based regimens are in different stages of clinical trials. Here, representative ACT approaches are introduced and the opportunities and challenges for clinical translation of ACT are discussed.
Collapse
Affiliation(s)
- Shi Du
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Jingyue Yan
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yonger Xue
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yichen Zhong
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yizhou Dong
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| |
Collapse
|
5
|
Wei F, Cheng XX, Xue JZ, Xue SA. Emerging Strategies in TCR-Engineered T Cells. Front Immunol 2022; 13:850358. [PMID: 35432319 PMCID: PMC9006933 DOI: 10.3389/fimmu.2022.850358] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy of cancer has made tremendous progress in recent years, as demonstrated by the remarkable clinical responses obtained from adoptive cell transfer (ACT) of patient-derived tumor infiltrating lymphocytes, chimeric antigen receptor (CAR)-modified T cells (CAR-T) and T cell receptor (TCR)-engineered T cells (TCR-T). TCR-T uses specific TCRS optimized for tumor engagement and can recognize epitopes derived from both cell-surface and intracellular targets, including tumor-associated antigens, cancer germline antigens, viral oncoproteins, and tumor-specific neoantigens (neoAgs) that are largely sequestered in the cytoplasm and nucleus of tumor cells. Moreover, as TCRS are naturally developed for sensitive antigen detection, they are able to recognize epitopes at far lower concentrations than required for CAR-T activation. Therefore, TCR-T holds great promise for the treatment of human cancers. In this focused review, we summarize basic, translational, and clinical insights into the challenges and opportunities of TCR-T. We review emerging strategies used in current ACT, point out limitations, and propose possible solutions. We highlight the importance of targeting tumor-specific neoAgs and outline a strategy of combining neoAg vaccines, checkpoint blockade therapy, and adoptive transfer of neoAg-specific TCR-T to produce a truly tumor-specific therapy, which is able to penetrate into solid tumors and resist the immunosuppressive tumor microenvironment. We believe such a combination approach should lead to a significant improvement in cancer immunotherapies, especially for solid tumors, and may provide a general strategy for the eradication of multiple cancers.
Collapse
Affiliation(s)
- Fang Wei
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - Xiao-Xia Cheng
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - John Zhao Xue
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| | - Shao-An Xue
- Genetic Engineering Laboratory, School of Biological & Environmental Engineering, Xi'An University, Xi'An, China
| |
Collapse
|
6
|
Liu Y, Yan X, Zhang F, Zhang X, Tang F, Han Z, Li Y. TCR-T Immunotherapy: The Challenges and Solutions. Front Oncol 2022; 11:794183. [PMID: 35145905 PMCID: PMC8822241 DOI: 10.3389/fonc.2021.794183] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/28/2021] [Indexed: 12/31/2022] Open
Abstract
T cell receptor-engineered T cell (TCR-T) therapy is free from the limit of surface antigen expression of the target cells, which is a potential cellular immunotherapy for cancer treatment. Significant advances in the treatment of hematologic malignancies with cellular immunotherapy have aroused the interest of researchers in the treatment of solid tumors. Nevertheless, the overall efficacy of TCR-T cell immunotherapy in solid tumors was not significantly high when compared with hematological malignancies. In this article, we pay attention to the barriers of TCR-T cell immunotherapy for solid tumors, as well as the strategies affecting the efficacy of TCR-T cell immunotherapy. To provide some reference for researchers to better overcome the impact of TCR-T cell efficiency in solid tumors.
Collapse
Affiliation(s)
- Yating Liu
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Xin Yan
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Fan Zhang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaoxia Zhang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhijian Han
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yumin Li
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Yumin Li,
| |
Collapse
|
7
|
Chen L, Dong L, Ma Y, Wang J, Qiao D, Tian G, Wang M. An efficient method to identify virus-specific TCRs for TCR-T cell immunotherapy against virus-associated malignancies. BMC Immunol 2021; 22:65. [PMID: 34583647 PMCID: PMC8480097 DOI: 10.1186/s12865-021-00455-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 11/21/2022] Open
Abstract
Adoptive transfer of T cells genetically engineered with a T cell receptor (TCR) is a promising cancer treatment modality that requires the identification of TCRs with good characteristics. Most T cell cloning methods involve a stringent singularization process, which necessitates either tedious hands-on operations or high cost. We present an efficient and nonstringent cloning approach based on existing techniques. We hypothesize that after elimination of most nonspecific T cells, a clonotype with high quality could outcompete other clonotypes and finally form a predominant population. This TCR identification method can be used to clone virus-specific TCRs efficiently from cancer patients and is easily adoptable by any laboratory.
Collapse
Affiliation(s)
- Lei Chen
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Lianhua Dong
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Yipeng Ma
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Juntao Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Dongjuan Qiao
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China
| | - Geng Tian
- Department of Oncology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Mingjun Wang
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen International Biological Valley-Life Science Industrial Park, Dapeng New District, Shenzhen, China.
| |
Collapse
|
8
|
Sun Y, Li F, Sonnemann H, Jackson KR, Talukder AH, Katailiha AS, Lizee G. Evolution of CD8 + T Cell Receptor (TCR) Engineered Therapies for the Treatment of Cancer. Cells 2021; 10:cells10092379. [PMID: 34572028 PMCID: PMC8469972 DOI: 10.3390/cells10092379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
Engineered T cell receptor T (TCR-T) cell therapy has facilitated the generation of increasingly reliable tumor antigen-specific adaptable cellular products for the treatment of human cancer. TCR-T cell therapies were initially focused on targeting shared tumor-associated peptide targets, including melanoma differentiation and cancer-testis antigens. With recent technological developments, it has become feasible to target neoantigens derived from tumor somatic mutations, which represents a highly personalized therapy, since most neoantigens are patient-specific and are rarely shared between patients. TCR-T therapies have been tested for clinical efficacy in treating solid tumors in many preclinical studies and clinical trials all over the world. However, the efficacy of TCR-T therapy for the treatment of solid tumors has been limited by a number of factors, including low TCR avidity, off-target toxicities, and target antigen loss leading to tumor escape. In this review, we discuss the process of deriving tumor antigen-specific TCRs, including the identification of appropriate tumor antigen targets, expansion of antigen-specific T cells, and TCR cloning and validation, including techniques and tools for TCR-T cell vector construction and expression. We highlight the achievements of recent clinical trials of engineered TCR-T cell therapies and discuss the current challenges and potential solutions for improving their safety and efficacy, insights that may help guide future TCR-T studies in cancer.
Collapse
Affiliation(s)
- Yimo Sun
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Fenge Li
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Heather Sonnemann
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Kyle R. Jackson
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Amjad H. Talukder
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Arjun S. Katailiha
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
| | - Gregory Lizee
- Department of Melanoma, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (Y.S.); (F.L.); (H.S.); (K.R.J.); (A.H.T.); (A.S.K.)
- Department of Immunology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
9
|
Schober K, Müller TR, Busch DH. Orthotopic T-Cell Receptor Replacement-An "Enabler" for TCR-Based Therapies. Cells 2020; 9:E1367. [PMID: 32492858 PMCID: PMC7348731 DOI: 10.3390/cells9061367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/25/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022] Open
Abstract
Natural adaptive immunity co-evolved with pathogens over millions of years, and adoptive transfer of non-engineered T cells to fight infections or cancer so far exhibits an exceptionally safe and functional therapeutic profile in clinical trials. However, the personalized nature of therapies using virus-specific T cells, donor lymphocyte infusion, or tumor-infiltrating lymphocytes makes implementation in routine clinical care difficult. In principle, genetic engineering can be used to make T-cell therapies more broadly applicable, but so far it significantly alters the physiology of cells. We recently demonstrated that orthotopic T-cell receptor (TCR) replacement (OTR) by clustered regularly interspaced short palindromic repeats (CRISPR)/ CRISPR-associated protein 9 (Cas9) can be used to generate engineered T cells with preservation of near-physiological function. In this review, we present the current status of OTR technology development and discuss its potential for TCR-based therapies. By providing the means to combine the therapeutic efficacy and safety profile of physiological T cells with the versatility of cell engineering, OTR can serve as an "enabler" for TCR-based therapies.
Collapse
Affiliation(s)
- Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Thomas R. Müller
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| |
Collapse
|
10
|
Malviya M, Saoudi A, Bauer J, Fillatreau S, Liblau R. Treatment of experimental autoimmune encephalomyelitis with engineered bi-specific Foxp3+ regulatory CD4+ T cells. J Autoimmun 2020; 108:102401. [PMID: 31948790 DOI: 10.1016/j.jaut.2020.102401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/01/2020] [Indexed: 12/22/2022]
Abstract
The use of autoantigen-specific regulatory T cells (Tregs) as a cellular therapy for autoimmune diseases is appealing. However, it is challenging to isolate and expand large quantity of Tregs expressing disease-relevant T-cell receptors (TCR). To overcome this problem, we used an approach aiming at redirecting the specificity of polyclonal Tregs through autoreactive TCR gene transfer technology. In this study, we examined whether Tregs engineered through retroviral transduction to express a TCR cross-reactive to two CNS autoantigens, myelin oligodendrocyte glycoprotein (MOG) and neurofilament-medium (NF-M), had a superior protective efficacy compared with Tregs expressing a MOG mono-specific TCR. We observed that engineered Tregs (engTregs) exhibited in vitro regulatory effects related to the antigenic specificity of the introduced TCR, and commensurate in potency with the avidity of the transduced TCR. In experimental autoimmune encephalomyelitis (EAE), adoptively transferred engTregs proliferated, and migrated to the CNS, while retaining FoxP3 expression. EngTregs expressing MOG/NF-M cross-reactive TCR had superior protective properties over engTregs expressing MOG-specific TCR in MOG-induced EAE. Remarkably, MOG/NF-M bi-specific TCR-engTregs also improved recovery from EAE induced by an unrelated CNS autoantigen, proteolipid protein (PLP). This study underlines the benefit of using TCRs cross-reacting towards multiple autoantigens, compared with mono-reactive TCR, for the generation of engTregs affording protection from autoimmune disease in adoptive cell therapy.
Collapse
Affiliation(s)
- Manish Malviya
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier (UPS), Toulouse, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier (UPS), Toulouse, France
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, A-1090, Austria
| | - Simon Fillatreau
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, Université Paris Descartes, Sorbonne Paris Cité, Bâtiment Leriche, 75993, Paris, France; AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Roland Liblau
- Centre de Physiopathologie Toulouse-Purpan (CPTP), Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Université Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Adoptive cell therapy using CD4FOXP3 regulatory T cells (Treg) has emerged as a promising therapeutic strategy to treat autoimmunity and alloimmunity. Preclinical studies suggest that the efficacy of Treg therapy can be improved by modifying the antigen specificity, stability and function of therapeutic Tregs. We review recent innovations that considerably enhance the possibilities of controlling these parameters. RECENT FINDINGS Antigen-specific Tregs can be generated by genetically modifying polyclonal Tregs to express designated T-cell receptors or single-chain chimeric antigen receptors. The benefits of this approach can be further extended by using novel strategies to fine-tune the antigen-specificity and affinity of Treg in vivo. CRISPR/Cas 9 technology now enables the modification of therapeutic Tregs so they are safer, more stable and long lived. The differentiation and homing properties of Tregs can also be modulated by gene editing or modifying ex-vivo stimulation conditions. SUMMARY A new wave of innovation has considerably increased the number of strategies that could be used to increase the therapeutic potential of Treg therapy. However, the increased complexity of these approaches may limit their wide accessibility. Third-party therapy with off-the-shelf Treg products could be a solution.
Collapse
|
12
|
Schober K, Müller TR, Gökmen F, Grassmann S, Effenberger M, Poltorak M, Stemberger C, Schumann K, Roth TL, Marson A, Busch DH. Orthotopic replacement of T-cell receptor α- and β-chains with preservation of near-physiological T-cell function. Nat Biomed Eng 2019; 3:974-984. [DOI: 10.1038/s41551-019-0409-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
|
13
|
Tendeiro Rego R, Morris EC, Lowdell MW. T-cell receptor gene-modified cells: past promises, present methodologies and future challenges. Cytotherapy 2019; 21:341-357. [PMID: 30655164 DOI: 10.1016/j.jcyt.2018.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Immunotherapy constitutes an exciting and rapidly evolving field, and the demonstration that genetically modified T-cell receptors (TCRs) can be used to produce T-lymphocyte populations of desired specificity offers new opportunities for antigen-specific T-cell therapy. Overall, TCR-modified T cells have the ability to target a wide variety of self and non-self targets through the normal biology of a T cell. Although major histocompatibility complex (MHC)-restricted and dependent on co-receptors, genetically engineered TCRs still present a number of characteristics that ensure they are an important alternative strategy to chimeric antigen receptors (CARs), and high-affinity TCRs can now be successfully engineered with the potential to enhance therapeutic efficacy while minimizing adverse events. This review will focus on the main characteristics of TCR gene-modified cells, their potential clinical application and promise to the field of adoptive cell transfer (ACT), basic manufacturing procedures and characterization protocols and overall challenges that need to be overcome so that redirection of TCR specificity may be successfully translated into clinical practice, beyond early-phase clinical trials.
Collapse
Affiliation(s)
- Rita Tendeiro Rego
- UCL Institute of Immunity and Transplantation, London, UK; Centre for Cell, Gene & Tissue Therapeutics, Royal Free London NHS Foundation Trust, London, UK
| | - Emma C Morris
- UCL Institute of Immunity and Transplantation, London, UK
| | - Mark W Lowdell
- UCL Cancer Institute, Department of Haematology, London, UK
| |
Collapse
|
14
|
Echchannaoui H, Petschenka J, Ferreira EA, Hauptrock B, Lotz-Jenne C, Voss RH, Theobald M. A Potent Tumor-Reactive p53-Specific Single-Chain TCR without On- or Off-Target Autoimmunity In Vivo. Mol Ther 2018; 27:261-271. [PMID: 30528087 DOI: 10.1016/j.ymthe.2018.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/25/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Genetic engineering of T cells with a T cell receptor (TCR) targeting tumor antigen is a promising strategy for cancer immunotherapy. Inefficient expression of the introduced TCR due to TCR mispairing may limit the efficacy and adversely affect the safety of TCR gene therapy. Here, we evaluated the safety and therapeutic efficiency of an optimized single-chain TCR (scTCR) specific for an HLA-A2.1-restricted (non-mutated) p53(264-272) peptide in adoptive T cell transfer (ACT) models using our unique transgenic mice expressing human p53 and HLA-A2.1 that closely mimic the human setting. Specifically, we showed that adoptive transfer of optimized scTCR-redirected T cells does not induce on-target and off-target autoimmunity. Furthermore, ACT resulted in full tumor protection and led to a long-lived effective, antigen-specific memory T cell response in syngeneic and xenograft models. Taken together, the study demonstrated that our scTCR specific for the broadly expressed tumor-associated antigen p53(264-272) can eradicate p53+A2.1+ tumor cells without inducing off-target or self-directed toxicities in mouse models of ACT. These data strongly support the improved safety and therapeutic efficacy of high-affinity p53scTCR for TCR-based immunotherapy of p53-associated malignancies.
Collapse
Affiliation(s)
- Hakim Echchannaoui
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center (UMC), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany; German Consortium for Translational Cancer Research (DKTK), Frankfurt/Mainz, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jutta Petschenka
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Edite Antunes Ferreira
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Beate Hauptrock
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Carina Lotz-Jenne
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Ralf-Holger Voss
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology, and Pneumology, University Medical Center (UMC) and University Cancer Center (UCT), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center (UMC), Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany; German Consortium for Translational Cancer Research (DKTK), Frankfurt/Mainz, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
15
|
Hull CM, Nickolay LE, Estorninho M, Richardson MW, Riley JL, Peakman M, Maher J, Tree TI. Generation of human islet-specific regulatory T cells by TCR gene transfer. J Autoimmun 2017; 79:63-73. [DOI: 10.1016/j.jaut.2017.01.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 01/12/2023]
|
16
|
A High-avidity WT1-reactive T-Cell Receptor Mediates Recognition of Peptide and Processed Antigen but not Naturally Occurring WT1-positive Tumor Cells. J Immunother 2016; 39:105-16. [PMID: 26938944 DOI: 10.1097/cji.0000000000000116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Wilms tumor gene 1 (WT1) is an attractive target antigen for cancer immunotherapy because it is overexpressed in many hematologic malignancies and solid tumors but has limited, low-level expression in normal adult tissues. Multiple HLA class I and class II restricted epitopes have been identified in WT1, and multiple investigators are pursuing the treatment of cancer patients with WT1-based vaccines and adoptively transferred WT1-reactive T cells. Here we isolated an HLA-A*0201-restricted WT1-reactive T-cell receptor (TCR) by stimulating peripheral blood lymphocytes of healthy donors with the peptide WT1:126-134 in vitro. This TCR mediated peptide recognition down to a concentration of ∼0.1 ng/mL when pulsed onto T2 cells as well as recognition of HLA-A*0201 target cells transfected with full-length WT1 cDNA. However, it did not mediate consistent recognition of many HLA-A*0201 tumor cell lines or freshly isolated leukemia cells that endogeneously expressed WT1. We dissected this pattern of recognition further and observed that WT1:126-134 was more efficiently processed by immunoproteasomes compared with standard proteasomes. However, pretreatment of WT1 tumor cell lines with interferon gamma did not appreciably enhance recognition by our TCR. In addition, we highly overexpressed WT1 in several leukemia cell lines by electroporation with full-length WT1 cDNA. Some of these lines were still not recognized by our TCR suggesting possible antigen processing defects in some leukemias. These results suggest WT1:126-134 may not be a suitable target for T-cell based tumor immunotherapies.
Collapse
|
17
|
Bethune MT, Gee MH, Bunse M, Lee MS, Gschweng EH, Pagadala MS, Zhou J, Cheng D, Heath JR, Kohn DB, Kuhns MS, Uckert W, Baltimore D. Domain-swapped T cell receptors improve the safety of TCR gene therapy. eLife 2016; 5. [PMID: 27823582 PMCID: PMC5101000 DOI: 10.7554/elife.19095] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/11/2016] [Indexed: 12/17/2022] Open
Abstract
T cells engineered to express a tumor-specific αβ T cell receptor (TCR) mediate anti-tumor immunity. However, mispairing of the therapeutic αβ chains with endogenous αβ chains reduces therapeutic TCR surface expression and generates self-reactive TCRs. We report a general strategy to prevent TCR mispairing: swapping constant domains between the α and β chains of a therapeutic TCR. When paired, domain-swapped (ds)TCRs assemble with CD3, express on the cell surface, and mediate antigen-specific T cell responses. By contrast, dsTCR chains mispaired with endogenous chains cannot properly assemble with CD3 or signal, preventing autoimmunity. We validate this approach in cell-based assays and in a mouse model of TCR gene transfer-induced graft-versus-host disease. We also validate a related approach whereby replacement of αβ TCR domains with corresponding γδ TCR domains yields a functional TCR that does not mispair. This work enables the design of safer TCR gene therapies for cancer immunotherapy. DOI:http://dx.doi.org/10.7554/eLife.19095.001 T cells enable the immune system to recognize invading microbes and diseased cells while ignoring healthy cells. The ability of a T cell to recognize a specific microbe or diseased cell is determined by two proteins that pair to form its “T cell receptor.” The paired receptors are exported to the surface of the T cell, where they bind to infected or cancerous cells. Those T cells that produce receptors that bind healthy cells are eliminated during development. T cells can generally distinguish between the body’s own cells and the cells of invading bacteria or other microbes. However, cancer cells are more difficult to identify because they are similar to healthy cells. Efforts to develop therapies that enhance the immune system’s ability to recognize cancer cells have had only limited success. One successful approach – known as T cell receptor gene therapy – modifies T cells to destroy cancer cells by arming them with a cancer-specific T cell receptor. This technique produces T cells possessing two T cell receptors – the cancer-specific receptor and the one it had originally – so it is possible for proteins from the two receptors to mispair. This impedes the correct pairing of the cancer-specific T cell receptor, reducing the effectiveness of the therapy. More importantly, mispaired T cell receptors may cause the immune cells to attack healthy cells in the body, leading to autoimmune disease. To make T cell receptor gene therapy safe, the cancer-specific receptor must not mispair with the resident receptor. Here, Bethune et al. describe a new strategy to prevent T cell receptors from mispairing. The researchers altered the arrangement of particular regions in a cancer-specific T cell receptor to make a new receptor called a domain-swapped T cell receptor (dsTCR). Like normal T cell receptors, the dsTCRs were exported to the T cell surface and were able to interact with other proteins involved in immune responses. Furthermore, T cells armed with dsTCRs were able to kill cancer cells and prevent tumor growth in mice. Unlike other cancer-specific receptors, dsTCRs did not mispair with the resident T cell receptors in mouse or human cells, and did not cause autoimmune disease in mice. The findings of Bethune et al. show that the structure of the T cell receptor is unexpectedly robust, in that it still works even if it is modified. The next step is to study dsTCRs in more detail with the aim of optimizing them so that they might be used in human clinical trials in the future. DOI:http://dx.doi.org/10.7554/eLife.19095.002
Collapse
Affiliation(s)
- Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Marvin H Gee
- Program in Immunology, Stanford University School of Medicine, Stanford, United States
| | - Mario Bunse
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Mark S Lee
- Department of Immunobiology, University of Arizona, Tucson, United States.,The BIO5 Institute, University of Arizona, Tucson, United States
| | - Eric H Gschweng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Meghana S Pagadala
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Jing Zhou
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, United States
| | - Donghui Cheng
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, United States
| | - James R Heath
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, United States
| | - Donald B Kohn
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Michael S Kuhns
- Department of Immunobiology, University of Arizona, Tucson, United States.,The BIO5 Institute, University of Arizona, Tucson, United States
| | - Wolfgang Uckert
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
18
|
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
|
19
|
Holler A, Zech M, Ghorashian S, Pike R, Hotblack A, Veliça P, Xue SA, Chakraverty R, Morris EC, Stauss HJ. Expression of a dominant T-cell receptor can reduce toxicity and enhance tumor protection of allogeneic T-cell therapy. Haematologica 2016; 101:482-90. [PMID: 26802053 PMCID: PMC5004405 DOI: 10.3324/haematol.2015.132712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 01/13/2016] [Indexed: 11/09/2022] Open
Abstract
Due to the lack of specificity for tumor antigens, allogeneic T-cell therapy is associated with graft-versus-host disease. Enhancing the anti-tumor specificity while reducing the graft-versus-host disease risk of allogeneic T cells has remained a research focus. In this study, we demonstrate that the introduction of 'dominant' T-cell receptors into primary murine T cells can suppress the expression of endogenous T-cell receptors in a large proportion of the gene-modified T cells. Adoptive transfer of allogeneic T cells expressing a 'dominant' T-cell receptor significantly reduced the graft-versus-host toxicity in recipient mice. Using two bone marrow transplant models, enhanced anti-tumor activity was observed in the presence of reduced graft-versus-host disease. However, although transfer of T-cell receptor gene-modified allogeneic T cells resulted in the elimination of antigen-positive tumor cells and improved the survival of treated mice, it was associated with accumulation of T cells expressing endogenous T-cell receptors and the development of delayed graft-versus-host disease. The in-vivo deletion of the engineered T cells, mediated by endogenous mouse mammary tumor virus MTV8 and MTV9, abolished graft-versus-host disease while retaining significant anti-tumor activity of adoptively transferred T cells. Together, this study shows that the in-vitro selection of allogeneic T cells expressing high levels of a 'dominant' T-cell receptor can lower acute graft-versus-host disease and enhance anti-tumor activity of adoptive cell therapy, while the in-vivo outgrowth of T cells expressing endogenous T-cell receptors remains a risk factor for the delayed onset of graft-versus-host disease.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation/methods
- Cell Line, Tumor
- Female
- Gene Expression
- Genes, Dominant
- Genetic Vectors/immunology
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/pathology
- Graft vs Host Disease/prevention & control
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocyte Depletion/methods
- Mammary Tumor Virus, Mouse/genetics
- Mammary Tumor Virus, Mouse/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Survival Analysis
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transgenes
- Transplantation, Homologous
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Angelika Holler
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Mathias Zech
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Sara Ghorashian
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Rebecca Pike
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Alastair Hotblack
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Pedro Veliça
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Shao-An Xue
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Ronjon Chakraverty
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London Department of Haematology, Cancer Institute, University College London, UK
| | - Emma C Morris
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| | - Hans J Stauss
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, University College London, Royal Free Hospital London
| |
Collapse
|
20
|
Nicholson E, Peggs KS. Cytomegalovirus-specific T-cell therapies: current status and future prospects. Immunotherapy 2015; 7:135-46. [PMID: 25713989 DOI: 10.2217/imt.14.99] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adoptive transfer of T cells specific for viral pathogens offers an attractive method for hastening immune reconstitution and protective immunity in patients following stem cell transplantation. The largest experience to date has been in the context of treatment or prevention of cytomegalovirus or Epstein-Barr virus. A number of technical hurdles have now been overcome allowing consideration of more widespread application of products compliant with Good Manufacturing Practice regulations, and of the development of commercialization pathways for these products. This review summarizes progress to date and highlights some of the areas that remain problematic and that require further innovation and evaluation before more widespread adoption is considered.
Collapse
Affiliation(s)
- Emma Nicholson
- Department of Haematology, University College London Hospital, London, NW1 2BU, UK
| | | |
Collapse
|
21
|
Gilham DE, Anderson J, Bridgeman JS, Hawkins RE, Exley MA, Stauss H, Maher J, Pule M, Sewell AK, Bendle G, Lee S, Qasim W, Thrasher A, Morris E. Adoptive T-cell therapy for cancer in the United kingdom: a review of activity for the British Society of Gene and Cell Therapy annual meeting 2015. Hum Gene Ther 2015; 26:276-85. [PMID: 25860661 PMCID: PMC4442586 DOI: 10.1089/hum.2015.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/31/2015] [Indexed: 12/25/2022] Open
Abstract
Adoptive T-cell therapy is delivering objective clinical responses across a number of cancer indications in the early phase clinical setting. Much of this clinical activity is taking place at major clinical academic centers across the United States. This review focuses upon cancer-focused cell therapy activity within the United Kingdom as a contribution to the 2015 British Society of Gene and Cell Therapy annual general meeting. This overview reflects the diversity and expansion of clinical and preclinical studies within the United Kingdom while considering the background context of this work against new infrastructural developments and the requirements of nationalized healthcare delivery within the UK National Health Service.
Collapse
Affiliation(s)
- David Edward Gilham
- Clinical and Experimental Immunotherapy Group, Institute of Cancer Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - John Anderson
- UCL Institute of Child Health, London WC1N 1EH, United Kingdom
| | | | - Robert Edward Hawkins
- Clinical and Experimental Immunotherapy Group, Institute of Cancer Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
- Cellular Therapeutics Ltd., UMIC Bio-incubator, Manchester M13 9XX, United Kingdom
| | - Mark Adrian Exley
- MCCIR, Faculty of Medicine, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Hans Stauss
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - John Maher
- Department of Research Oncology, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Martin Pule
- Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, United Kingdom
| | - Andrew Kelvin Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, United Kingdom
| | - Gavin Bendle
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Steven Lee
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Waseem Qasim
- Molecular and Cellular Immunology Institute, Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Adrian Thrasher
- Molecular and Cellular Immunology Institute, Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Emma Morris
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| |
Collapse
|
22
|
Sharpe M, Mount N. Genetically modified T cells in cancer therapy: opportunities and challenges. Dis Model Mech 2015; 8:337-50. [PMID: 26035842 PMCID: PMC4381333 DOI: 10.1242/dmm.018036] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tumours use many strategies to evade the host immune response, including downregulation or weak immunogenicity of target antigens and creation of an immune-suppressive tumour environment. T cells play a key role in cell-mediated immunity and, recently, strategies to genetically modify T cells either through altering the specificity of the T cell receptor (TCR) or through introducing antibody-like recognition in chimeric antigen receptors (CARs) have made substantial advances. The potential of these approaches has been demonstrated in particular by the successful use of genetically modified T cells to treat B cell haematological malignancies in clinical trials. This clinical success is reflected in the growing number of strategic partnerships in this area that have attracted a high level of investment and involve large pharmaceutical organisations. Although our understanding of the factors that influence the safety and efficacy of these therapies has increased, challenges for bringing genetically modified T-cell immunotherapy to many patients with different tumour types remain. These challenges range from the selection of antigen targets and dealing with regulatory and safety issues to successfully navigating the routes to commercial development. However, the encouraging clinical data, the progress in the scientific understanding of tumour immunology and the improvements in the manufacture of cell products are all advancing the clinical translation of these important cellular immunotherapies.
Collapse
Affiliation(s)
- Michaela Sharpe
- Cell Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Natalie Mount
- Cell Therapy Catapult, 12th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
23
|
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: 1.9] [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
|
24
|
Oren R, Hod-Marco M, Haus-Cohen M, Thomas S, Blat D, Duvshani N, Denkberg G, Elbaz Y, Benchetrit F, Eshhar Z, Stauss H, Reiter Y. Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody-based chimeric antigen receptors indicates affinity/avidity thresholds. THE JOURNAL OF IMMUNOLOGY 2014; 193:5733-43. [PMID: 25362181 DOI: 10.4049/jimmunol.1301769] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adoptive transfer of Ag-specific T lymphocytes is an attractive form of immunotherapy for cancers. However, acquiring sufficient numbers of host-derived tumor-specific T lymphocytes by selection and expansion is challenging, as these cells may be rare or anergic. Using engineered T cells can overcome this difficulty. Such engineered cells can be generated using a chimeric Ag receptor based on common formats composed from Ag-recognition elements such as αβ-TCR genes with the desired specificity, or Ab variable domain fragments fused with T cell-signaling moieties. Combining these recognition elements are Abs that recognize peptide-MHC. Such TCR-like Abs mimic the fine specificity of TCRs and exhibit both the binding properties and kinetics of high-affinity Abs. In this study, we compared the functional properties of engineered T cells expressing a native low affinity αβ-TCR chains or high affinity TCR-like Ab-based CAR targeting the same specificity. We isolated high-affinity TCR-like Abs recognizing HLA-A2-WT1Db126 complexes and constructed CAR that was transduced into T cells. Comparative analysis revealed major differences in function and specificity of such CAR-T cells or native TCR toward the same antigenic complex. Whereas the native low-affinity αβ-TCR maintained potent cytotoxic activity and specificity, the high-affinity TCR-like Ab CAR exhibited reduced activity and loss of specificity. These results suggest an upper affinity threshold for TCR-based recognition to mediate effective functional outcomes of engineered T cells. The rational design of TCRs and TCR-based constructs may need to be optimized up to a given affinity threshold to achieve optimal T cell function.
Collapse
Affiliation(s)
- Ravit Oren
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Moran Hod-Marco
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Maya Haus-Cohen
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Sharyn Thomas
- Department of Immunology, Institute of Immunity, Infection and Transplantation, Royal Free Hospital, University College London, London NW3 2PF, United Kingdom
| | - Dan Blat
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel; and
| | - Nerri Duvshani
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | | - Yael Elbaz
- Applied Immune Technologies, Haifa 32000, Israel
| | | | - Zelig Eshhar
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel; and
| | - Hans Stauss
- Department of Immunology, Institute of Immunity, Infection and Transplantation, Royal Free Hospital, University College London, London NW3 2PF, United Kingdom
| | - Yoram Reiter
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel;
| |
Collapse
|
25
|
Xue SA, Gao L, Ahmadi M, Ghorashian S, Barros RD, Pospori C, Holler A, Wright G, Thomas S, Topp M, Morris EC, Stauss HJ. Human MHC Class I-restricted high avidity CD4 + T cells generated by co-transfer of TCR and CD8 mediate efficient tumor rejection in vivo. Oncoimmunology 2014; 2:e22590. [PMID: 23483821 PMCID: PMC3583927 DOI: 10.4161/onci.22590] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this study, we generated human MHC Class I-restricted CD4+ T cells specific for Epstein-Barr virus (EBV) and cytomegalovirus (CMV), two herpesviridae associated with lymphoma, nasopharyngeal carcinoma and medulloblastoma, respectively. Retroviral transfer of virus-specific, HLA-A2-restricted TCR-coding genes generated CD4+ T cells that recognized HLA-A2/peptide multimers and produced cytokines when stimulated with MHC Class II-deficient cells presenting the relevant viral peptides in the context of HLA-A2. Peptide titration revealed that CD4+ T cells had a 10-fold lower avidity than CD8+ T cells expressing the same TCR. The impaired avidity of CD4+ T cells was corrected by simultaneously transferring TCR- and CD8-coding genes. The CD8 co-receptor did not alter the cytokine signature of CD4+ T cells, which remained distinct from that of CD8+ T cells. Using the xenogeneic NOD/SCID mouse model, we demonstrated that human CD4+ T cells expressing a specific TCR and CD8 can confer efficient protection against the growth of tumors expressing the EBV or CMV antigens recognized by the TCR. In summary, we describe a robust approach for generating therapeutic CD4+ T cells capable of providing MHC Class I-restricted immunity against MHC Class II-negative tumors in vivo.
Collapse
Affiliation(s)
- Shao-An Xue
- Department of Immunology; University College London; Royal Free Hospital; London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Adoptive immunotherapy, or the infusion of lymphocytes, is a promising approach for the treatment of cancer and certain chronic viral infections. The application of the principles of synthetic biology to enhance T cell function has resulted in substantial increases in clinical efficacy. The primary challenge to the field is to identify tumor-specific targets to avoid off-tumor, on-target toxicity. Given recent advances in efficacy in numerous pilot trials, the next steps in clinical development will require multicenter trials to establish adoptive immunotherapy as a mainstream technology.
Collapse
Affiliation(s)
- Marcela V Maus
- Translational Research Program, Abramson Cancer Center and
| | | | | | | | | | | |
Collapse
|
27
|
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: 1.0] [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]
|
28
|
Kunert A, Straetemans T, Govers C, Lamers C, Mathijssen R, Sleijfer S, Debets R. TCR-Engineered T Cells Meet New Challenges to Treat Solid Tumors: Choice of Antigen, T Cell Fitness, and Sensitization of Tumor Milieu. Front Immunol 2013; 4:363. [PMID: 24265631 PMCID: PMC3821161 DOI: 10.3389/fimmu.2013.00363] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/24/2013] [Indexed: 01/18/2023] Open
Abstract
Adoptive transfer of T cells gene-engineered with antigen-specific T cell receptors (TCRs) has proven its feasibility and therapeutic potential in the treatment of malignant tumors. To ensure further clinical development of TCR gene therapy, it is necessary to target immunogenic epitopes that are related to oncogenesis and selectively expressed by tumor tissue, and implement strategies that result in optimal T cell fitness. In addition, in particular for the treatment of solid tumors, it is equally necessary to include strategies that counteract the immune-suppressive nature of the tumor micro-environment. Here, we will provide an overview of the current status of TCR gene therapy, and redefine the following three challenges of improvement: “choice of target antigen”; “fitness of T cells”; and “sensitization of tumor milieu.” We will categorize and discuss potential strategies to address each of these challenges, and argue that advancement of clinical TCR gene therapy critically depends on developments toward each of the three challenges.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Experimental Tumor Immunology, Erasmus MC Cancer Institute , Rotterdam , Netherlands ; Department of Medical Oncology, Erasmus MC Cancer Institute , Rotterdam , Netherlands
| | | | | | | | | | | | | |
Collapse
|
29
|
Reuß S, Sebestyén Z, Heinz N, Loew R, Baum C, Debets R, Uckert W. TCR-engineered T cells: a model of inducible TCR expression to dissect the interrelationship between two TCRs. Eur J Immunol 2013; 44:265-74. [PMID: 24114521 PMCID: PMC4209802 DOI: 10.1002/eji.201343591] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 08/01/2013] [Accepted: 09/12/2013] [Indexed: 11/30/2022]
Abstract
TCR gene modified T cells for adoptive therapy simultaneously express the Tg TCR and the endogenous TCR, which might lead to mispaired TCRs with harmful unknown specificity and to a reduced function of TCR-Tg T cells. We generated dual TCR T cells in two settings in which either TCR was constitutively expressed by a retroviral promoter while the second TCR expression was regulable by a Tet-on system. Constitutively expressed TCR molecules were reduced on the cell surface depending on the induced TCR expression leading to strongly hampered function. Besides that, using fluorescence resonance energy transfer we detected mispaired TCR dimers and different pairing behaviors of individual TCR chains with a mutual influence on TCR chain expression. The loss of function and mispairing could not be avoided by changing the TCR expression level or by introduction of an additional cysteine bridge. However, in polyclonal T cells, optimized TCR formats (cysteineization, codon optimization) enhanced correct pairing and function. We conclude from our data that (i) the level of mispairing depends on the individual TCRs and is not reduced by increasing the level of one TCR, and (ii) modifications (cysteineization, codon optimization) improve correct pairing but do not completely exclude mispairing (cysteineization).
Collapse
Affiliation(s)
- Simone Reuß
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Stauss HJ, Morris EC. Immunotherapy with gene-modified T cells: limiting side effects provides new challenges. Gene Ther 2013; 20:1029-32. [DOI: 10.1038/gt.2013.34] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/03/2013] [Accepted: 05/20/2013] [Indexed: 01/19/2023]
|
31
|
Iwami K, Natsume A, Ohno M, Ikeda H, Mineno J, Nukaya I, Okamoto S, Fujiwara H, Yasukawa M, Shiku H, Wakabayashi T. Adoptive transfer of genetically modified Wilms' tumor 1-specific T cells in a novel malignant skull base meningioma model. Neuro Oncol 2013; 15:747-58. [PMID: 23460320 DOI: 10.1093/neuonc/not007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Meningiomas are the most commonly diagnosed primary intracranial neoplasms. Despite significant advances in modern therapies, the management of malignant meningioma and skull base meningioma remains a challenge. Thus, the development of new treatment modalities is urgently needed for these difficult-to-treat meningiomas. The goal of this study was to investigate the potential of build-in short interfering RNA-based Wilms' tumor protein (WT1)-targeted adoptive immunotherapy in a reproducible mouse model of malignant skull base meningioma that we recently established. METHODS We compared WT1 mRNA expression in human meningioma tissues and gliomas by quantitative real-time reverse-transcription polymerase chain reaction. Human malignant meningioma cells (IOMM-Lee cells) were labeled with green fluorescent protein (GFP) and implanted at the skull base of immunodeficient mice by using the postglenoid foramen injection (PGFi) technique. The animals were sacrificed at specific time points for analysis of tumor formation. Two groups of animals received adoptive immunotherapy with control peripheral blood mononuclear cells (PBMCs) or WT1-targeted PBMCs. RESULTS High levels of WT1 mRNA expression were observed in many meningioma tissues and all meningioma cell lines. IOMM-Lee-GFP cells were successfully implanted using the PGFi technique, and malignant skull base meningiomas were induced in all mice. The systemically delivered WT1-targeted PBMCs infiltrated skull base meningiomas and significantly delayed tumor growth and increased survival time. CONCLUSIONS We have established a reproducible mouse model of malignant skull base meningioma. WT1-targeted adoptive immunotherapy appears to be a promising approach for the treatment of difficult-to-treat meningiomas.
Collapse
Affiliation(s)
- Kenichiro Iwami
- Department of Neurosurgery, Nagoya University, Graduate School of Medicine, 65, Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Berdien B, Reinhard H, Meyer S, Spöck S, Kröger N, Atanackovic D, Fehse B. Influenza virus-specific TCR-transduced T cells as a model for adoptive immunotherapy. Hum Vaccin Immunother 2013; 9:1205-16. [PMID: 23428899 DOI: 10.4161/hv.24051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adoptive transfer of T lymphocytes equipped with tumor-antigen specific T-cell receptors (TCRs) represents a promising strategy in cancer immunotherapy, but the approach remains technically demanding. Using influenza virus (Flu)-specific T-cell responses as a model system we compared different methods for the generation of T-cell clones and isolation of antigen-specific TCRs. Altogether, we generated 12 CD8(+) T-cell clones reacting to the Flu matrix protein (Flu-M) and 6 CD4(+) T-cell clones reacting to the Flu nucleoprotein (Flu-NP) from 4 healthy donors. IFN-γ-secretion-based enrichment of antigen-specific cells, optionally combined with tetramer staining, was the most efficient way for generating T-cell clones. In contrast, the commonly used limiting dilution approach was least efficient. TCR genes were isolated from T-cell clones and cloned into both a previously used gammaretroviral LTR-vector, MP91 and the novel lentiviral self-inactivating vector LeGO-MP that contains MP91-derived promotor and regulatory elements. To directly compare their functional efficiencies, we in parallel transduced T-cell lines and primary T cells with the two vectors encoding identical TCRs. Transduction efficiencies were approximately twice higher with the gammaretroviral vector. Secretion of high amounts of IFN-γ, IL-2 and TNF-α by transduced cells after exposure to the respective influenza target epitope proved efficient specificity transfer of the isolated TCRs to primary T-cells for both vectors, at the same time indicating superior functionality of MP91-transduced cells. In conclusion, we have developed optimized strategies to obtain and transfer antigen-specific TCRs as well as designed a novel lentiviral vector for TCR-gene transfer. Our data may help to improve adoptive T-cell therapies.
Collapse
Affiliation(s)
- Belinda Berdien
- Research Department Cell and Gene Therapy; Department of Stem Cell Transplantation (SCT); University Medical Center (UMC) Hamburg-Eppendorf; Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Choi D, Kim TG, Sung YC. The past, present, and future of adoptive T cell therapy. Immune Netw 2012; 12:139-47. [PMID: 23091437 PMCID: PMC3467412 DOI: 10.4110/in.2012.12.4.139] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 12/20/2022] Open
Abstract
Although adoptive T cell therapy (ACT) has become a promising immunotherapeutic regime for cancer treatment, its effectiveness has been hindered by several inherent shortcomings regarding safety and efficacy. During the past few decades, several strategies for enhancing the efficacy of ACT have been developed and introduced in clinic. This review will summarize not only the past approaches but also the latest strategies which have been shown to enhance the anticancer activity of ACT.
Collapse
Affiliation(s)
- Donghoon Choi
- Cellular Immunology Laboratory, Division of Molecular and Life Science, POSTECH Biotech Center, Pohang University of Science & Technology, Pohang 790-784, Korea
| | | | | |
Collapse
|
34
|
T-cell receptor gene transfer exclusively to human CD8(+) cells enhances tumor cell killing. Blood 2012; 120:4334-42. [PMID: 22898597 DOI: 10.1182/blood-2012-02-412973] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transfer of tumor-specific T-cell receptor (TCR) genes into patient T cells is a promising strategy in cancer immunotherapy. We describe here a novel vector (CD8-LV) derived from lentivirus, which delivers genes exclusively and specifically to CD8(+) cells. CD8-LV mediated stable in vitro and in vivo reporter gene transfer as well as efficient transfer of genes encoding TCRs recognizing the melanoma antigen tyrosinase. Strikingly, T cells genetically modified with CD8-LV killed melanoma cells reproducibly more efficiently than CD8(+) cells transduced with a conventional lentiviral vector. Neither TCR expression levels, nor the rate of activation-induced death of transduced cells differed between both vector types. Instead, CD8-LV transduced cells showed increased granzyme B and perforin levels as well as an up-regulation of CD8 surface expression in a small subpopulation of cells. Thus, a possible mechanism for CD8-LV enhanced tumor cell killing may be based on activation of the effector functions of CD8(+) T cells by the vector particle displaying OKT8-derived CD8-scFv and an increase of the surface density of CD8, which functions as coreceptor for tumor-cell recognition. CD8-LV represents a powerful novel vector for TCR gene therapy and other applications in immunotherapy and basic research requiring CD8(+) cell-specific gene delivery.
Collapse
|
35
|
Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer. Nat Med 2012; 18:807-815. [PMID: 22466705 DOI: 10.1038/nm.2700] [Citation(s) in RCA: 348] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 02/08/2012] [Indexed: 11/09/2022]
Abstract
The transfer of high-avidity T cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal T cells is an attractive cancer immunotherapy strategy. However, TCR gene transfer results in competition for surface expression and inappropriate pairing between the exogenous and endogenous TCR chains, resulting in suboptimal activity and potentially harmful unpredicted antigen specificities of the resultant TCRs. We designed zinc-finger nucleases (ZFNs) that promoted the disruption of endogenous TCR β- and α-chain genes. Lymphocytes treated with ZFNs lacked surface expression of CD3-TCR and expanded with the addition of interleukin-7 (IL-7) and IL-15. After lentiviral transfer of a TCR specific for the Wilms tumor 1 (WT1) antigen, these TCR-edited cells expressed the new TCR at high levels, were easily expanded to near purity and were superior at specific antigen recognition compared to donor-matched, unedited TCR-transferred cells. In contrast to unedited TCR-transferred cells, the TCR-edited lymphocytes did not mediate off-target reactivity while maintaining their anti-tumor activity in vivo, thus showing that complete editing of T cell specificity generates tumor-specific lymphocytes with improved biosafety profiles.
Collapse
|
36
|
Daniel-Meshulam I, Ya'akobi S, Ankri C, Cohen CJ. How (specific) would like your T-cells today? Generating T-cell therapeutic function through TCR-gene transfer. Front Immunol 2012; 3:186. [PMID: 22783259 PMCID: PMC3390604 DOI: 10.3389/fimmu.2012.00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/15/2012] [Indexed: 01/02/2023] Open
Abstract
T-cells are central players in the immune response against both pathogens and cancer. Their specificity is solely dictated by the T-cell receptor (TCR) they clonally express. As such, the genetic modification of T lymphocytes using pathogen- or cancer-specific TCRs represents an appealing strategy to generate a desired immune response from peripheral blood lymphocytes. Moreover, notable objective clinical responses were observed in terminally ill cancer patients treated with TCR-gene modified cells in several clinical trials conducted recently. Nevertheless, several key aspects of this approach are the object of intensive research aimed at improving the reliability and efficacy of this strategy. Herein, we will survey recent studies in the field of TCR-gene transfer dealing with the improvement of this approach and its application for the treatment of malignant, autoimmune, and infectious diseases.
Collapse
Affiliation(s)
- Inbal Daniel-Meshulam
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences , Bar-Ilan University, Ramat Gan, Israel
| | | | | | | |
Collapse
|
37
|
Haga-Friedman A, Horovitz-Fried M, Cohen CJ. Incorporation of transmembrane hydrophobic mutations in the TCR enhance its surface expression and T cell functional avidity. THE JOURNAL OF IMMUNOLOGY 2012; 188:5538-46. [PMID: 22544927 DOI: 10.4049/jimmunol.1103020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TCR-gene transfer represents an effective way to redirect the specificity of T lymphocytes for therapeutic purposes. Recent successful clinical trials have underscored the potential of this approach in which efficient expression of the exogenous TCR has been directly linked to the efficacy of T cell activity. It has been also demonstrated that the TCR exhibits a lack of stability associated with the presence of positively charged residues in its transmembrane (TM) region. In this study, we designed an original approach selectively to improve exogenous TCR stability by increasing the hydrophobic nature of the TCRα TM region. Incorporation of hydrophobic residues at evolutionarily permissive positions resulted in an enhanced surface expression of the TCR chains, leading to an improved cellular avidity and anti-tumor TCR activity. Furthermore, this strategy was successfully applied to different TCRs, enabling the targeting of human tumors from different histologies. We also show that the combination of these hydrophobic mutations with another TCR-enhancing approach further improved TCR expression and function. Overall, these findings provide information regarding TCR TM composition that can be applied for the improvement of TCR-gene transfer-based treatments.
Collapse
Affiliation(s)
- Astar Haga-Friedman
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel
| | | | | |
Collapse
|
38
|
Marr LA, Gilham DE, Campbell JDM, Fraser AR. Immunology in the clinic review series; focus on cancer: double trouble for tumours: bi-functional and redirected T cells as effective cancer immunotherapies. Clin Exp Immunol 2012; 167:216-25. [PMID: 22235997 PMCID: PMC3278687 DOI: 10.1111/j.1365-2249.2011.04517.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2011] [Indexed: 01/04/2023] Open
Abstract
Cancer is one of the most important pathological conditions facing mankind in the 21st century, and is likely to become the most important cause of death as improvements continue in health, diet and life expectancy. The immune response is responsible for controlling nascent cancer through immunosurveillance. If tumours escape this control, they can develop into clinical cancer. Although surgery and chemo- or radiotherapy have improved survival rates significantly, there is a drive to reharness immune responses to treat disease. As T cells are one of the key immune cells in controlling cancer, research is under way to enhance their function and improve tumour targeting. This can be achieved by transduction with tumour-specific T cell receptor (TCR) or chimaeric antigen receptors (CAR) to generate redirected T cells. Virus-specific cells can also be transduced with TCR or CAR to create bi-functional T cells with specificity for both virus and tumour. In this review we outline the development and optimization of redirected and bi-functional T cells, and outline the results from current clinical trials using these cells. From this we discuss the challenges involved in generating effective anti-tumour responses while avoiding concomitant damage to normal tissues and organs.
Collapse
Affiliation(s)
- L A Marr
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
39
|
Improving TCR Gene Therapy for Treatment of Haematological Malignancies. Adv Hematol 2012; 2012:404081. [PMID: 22319532 PMCID: PMC3272793 DOI: 10.1155/2012/404081] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 10/10/2011] [Indexed: 12/14/2022] Open
Abstract
Adoptive immunotherapy using TCR gene modified T cells may allow separation of beneficial Graft versus tumour responses from harmful GvHD. Improvements to this include methods to generate high avidity or high affinity TCR, improvements in vector design and reduction in mispairing. Following adoptive transfer, TCR transduced T cells must be able to survive and persist in vivo to give most effective antitumour responses. Central memory or naive T cells have both been shown to be more effective than effector cells at expanding and persisting in vivo. Lymphodepletion may enhance persistence of transferred T cell populations. TCR gene transfer can be used to redirect CD4 helper T cells, and these could be used in combination with CD8+ tumour specific T cells to provide help for the antitumour response. Antigen specific T regulatory T cells can also be generated by TCR gene transfer and could be used to suppress unwanted alloresponses.
Collapse
|
40
|
Merhavi-Shoham E, Haga-Friedman A, Cohen CJ. Genetically modulating T-cell function to target cancer. Semin Cancer Biol 2011; 22:14-22. [PMID: 22210183 DOI: 10.1016/j.semcancer.2011.12.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/15/2011] [Indexed: 12/14/2022]
Abstract
The adoptive transfer of tumor-specific T-lymphocytes holds promise for the treatment of metastatic cancer. Genetic modulation of T-lymphocytes using TCR transfer with tumor-specific TCR genes is an attractive strategy to generate anti-tumor response, especially against large solid tumors. Recently, several clinical trials have demonstrated the therapeutic potential of this approach which lead to impressive tumor regression in cancer patients. Still, several factors may hinder the clinical benefit of this approach, such as the type of cells to modulate, the vector configuration or the safety of the procedure. In the present review we will aim at giving an overview of the recent developments related to the immune modulation of the anti-tumor adaptive response using genetically engineered lymphocytes and will also elaborate the development of other genetic modifications to enhance their anti-tumor immune response.
Collapse
Affiliation(s)
- Efrat Merhavi-Shoham
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel
| | | | | |
Collapse
|
41
|
Ghorashian S, Nicholson E, Stauss HJ. T cell gene-engineering to enhance GVT and suppress GVHD. Best Pract Res Clin Haematol 2011; 24:421-33. [DOI: 10.1016/j.beha.2011.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Capitini CM, Gottschalk S, Brenner M, Cooper LJN, Handgretinger R, Mackall CL. Highlights of the second international conference on "Immunotherapy in Pediatric Oncology". Pediatr Hematol Oncol 2011; 28:459-60. [PMID: 21854215 DOI: 10.3109/08880018.2011.596615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Second International Conference on Immunotherapy in Pediatric Oncology was held in Houston, Texas, USA, October 11-12, 2010, to discuss the progress and challenges that have occurred in cutting edge immunotherapeutic strategies currently being developed for pediatric oncology. Major topics included immune targeting of acute lymphoblastic leukemia and pediatric solid tumors, chimeric antigen receptors (CARs) for hematologic malignancies and solid tumors, enhancing graft-versus-leukemia for pediatric cancers, overcoming hurdles of immunotherapy, strategies to active the innate immune system, and moving immunotherapy beyond phase I studies. Significant progress has been made in the last 2 years both in the development of novel immunobiologics such as CARs, and in establishing survival benefits of an anti-GD2 monoclonal antibody in randomized studies. Although there is much excitement going forward, a great deal of laboratory and regulatory challenges lie ahead in improving the efficacy of each of these modalities as well as getting them to patients in a timely and cost-effective fashion. The resulting discussions will hopefully lead to new collaborations and insight for further translational and clinical studies.
Collapse
Affiliation(s)
- Christian M Capitini
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Falkenburg WJJ, Melenhorst JJ, van de Meent M, Kester MGD, Hombrink P, Heemskerk MHM, Hagedoorn RS, Gostick E, Price DA, Falkenburg JHF, Barrett AJ, Jedema I. Allogeneic HLA-A*02-restricted WT1-specific T cells from mismatched donors are highly reactive but show off-target promiscuity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:2824-33. [PMID: 21821799 DOI: 10.4049/jimmunol.1100852] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
T cells recognizing tumor-associated Ags such as Wilms tumor protein (WT1) are thought to exert potent antitumor reactivity. However, no consistent high-avidity T cell responses have been demonstrated in vaccination studies with WT1 as target in cancer immunotherapy. The aim of this study was to investigate the possible role of negative thymic selection on the avidity and specificity of T cells directed against self-antigens. T cell clones directed against the HLA-A*0201-binding WT1(126-134) peptide were generated from both HLA-A*02-positive (self-HLA-restricted) and HLA-A*02-negative [nonself (allogeneic) HLA [allo-HLA]-restricted] individuals by direct ex vivo isolation using tetramers or after in vitro priming and selection. The functional avidity and specificity of these T cell clones was analyzed in-depth. Self-HLA-restricted WT1-specific clones only recognized WT1(126-134) with low avidities. In contrast, allo-HLA-restricted WT1 clones exhibited profound functional reactivity against a multitude of HLA-A*02-positive targets, even in the absence of exogenously loaded WT1 peptide, indicative of Ag-binding promiscuity. To characterize this potential promiscuity, reactivity of the T cell clones against 400 randomly selected HLA-A*0201-binding peptides was investigated. The self-HLA-restricted WT1-specific T cell clones only recognized the WT1 peptide. In contrast, the allo-HLA-restricted WT1-reactive clones recognized besides WT1 various other HLA-A*0201-binding peptides. In conclusion, allogeneic HLA-A*02-restricted WT1-specific T cells isolated from mismatched donors may be more tumor-reactive than their autologous counterparts but can show specific off-target promiscuity of potential clinical importance. As a result of this, administration of WT1-specific T cells generated from HLA-mismatched donors should be performed with appropriate precautions against potential off-target effects.
Collapse
Affiliation(s)
- Willem J J Falkenburg
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Single-chain VαVβ T-cell receptors function without mispairing with endogenous TCR chains. Gene Ther 2011; 19:365-74. [PMID: 21753797 DOI: 10.1038/gt.2011.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transduction of exogenous T-cell receptor (TCR) genes into patients' activated peripheral blood T cells is a potent strategy to generate large numbers of specific T cells for adoptive therapy of cancer and viral diseases. However, the remarkable clinical promise of this powerful approach is still being overshadowed by a serious potential consequence: mispairing of the exogenous TCR chains with endogenous TCR chains. These 'mixed' heterodimers can generate new specificities that result in graft-versus-host reactions. Engineering TCR constant regions of the exogenous chains with a cysteine promotes proper pairing and reduces the mispairing, but, as we show here, does not eliminate the formation of mixed heterodimers. By contrast, deletion of the constant regions, through use of a stabilized Vα/Vβ single-chain TCR (scTv), avoided mispairing completely. By linking a high-affinity scTv to intracellular signaling domains, such as Lck and CD28, the scTv was capable of activating functional T-cell responses in the absence of either the CD3 subunits or the co-receptors, and circumvented mispairing with endogenous TCRs. Such transduced T cells can respond to the targeted antigen independent of CD3 subunits via the introduced scTv, without the transduced T cells acquiring any new undefined and potentially dangerous specificities.
Collapse
|
45
|
Abstract
The function of T-cell receptor (TCR) gene modified T cells is dependent on efficient surface expression of the introduced TCR α/β heterodimer. We tested whether endogenous CD3 chains are rate-limiting for TCR expression and antigen-specific T-cell function. We show that co-transfer of CD3 and TCR genes into primary murine T cells enhanced TCR expression and antigen-specific T-cell function in vitro. Peptide titration experiments showed that T cells expressing introduced CD3 and TCR genes recognized lower concentration of antigen than T cells expressing TCR only. In vivo imaging revealed that TCR+CD3 gene modified T cells infiltrated tumors faster and in larger numbers, which resulted in more rapid tumor elimination compared with T cells modified by TCR only. After tumor clearance, TCR+CD3 engineered T cells persisted in larger numbers than TCR-only T cells and mounted a more effective memory response when rechallenged with antigen. The data demonstrate that provision of additional CD3 molecules is an effective strategy to enhance the avidity, anti-tumor activity and functional memory formation of TCR gene modified T cells in vivo.
Collapse
|
46
|
Gehring AJ, Xue SA, Ho ZZ, Teoh D, Ruedl C, Chia A, Koh S, Lim SG, Maini MK, Stauss H, Bertoletti A. Engineering virus-specific T cells that target HBV infected hepatocytes and hepatocellular carcinoma cell lines. J Hepatol 2011; 55:103-10. [PMID: 21145860 DOI: 10.1016/j.jhep.2010.10.025] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 10/01/2010] [Accepted: 10/20/2010] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Virus-specific T cells capable of controlling HBV and eliminating hepatocellular carcinoma (HCC) expressing HBV antigens are deleted or dysfunctional in patients with chronic HBV or HBV-related HCC. The goal of this study was to determine if T cell receptor (TCR) gene transfer can reconstitute HBV-specific T cell immunity in lymphocytes of chronic HBV patients and investigate whether HCC cells with natural HBV-DNA integration can be recognized by genetically modified T cells. METHODS We used vector-mediated gene transfer to introduce HLA-A2-restricted, HBV-specific TCRs into T cells of chronic HBV as well as HBV-related HCC patients. RESULTS The introduced TCRs were expressed on the cell surface, evidenced by Vβ and pentamer staining. TCR transduced T cells produced IFN-γ, TNF-α, IL-2, and lysed HBV infected hepatocyte-like cell lines. Furthermore, HCC cell lines with natural HBV-DNA integration could be recognized by HBV-specific TCR-re-directed T cells. CONCLUSIONS TCR re-directed HBV-specific T cells generated from PBMC of chronic HBV and HBV-related HCC patients were multifunctional and capable of recognizing HBV-infected cells and HCC tumor cells expressing viral antigens from naturally integrated HBV DNA. These genetically modified T cells could be used to reconstitute virus-specific T cell immunity in chronic HBV patients and target tumors in HBV-related HCC.
Collapse
Affiliation(s)
- Adam J Gehring
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ribas A, Koya RC. Adoptive cell transfer of T-cell receptor-engineered lymphocytes: lessons from recent modeling. Future Oncol 2011; 6:1671-3. [PMID: 21142653 DOI: 10.2217/fon.10.137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
48
|
Ochi T, Fujiwara H, Yasukawa M. Requisite considerations for successful adoptive immunotherapy with engineered T-lymphocytes using tumor antigen-specific T-cell receptor gene transfer. Expert Opin Biol Ther 2011; 11:699-713. [PMID: 21413911 DOI: 10.1517/14712598.2011.566853] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Although engineered T-cell-based antitumor immunotherapy using tumor-antigen-specific T-cell receptor (TCR) gene transfer is undoubtedly a promising strategy, a number of studies have revealed that it has several drawbacks. AREAS COVERED This review covers selected articles detailing recent progress in this field, not only for solid tumors, but also for leukemias. In terms of achieving uniform therapeutic quality of TCR gene-modified T cells as an 'off-the-shelf' product, the authors abstract and discuss the requisite conditions for successful outcome, including: i) the optimal target choice reflecting the specificity of the introduced TCR, ii) the quality and quantity of expressed TCRs in gene-modified T cells, and additional genetic modification reflecting enhanced antitumor functionality, and iii) 'on-' and 'off-target' adverse events caused by the quality of the introduced TCRs and other adverse events related to genetic modification itself. Readers will be able to readily abstract recent advances in TCR gene-transferred T-cell therapy, centering notably on efforts to obtain uniformity in the therapeutic functionality of engineered T cells. EXPERT OPINION Harmonizing the functionality and target specificity of TCR will allow the establishment of clinically useful adoptive immunotherapy in the near future.
Collapse
Affiliation(s)
- Toshiki Ochi
- Department of Bioregulatory Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791 0295, Japan.
| | | | | |
Collapse
|
49
|
Abstract
The adoptive transfer of tumor-reactive cells is a promising approach for the treatment of melanoma and some other cancers. To remedy the difficulties associated with the isolation and expansion of tumor-reactive T cells in most cancer patients, peripheral blood T cells can be retargeted to any chosen tumor antigen by the genetic transfer of an antigen-specific receptor. The transduced receptors may be human leukocyte antigen-restricted, heterodimeric T-cell antigen receptor (TCRs), or chimeric antigen receptors (CARs), which typically recognize native cell-surface antigens. Considerable progress has been made in recent years to address the challenges posed by the transfer of either receptor type. Vector and protein modifications enable the expression of TCR chains in human T cells at functional levels and with a reduced risk of mis-pairing with endogenous TCR chains. The combinatorial inclusion of activating and costimulatory domains in CARs has dramatically enhanced the signaling properties of the chimeric receptors described over a decade ago. Based on the effective T-cell transduction and expansion procedures now available to support clinical investigation, improved designer TCRs and second generation CARs targeting an array of antigens are being evaluated in a range of hematological malignancies and solid tumors.
Collapse
|
50
|
Canderan G, Gruarin P, Montagna D, Fontana R, Melloni G, Traversari C, Dellabona P, Casorati G. An efficient strategy to induce and maintain in vitro human T cells specific for autologous non-small cell lung carcinoma. PLoS One 2010; 5:e12014. [PMID: 20711505 PMCID: PMC2918513 DOI: 10.1371/journal.pone.0012014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Accepted: 07/15/2010] [Indexed: 11/18/2022] Open
Abstract
Background The efficient expansion in vitro of cytolytic CD8+ T cells (CTLs) specific for autologous tumors is crucial both for basic and translational aspects of tumor immunology. We investigated strategies to generate CTLs specific for autologous Non-Small Cell Lung Carcinoma (NSCLC), the most frequent tumor in mankind, using circulating lymphocytes. Principal Findings Classic Mixed Lymphocyte Tumor Cultures with NSCLC cells consistently failed to induce tumor-specific CTLs. Cross-presentation in vitro of irradiated NSCLC cells by autologous dendritic cells, by contrast, induced specific CTL lines from which we obtained a high number of tumor-specific T cell clones (TCCs). The TCCs displayed a limited TCR diversity, suggesting an origin from few tumor-specific T cell precursors, while their TCR molecular fingerprints were detected in the patient's tumor infiltrating lymphocytes, implying a role in the spontaneous anti-tumor response. Grafting NSCLC-specific TCR into primary allogeneic T cells by lentiviral vectors expressing human V-mouse C chimeric TCRα/β chains overcame the growth limits of these TCCs. The resulting, rapidly expanding CD4+ and CD8+ T cell lines stably expressed the grafted chimeric TCR and specifically recognized the original NSCLC. Conclusions This study defines a strategy to efficiently induce and propagate in vitro T cells specific for NSCLC starting from autologous peripheral blood lymphocytes.
Collapse
Affiliation(s)
- Glenda Canderan
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Paola Gruarin
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Montagna
- Laboratory of Immunology, Department of Pediatrics, University of Pavia, Pavia, Italy
| | - Raffaella Fontana
- Cancer Gene Therapy Unit, Division of Molecular Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Melloni
- Department of Thoracic Surgery, San Raffaele Scientific Institute, Milan, Italy
| | | | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- * E-mail: (PD); (GC)
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- * E-mail: (PD); (GC)
| |
Collapse
|