1
|
Colina AS, Shah V, Shah RK, Kozlik T, Dash RK, Terhune S, Zamora AE. Current advances in experimental and computational approaches to enhance CAR T cell manufacturing protocols and improve clinical efficacy. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1310002. [PMID: 39086435 PMCID: PMC11285593 DOI: 10.3389/fmmed.2024.1310002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 08/02/2024]
Abstract
Since the FDA's approval of chimeric antigen receptor (CAR) T cells in 2017, significant improvements have been made in the design of chimeric antigen receptor constructs and in the manufacturing of CAR T cell therapies resulting in increased in vivo CAR T cell persistence and improved clinical outcome in certain hematological malignancies. Despite the remarkable clinical response seen in some patients, challenges remain in achieving durable long-term tumor-free survival, reducing therapy associated malignancies and toxicities, and expanding on the types of cancers that can be treated with this therapeutic modality. Careful analysis of the biological factors demarcating efficacious from suboptimal CAR T cell responses will be of paramount importance to address these shortcomings. With the ever-expanding toolbox of experimental approaches, single-cell technologies, and computational resources, there is renowned interest in discovering new ways to streamline the development and validation of new CAR T cell products. Better and more accurate prognostic and predictive models can be developed to help guide and inform clinical decision making by incorporating these approaches into translational and clinical workflows. In this review, we provide a brief overview of recent advancements in CAR T cell manufacturing and describe the strategies used to selectively expand specific phenotypic subsets. Additionally, we review experimental approaches to assess CAR T cell functionality and summarize current in silico methods which have the potential to improve CAR T cell manufacturing and predict clinical outcomes.
Collapse
Affiliation(s)
- Alfredo S. Colina
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Viren Shah
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Ravi K. Shah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tanya Kozlik
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ranjan K. Dash
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Scott Terhune
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, United States
| | - Anthony E. Zamora
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
2
|
Thieme CJ, Schulz M, Wehler P, Anft M, Amini L, Blàzquez-Navarro A, Stervbo U, Hecht J, Nienen M, Stittrich AB, Choi M, Zgoura P, Viebahn R, Schmueck-Henneresse M, Reinke P, Westhoff TH, Roch T, Babel N. In vitro and in vivo evidence that the switch from calcineurin to mTOR inhibitors may be a strategy for immunosuppression in Epstein-Barr virus-associated post-transplant lymphoproliferative disorder. Kidney Int 2022; 102:1392-1408. [PMID: 36103953 DOI: 10.1016/j.kint.2022.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/12/2023]
Abstract
Post-transplant lymphoproliferative disorder is a life-threatening complication of immunosuppression following transplantation mediated by failure of T cells to control Epstein-Barr virus (EBV)-infected and transformed B cells. Typically, a modification or reduction of immunosuppression is recommended, but insufficiently defined thus far. In order to help delineate this, we characterized EBV-antigen-specific T cells and lymphoblastoid cell lines from healthy donors and in patients with a kidney transplant in the absence or presence of the standard immunosuppressants tacrolimus, cyclosporin A, prednisolone, rapamycin, and mycophenolic acid. Phenotypes of lymphoblastoid cell-lines and T cells, T cell-receptor-repertoire diversity, and T-cell reactivity upon co-culture with autologous lymphoblastoid cell lines were analyzed. Rapamycin and mycophenolic acid inhibited lymphoblastoid cell-line proliferation. T cells treated with prednisolone and rapamycin showed nearly normal cytokine production. Proliferation and the viability of T cells were decreased by mycophenolic acid, while tacrolimus and cyclosporin A were strong suppressors of T-cell function including their killing activity. Overall, our study provides a basis for the clinical decision for the modification and reduction of immunosuppression and adds information to the complex balance of maintaining anti-viral immunity while preventing acute rejection. Thus, an immunosuppressive regime based on mTOR inhibition and reduced or withdrawn calcineurin inhibitors could be a promising strategy for patients with increased risk of or manifested EBV-associated post-transplant lymphoproliferative disorder.
Collapse
Affiliation(s)
- Constantin J Thieme
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Malissa Schulz
- Hochschule für Technik und Wirtschaft Berlin (HTW), Berlin, Germany
| | - Patrizia Wehler
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Anft
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Leila Amini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arturo Blàzquez-Navarro
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Jochen Hecht
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain; Experimental and Health Sciences Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mikalai Nienen
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Panagiota Zgoura
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Richard Viebahn
- Department of Surgery, University Hospital Knappschaftskrankenhaus Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Michael Schmueck-Henneresse
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Timm H Westhoff
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Toralf Roch
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany.
| |
Collapse
|
3
|
Peter L, Wendering DJ, Schlickeiser S, Hoffmann H, Noster R, Wagner DL, Zarrinrad G, Münch S, Picht S, Schulenberg S, Moradian H, Mashreghi MF, Klein O, Gossen M, Roch T, Babel N, Reinke P, Volk HD, Amini L, Schmueck-Henneresse M. Tacrolimus-resistant SARS-CoV-2-specific T cell products to prevent and treat severe COVID-19 in immunosuppressed patients. Mol Ther Methods Clin Dev 2022; 25:52-73. [PMID: 35252469 PMCID: PMC8882037 DOI: 10.1016/j.omtm.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Solid organ transplant (SOT) recipients receive therapeutic immunosuppression that compromises their immune response to infections and vaccines. For this reason, SOT patients have a high risk of developing severe coronavirus disease 2019 (COVID-19) and an increased risk of death from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Moreover, the efficiency of immunotherapies and vaccines is reduced due to the constant immunosuppression in this patient group. Here, we propose adoptive transfer of SARS-CoV-2-specific T cells made resistant to a common immunosuppressant, tacrolimus, for optimized performance in the immunosuppressed patient. Using a ribonucleoprotein approach of CRISPR-Cas9 technology, we have generated tacrolimus-resistant SARS-CoV-2-specific T cell products from convalescent donors and demonstrate their specificity and function through characterizations at the single-cell level, including flow cytometry, single-cell RNA (scRNA) Cellular Indexing of Transcriptomes and Epitopes (CITE), and T cell receptor (TCR) sequencing analyses. Based on the promising results, we aim for clinical validation of this approach in transplant recipients. Additionally, we propose a combinatory approach with tacrolimus, to prevent an overshooting immune response manifested as bystander T cell activation in the setting of severe COVID-19 immunopathology, and tacrolimus-resistant SARS-CoV-2-specific T cell products, allowing for efficient clearance of viral infection. Our strategy has the potential to prevent severe COVID-19 courses in SOT or autoimmunity settings and to prevent immunopathology while providing viral clearance in severe non-transplant COVID-19 cases.
Collapse
Affiliation(s)
- Lena Peter
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Einstein Center for Regenerative Therapies at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Désirée Jacqueline Wendering
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Stephan Schlickeiser
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Henrike Hoffmann
- Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Rebecca Noster
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Dimitrios Laurin Wagner
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Transfusion Medicine, Charitéplatz 1, 10117 Berlin, Germany
| | - Ghazaleh Zarrinrad
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Einstein Center for Regenerative Therapies at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Sandra Münch
- Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Samira Picht
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Sarah Schulenberg
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Einstein Center for Regenerative Therapies at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Hanieh Moradian
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Kantstr. 55, 14513 Teltow, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Mir-Farzin Mashreghi
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Deutsches Rheuma-Forschungszentrum Berlin, a Leibniz Institute, Charitéplatz 1, 10117 Berlin, Germany
| | - Oliver Klein
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany
| | - Manfred Gossen
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Kantstr. 55, 14513 Teltow, Germany
| | - Toralf Roch
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353 Berlin, Germany.,Center for Translational Medicine, Immunology, and Transplantation, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Nina Babel
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353 Berlin, Germany.,Center for Translational Medicine, Immunology, and Transplantation, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Hölkeskampring 40, 44625 Herne, Germany
| | - Petra Reinke
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Leila Amini
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT) at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
4
|
Amini L, Wagner DL, Rössler U, Zarrinrad G, Wagner LF, Vollmer T, Wendering DJ, Kornak U, Volk HD, Reinke P, Schmueck-Henneresse M. CRISPR-Cas9-Edited Tacrolimus-Resistant Antiviral T Cells for Advanced Adoptive Immunotherapy in Transplant Recipients. Mol Ther 2021; 29:32-46. [PMID: 32956624 PMCID: PMC7791012 DOI: 10.1016/j.ymthe.2020.09.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
Viral infections, such as with cytomegalovirus (CMV), remain a major risk factor for mortality and morbidity of transplant recipients because of their requirement for lifelong immunosuppression (IS). Antiviral drugs often cause toxicity and sometimes fail to control disease. Thus, regeneration of the antiviral immune response by adoptive antiviral T cell therapy is an attractive alternative. Our recent data, however, show only short-term efficacy in some solid organ recipients, possibly because of malfunction in transferred T cells caused by ongoing IS. We developed a vector-free clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-based good manufacturing practice (GMP)-compliant protocol that efficiently targets and knocks out the gene for the adaptor protein FK506-binding protein 12 (FKBP12), required for the immunosuppressive function of tacrolimus. This was achieved by transient delivery of ribonucleoprotein complexes into CMV-specific T cells by electroporation. We confirmed the tacrolimus resistance of our gene-edited T cell products in vitro and demonstrated performance comparable with non-tacrolimus-treated unmodified T cells. The alternative calcineurin inhibitor cyclosporine A can be administered as a safety switch to shut down tacrolimus-resistant T cell activity in case of adverse effects. Furthermore, we performed safety assessments as a prerequisite for translation to first-in-human applications.
Collapse
Affiliation(s)
- Leila Amini
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dimitrios Laurin Wagner
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Uta Rössler
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Ghazaleh Zarrinrad
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Einstein Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Livia Felicitas Wagner
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Tino Vollmer
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Désirée Jacqueline Wendering
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Uwe Kornak
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (B-CRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany.
| |
Collapse
|
5
|
Thieme CJ, Anft M, Paniskaki K, Blazquez-Navarro A, Doevelaar A, Seibert FS, Hoelzer B, Konik MJ, Berger MM, Brenner T, Tempfer C, Watzl C, Meister TL, Pfaender S, Steinmann E, Dolff S, Dittmer U, Westhoff TH, Witzke O, Stervbo U, Roch T, Babel N. Robust T Cell Response Toward Spike, Membrane, and Nucleocapsid SARS-CoV-2 Proteins Is Not Associated with Recovery in Critical COVID-19 Patients. Cell Rep Med 2020; 1:100092. [PMID: 32904468 PMCID: PMC7456276 DOI: 10.1016/j.xcrm.2020.100092] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/06/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023]
Abstract
T cell immunity toward SARS-CoV-2 spike (S-), membrane (M-), and nucleocapsid (N-) proteins may define COVID-19 severity. Therefore, we compare the SARS-CoV-2-reactive T cell responses in moderate, severe, and critical COVID-19 patients and unexposed donors. Overlapping peptide pools of all three proteins induce SARS-CoV-2-reactive T cell response with dominance of CD4+ over CD8+ T cells and demonstrate interindividual immunity against the three proteins. M-protein induces the highest frequencies of CD4+ T cells, suggesting its relevance for diagnosis and vaccination. The T cell response of critical COVID-19 patients is robust and comparable or even superior to non-critical patients. Virus clearance and COVID-19 survival are not associated with either SARS-CoV-2 T cell kinetics or magnitude of T cell responses, respectively. Thus, our data do not support the hypothesis of insufficient SARS-CoV-2-reactive immunity in critical COVID-19. Conversely, it indicates that activation of differentiated memory effector T cells could cause hyperreactivity and immunopathogenesis in critical patients.
Collapse
Affiliation(s)
- Constantin J. Thieme
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, BIH Center for Regenerative Therapies, Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Berlin, Germany
| | - Moritz Anft
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Krystallenia Paniskaki
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Arturo Blazquez-Navarro
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, BIH Center for Regenerative Therapies, Berlin, Berlin, Germany
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Adrian Doevelaar
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Felix S. Seibert
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Bodo Hoelzer
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Margarethe Justine Konik
- University Duisburg-Essen, University Hospital Essen, Department of Infectious Diseases, West-German Centre for Infectious Diseases, Essen, North Rhine-Westphalia, Germany
| | - Marc Moritz Berger
- University Duisburg-Essen, University Hospital Essen, Department of Anesthesiology and Intensive Care Medicine, Essen, North Rhine-Westphalia, Germany
| | - Thorsten Brenner
- University Duisburg-Essen, University Hospital Essen, Department of Anesthesiology and Intensive Care Medicine, Essen, North Rhine-Westphalia, Germany
| | - Clemens Tempfer
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Department of Gynecology and Obstetrics, Herne, North Rhine-Westphalia, Germany
| | - Carsten Watzl
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund (IfADo), Department of Immunology Dortmund, North Rhine-Westphalia, Germany
| | - Toni L. Meister
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, North Rhine-Westphalia, Germany
| | - Stephanie Pfaender
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, North Rhine-Westphalia, Germany
| | - Eike Steinmann
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, North Rhine-Westphalia, Germany
| | - Sebastian Dolff
- University Duisburg-Essen, University Hospital Essen, Department of Infectious Diseases, West-German Centre for Infectious Diseases, Essen, North Rhine-Westphalia, Germany
| | - Ulf Dittmer
- University Duisburg-Essen, University Hospital Essen, Institute for Virology, Essen, North Rhine-Westphalia, Germany
| | - Timm H. Westhoff
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Oliver Witzke
- University Duisburg-Essen, University Hospital Essen, Department of Infectious Diseases, West-German Centre for Infectious Diseases, Essen, North Rhine-Westphalia, Germany
| | - Ulrik Stervbo
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Toralf Roch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, BIH Center for Regenerative Therapies, Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Berlin, Germany
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| | - Nina Babel
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, BIH Center for Regenerative Therapies, Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Medical Immunology, Berlin, Berlin, Germany
- Ruhr-University Bochum, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Herne, North Rhine-Westphalia, Germany
| |
Collapse
|
6
|
Boucault L, Lopez Robles MD, Thiolat A, Bézie S, Schmueck-Henneresse M, Braudeau C, Vimond N, Freuchet A, Autrusseau E, Charlotte F, Redjoul R, Beckerich F, Leclerc M, Piaggio E, Josien R, Volk HD, Maury S, Cohen JL, Anegon I, Guillonneau C. Transient antibody targeting of CD45RC inhibits the development of graft-versus-host disease. Blood Adv 2020; 4:2501-2515. [PMID: 32511714 PMCID: PMC7284095 DOI: 10.1182/bloodadvances.2020001688] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is a widely spread treatment of many hematological diseases, but its most important side effect is graft-versus-host disease (GVHD). Despite the development of new therapies, acute GVHD (aGVHD) occurs in 30% to 50% of allogeneic BMT and is characterized by the generation of effector T (Teff) cells with production of inflammatory cytokines. We previously demonstrated that a short anti-CD45RC monoclonal antibody (mAb) treatment in a heart allograft rat model transiently decreased CD45RChigh Teff cells and increased regulatory T cell (Treg) number and function allowing long-term donor-specific tolerance. Here, we demonstrated in rat and mouse allogeneic GVHD, as well as in xenogeneic GVHD mediated by human T cells in NSG mice, that both ex vivo depletion of CD45RChigh T cells and in vivo treatment with short-course anti-CD45RC mAbs inhibited aGVHD. In the rat model, we demonstrated that long surviving animals treated with anti-CD45RC mAbs were fully engrafted with donor cells and developed a donor-specific tolerance. Finally, we validated the rejection of a human tumor in NSG mice infused with human cells and treated with anti-CD45RC mAbs. The anti-human CD45RC mAbs showed a favorable safety profile because it did not abolish human memory antiviral immune responses, nor trigger cytokine release in in vitro assays. Altogether, our results show the potential of a prophylactic treatment with anti-human CD45RC mAbs in combination with rapamycin as a new therapy to treat aGVHD without abolishing the antitumor effect.
Collapse
Affiliation(s)
- Laetitia Boucault
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Maria-Dolores Lopez Robles
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Allan Thiolat
- Université Paris-Est Créteil, INSERM, Institut Mondor de Recherche Biomédicale (IMRB), Creteil, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Michael Schmueck-Henneresse
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Nadège Vimond
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Frédéric Charlotte
- Service d'Anatomo-Pathologie, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Rabah Redjoul
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Florence Beckerich
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Mathieu Leclerc
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - Eliane Piaggio
- Translational Research Department, Institut Curie Research Center, Paris Sciences & Lettres (PSL) Research University, U932, INSERM, Paris, France; and
| | - Regis Josien
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
- Laboratoire d'Immunologie, Centre d'Immunomonitorage Nantes Atlantique (CIMNA), Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin/Berlin Institute of Health (BIH), Berlin, Germany
| | - Sébastien Maury
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Service d'Hematologie Clinique, Creteil, France
| | - José L Cohen
- Université Paris-Est Créteil, INSERM, IMRB, AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d'Investigation Clinique Biotherapie, Creteil, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie, Institut de Transplantation Urologie-Néphrologie (ITUN), Unité Mixte de Recherche (UMR) 1064, INSERM/Université de Nantes, Nantes, France
- Immunotherapy, Graft, Oncology (IGO), LabEx, Nantes, France
| |
Collapse
|
7
|
Amini L, Vollmer T, Wendering DJ, Jurisch A, Landwehr-Kenzel S, Otto NM, Jürchott K, Volk HD, Reinke P, Schmueck-Henneresse M. Comprehensive Characterization of a Next-Generation Antiviral T-Cell Product and Feasibility for Application in Immunosuppressed Transplant Patients. Front Immunol 2019; 10:1148. [PMID: 31191530 PMCID: PMC6546853 DOI: 10.3389/fimmu.2019.01148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 05/07/2019] [Indexed: 11/13/2022] Open
Abstract
Viral infections have a major impact on morbidity and mortality of immunosuppressed solid organ transplant (SOT) patients because of missing or failure of adequate pharmacologic antiviral treatment. Adoptive antiviral T-cell therapy (AVTT), regenerating disturbed endogenous T-cell immunity, emerged as an attractive alternative approach to combat severe viral complications in immunocompromised patients. AVTT is successful in patients after hematopoietic stem cell transplantation where T-cell products (TCPs) are manufactured from healthy donors. In contrast, in the SOT setting TCPs are derived from/applied back to immunosuppressed patients. We and others demonstrated feasibility of TCP generation from SOT patients and first clinical proof-of-concept trials revealing promising data. However, the initial efficacy is frequently lost long-term, because of limited survival of transferred short-lived T-cells indicating a need for next-generation TCPs. Our recent data suggest that Rapamycin treatment during TCP manufacture, conferring partial inhibition of mTOR, might improve its composition. The aim of this study was to confirm these promising observations in a setting closer to clinical challenges and to deeply characterize the next-generation TCPs. Using cytomegalovirus (CMV) as model, our next-generation Rapamycin-treated (Rapa-)TCP showed consistently increased proportions of CD4+ T-cells as well as CD4+ and CD8+ central-memory T-cells (TCM). In addition, Rapamycin sustained T-cell function despite withdrawal of Rapamycin, showed superior T-cell viability and resistance to apoptosis, stable metabolism upon activation, preferential expansion of TCM, partial conversion of other memory T-cell subsets to TCM and increased clonal diversity. On transcriptome level, we observed a gene expression profile denoting long-lived early memory T-cells with potent effector functions. Furthermore, we successfully applied the novel protocol for the generation of Rapa-TCPs to 19/19 SOT patients in a comparative study, irrespective of their history of CMV reactivation. Moreover, comparison of paired TCPs generated before/after transplantation did not reveal inferiority of the latter despite exposition to maintenance immunosuppression post-SOT. Our data imply that the Rapa-TCPs, exhibiting longevity and sustained T-cell memory, are a reasonable treatment option for SOT patients. Based on our success to manufacture Rapa-TCPs from SOT patients under maintenance immunosuppression, now, we seek ultimate clinical proof of efficacy in a clinical study.
Collapse
Affiliation(s)
- Leila Amini
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Tino Vollmer
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Desiree J Wendering
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Anke Jurisch
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany
| | - Sybille Landwehr-Kenzel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany.,Department for Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité University Medicine Berlin, Berlin, Germany
| | - Natalie Maureen Otto
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Karsten Jürchott
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Petra Reinke
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Institute for Medical Immunology, Charité University Medicine Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
8
|
Schmueck-Henneresse M, Omer B, Shum T, Tashiro H, Mamonkin M, Lapteva N, Sharma S, Rollins L, Dotti G, Reinke P, Volk HD, Rooney CM. Comprehensive Approach for Identifying the T Cell Subset Origin of CD3 and CD28 Antibody-Activated Chimeric Antigen Receptor-Modified T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:348-362. [PMID: 28550199 PMCID: PMC5536854 DOI: 10.4049/jimmunol.1601494] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/03/2017] [Indexed: 12/27/2022]
Abstract
The outcome of therapy with chimeric Ag receptor (CAR)-modified T cells is strongly influenced by the subset origin of the infused T cells. However, because polyclonally activated T cells acquire a largely CD45RO+CCR7- effector memory phenotype after expansion, regardless of subset origin, it is impossible to know which subsets contribute to the final T cell product. To determine the contribution of naive T cell, memory stem T cell, central memory T cell, effector memory T cell, and terminally differentiated effector T cell populations to the CD3 and CD28-activated CAR-modified T cells that we use for therapy, we followed the fate and function of individually sorted CAR-modified T cell subsets after activation with CD3 and CD28 Abs (CD3/28), transduction and culture alone, or after reconstitution into the relevant subset-depleted population. We show that all subsets are sensitive to CAR transduction, and each developed a distinct T cell functional profile during culture. Naive-derived T cells showed the greatest rate of proliferation but had more limited effector functions and reduced killing compared with memory-derived populations. When cultured in the presence of memory T cells, naive-derived T cells show increased differentiation, reduced effector cytokine production, and a reduced reproliferative response to CAR stimulation. CD3/28-activated T cells expanded in IL-7 and IL-15 produced greater expansion of memory stem T cells and central memory T cell-derived T cells compared with IL-2. Our strategy provides a powerful tool to elucidate the characteristics of CAR-modified T cells, regardless of the protocol used for expansion, reveals the functional properties of each expanded T cell subset, and paves the way for a more detailed evaluation of the effects of manufacturing changes on the subset contribution to in vitro-expanded T cells.
Collapse
Affiliation(s)
- Michael Schmueck-Henneresse
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany;
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
- Division of Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Thomas Shum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
- Graduate Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
- Graduate Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Lisa Rollins
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
| | - Gianpietro Dotti
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
| | - Petra Reinke
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
- Houston Methodist Hospital, Houston, TX 77030
- Texas Children's Hospital, Houston, TX 77030
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030; and
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
9
|
Liu J, Chang YJ, Yan CH, Xu LP, Jiang ZF, Zhang XH, Liu KY, Huang XJ. Poor CMV-specific CD8+ T central memory subset recovery at early stage post-HSCT associates with refractory and recurrent CMV reactivation. J Infect 2016; 73:261-70. [DOI: 10.1016/j.jinf.2016.04.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 04/07/2016] [Accepted: 04/21/2016] [Indexed: 11/25/2022]
|
10
|
Chen SF, Holmes TH, Slifer T, Ramachandran V, Mackey S, Hebson C, Arvin AM, Lewis DB, Dekker CL. Longitudinal Kinetics of Cytomegalovirus-Specific T-Cell Immunity and Viral Replication in Infants With Congenital Cytomegalovirus Infection. J Pediatric Infect Dis Soc 2016; 5:14-20. [PMID: 26908487 PMCID: PMC4765489 DOI: 10.1093/jpids/piu089] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 08/08/2014] [Indexed: 11/14/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) is reported to affect up to 1% of all live births in the United States. T-cell immunity may be important for controlling CMV replication in congenital CMV-infected infants. We describe the natural history of CMV-specific T-cell evolution and CMV replication in infants with congenital CMV infection. METHODS Cytomegalovirus viral load, CMV urine culture, and CMV-specific CD4 and CD8 T-cell responses were assessed in a prospective longitudinal cohort of 51 infants with congenital CMV infection who were observed from birth to 3 years of age. RESULTS We found a kinetic pattern of decreasing urinary CMV replication and increasing CMV-specific CD4 and CD8 T-cell responses during the first 3 years of life. We also found higher CMV-specific CD8 T-cell responses were associated with subsequent reduction of urine CMV viral load. CONCLUSION For infants with congenital CMV infection, our data suggest an age-related maturation of both CMV-specific CD4 and CD8 T-cell immunity that is associated with an age-related decline in urinary CMV replication.
Collapse
Affiliation(s)
| | - Tyson H. Holmes
- Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
| | | | | | | | - Cathleen Hebson
- Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, California
| | | | | | | |
Collapse
|
11
|
Abstract
Human cytomegalovirus (CMV) is the major cause of congenital neurological defects in the United States and also causes significant morbidity and mortality for hematopoietic and solid organ transplant patients. Primary infection in immunocompetent individuals rarely causes disease but resolves as a life-long latent infection, characterized by sustained antibody and cellular responses. Despite considerable efforts over the last 40 years to develop live attenuated and subunit vaccines, none is close to receiving regulatory approval. However, there is evidence that antibodies can prevent primary infection and cytotoxic T cells can suppress secondary infection. Prior maternal infection decreases the risk a fetus will contract CMV, while adoptive transfer of virus-specific CD8+ T cells is highly protective against CMV disease in hematopoietic stem cell transplant recipients. As a result, three polyclonal immunoglobulin preparations are approved for clinical use and one monoclonal antibody has reached phase III trials. Enhanced understanding of the viral life cycle from a biochemical perspective has revealed additional targets for neutralizing antibodies in the gH/gL/UL128-131 pentamer. Until an effective vaccine is licensed, passive immunotherapeutics may present an alternative to maintain viral loads and prevent CMV disease in susceptible populations. This review summarizes the progress and potential of immunotherapeutics to treat CMV infection.
Collapse
|
12
|
Roemhild A, Reinke P. Virus-specific T-cell therapy in solid organ transplantation. Transpl Int 2015; 29:515-26. [PMID: 26284570 DOI: 10.1111/tri.12659] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/07/2015] [Accepted: 08/12/2015] [Indexed: 12/12/2022]
Abstract
This article reviews the current state of T-cell therapy as therapeutic option for virus-associated diseases against the background of the most common viral complications and their standard treatment regimens after SOT. The available data of clinical T-cell trials in SOT are summarized. References to the hematopoietic stem cell transplantation are made if applicable data in SOT are not available and their content was considered likewise valid for cell therapy in SOT. Moreover, aspects of different manufacturing approaches including beneficial product characteristics and the importance of GMP compliance are addressed.
Collapse
Affiliation(s)
- Andy Roemhild
- Department of Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Department of Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
Weist BJD, Wehler P, El Ahmad L, Schmueck-Henneresse M, Millward JM, Nienen M, Neumann AU, Reinke P, Babel N. A revised strategy for monitoring BKV-specific cellular immunity in kidney transplant patients. Kidney Int 2015. [PMID: 26221751 DOI: 10.1038/ki.2015.215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactivation of Polyomavirus BKV is a severe complication in kidney transplant patients. Current treatment requires close monitoring, and modification of immunosuppressive drugs. As an important additional tool, the monitoring of BKV immunity has been based on detection of cytokine-secreting T cells upon BKV-antigen challenge. However, low frequent BKV-specific T cells are often barely detectable and their roles in BKV clearance remain unclear. Here, we analyzed the effects of immunosuppressive agents on BKV-specific T cells in vitro. Significant reductions in expression of several markers, and reduced killing functions upon treatment with calcineurin but not mTOR inhibitors were detected. However, effects of these drugs on expression of surface markers and GranzymeB were substantially less striking than effects on cytokine expression. Consequently, we applied a novel detection strategy for BKV-specific T cells in immunosuppressed kidney transplant patients using these more robust markers, and showed significantly improved sensitivity compared with the conventional IFNγ-based method. Using this strategy and 17-color flow cytometry, we found BKV-specific helper and cytolytic CD4+ T-cell subsets that differed in their memory phenotype, which corresponded with BKV clearance in kidney transplant patients. Thus, our results offer an improved detection strategy for BKV-specific T cells in kidney transplant patients, and shed light on the contributions of these cells to BKV clearance.
Collapse
Affiliation(s)
- Benjamin J D Weist
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Patrizia Wehler
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Linda El Ahmad
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | | | - Jason M Millward
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Mikalai Nienen
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Avidan U Neumann
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany.,Department of Nephrology, Charité Universitätsmedizin Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany.,Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| |
Collapse
|
14
|
Schmueck-Henneresse M, Sharaf R, Vogt K, Weist BJD, Landwehr-Kenzel S, Fuehrer H, Jurisch A, Babel N, Rooney CM, Reinke P, Volk HD. Peripheral blood-derived virus-specific memory stem T cells mature to functional effector memory subsets with self-renewal potency. THE JOURNAL OF IMMUNOLOGY 2015; 194:5559-67. [PMID: 25917088 DOI: 10.4049/jimmunol.1402090] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 03/27/2015] [Indexed: 12/31/2022]
Abstract
Memory T cells expressing stem cell-like properties have been described recently. The capacity of self-renewal and differentiation into various memory/effector subsets make them attractive for adoptive T cell therapy to combat severe virus infections and tumors. The very few reports on human memory stem T cells (T(SCM)) are restricted to analyses on polyclonal T cells, but extensive data on Ag-specific T(SCM )are missing. This might be due to their very low frequency limiting their enrichment and characterization. In this article, we provide functional and phenotypic data on human viral-specific T(SCM), defined as CD8(+)CD45RA(+)CCR7(+)CD127(+)CD95(+). Whereas <1% of total T cells express the T(SCM) phenotype, human CMV-specific T(SCM) can be detected at frequencies similar to those seen in other subsets, resulting in ∼ 1 /10,000 human CMV-specific T(SCM). A new virus-specific expansion protocol of sort-purified T(SCM) reveals both upregulation of various T cell subset markers and preservation of their stem cell phenotype in a significant proportion, indicating both self-renewal and differentiation potency of virus-specific T cells sharing their TCR repertoire. Furthermore, we describe a simplified culture protocol that allows fast expansion of virus-specific T(SCM) starting from a mixed naive T/T(SCM) pool of PBLs. Due to the clinical-grade compatibility, this might be the basis for novel cell therapeutic options in life-threatening courses of viral and tumor disease.
Collapse
Affiliation(s)
- Michael Schmueck-Henneresse
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany; Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany;
| | - Radwa Sharaf
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Katrin Vogt
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Benjamin J D Weist
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Sybille Landwehr-Kenzel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany; Department for Pediatric Pulmonology and Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Henrike Fuehrer
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Anke Jurisch
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany; Marien Hospital Herne, Ruhr University Bochum, D-44625 Herne, Germany; and
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030
| | - Petra Reinke
- Renal and Transplant Research Unit, Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| | - Hans-Dieter Volk
- Institute for Medical Immunology, Charité University Medicine Berlin, D-13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin, D-13353 Berlin, Germany
| |
Collapse
|
15
|
Gibson L, Barysauskas CM, McManus M, Dooley S, Lilleri D, Fisher D, Srivastava T, Diamond DJ, Luzuriaga K. Reduced frequencies of polyfunctional CMV-specific T cell responses in infants with congenital CMV infection. J Clin Immunol 2015; 35:289-301. [PMID: 25712611 PMCID: PMC4366322 DOI: 10.1007/s10875-015-0139-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 02/04/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE CMV infection remains a priority for vaccine development. Vaccination of infants could modify congenital infection and provide lifetime immunity. Properties of CMV-specific T cells associated with control of viral replication in early life have not been fully defined. METHODS CMV-specific CD4 and CD8 T cell responses were investigated in infants with congenital CMV infection and compared to adults with primary or chronic infection. PBMC were stimulated with UL83 (pp65) or UL122 (IE-2) peptide pools then stained with antibodies to markers of T cell subset (CD4 or CD8), phenotype (CD45RA, CCR7), or function (MIP1β, CD107, IFNγ, IL2) for flow cytometry analysis. RESULTS Detection of CMV pp65-specific CD4 T cells was less common in infants than adults. Responder cells were primarily effector memory (EM, CD45RA-CCR7-) in adults, but mixed memory subsets in infants. Detection of CMV pp65-specific CD8 T cells did not differ between the groups, but infants had lower frequencies of total responding cells and of MIP1β- or CD107-expressing cells. Responder cells were EM or effector memory RA (CD45RA + CCR7-) in all groups. Polyfunctional T cells were less commonly detected in infants than adults. Responses to IE-2 were detected in adults but not infants. All infants had detectable circulating CMV DNA at initial study (versus 60 % of adults with primary infection) despite longer duration of CMV infection. CONCLUSIONS Reduced frequencies and altered functional profile of CMV-specific CD4 and CD8 T cell responses were detected in infants compared to adults, and were associated with persistent CMV DNA in peripheral blood.
Collapse
Affiliation(s)
- Laura Gibson
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Weist BJD, Schmueck M, Fuehrer H, Sattler A, Reinke P, Babel N. The role of CD4(+) T cells in BKV-specific T cell immunity. Med Microbiol Immunol 2014; 203:395-408. [PMID: 25052009 DOI: 10.1007/s00430-014-0348-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/05/2014] [Indexed: 12/11/2022]
Abstract
Reactivation of polyomavirus BK (BKV) infection represents a severe complication in kidney transplant (KTX) patients. We previously reported an association between a declining BK viral load and the reconstitution of CD4(+) T cell BKV-specific immunity in patients following kidney transplantation. However, the specific contribution of CD4(+) T cells in the regulation of BKV-replication is unknown. Nevertheless, in vitro enrichment of BKV-specific T cells and subsequent adoptive T cell transfer may improve the restoration of immune competence in KTX patients with BKV infection. To date, strategies to capture human BKV-specific T cells with the ensuing expansion to clinically useful numbers are lacking. Here, we demonstrated a comprehensive flow cytometric analysis of the BKV-specific T cell response that permits access to the majority of T cells specific for immunodominant BKV antigens. A full-spectrum evaluation of the BKV-specific T cell response was performed by stimulating peripheral blood mononuclear cells (PBMC) with a mixture of BKV immunodominant peptide pools at varying concentrations and measuring activation marker expression and cytokine secretion. We also examined the effects of co-stimulation and PBMC resting time prior to activation. We defined the narrow range of stimulation conditions that permit the capture and expansion of functional BKV-specific T cell lines. The generated BKV-specific T cell lines showed the highest specificity and functionality when the T cells were captured according to IFNγ-secretion. This study highlights the multifunctional and cytolytic BKV-specific CD4(+) T cells as a dominant population within the generated T cell product. This method offers a novel approach for the generation of BKV-specific T cell lines for adoptive immunotherapy and underscores the critical role of CD4(+) T cells in the clearance of BKV.
Collapse
Affiliation(s)
- B J D Weist
- Department of Nephrology, Charité University Medicine, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Yamaji Y, Nakayama T. Recombinant measles viruses expressing respiratory syncytial virus proteins induced virus-specific CTL responses in cotton rats. Vaccine 2014; 32:4529-4536. [PMID: 24951869 DOI: 10.1016/j.vaccine.2014.06.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/27/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Respiratory syncytial virus (RSV) is a common cause of serious lower respiratory tract illnesses in infants. Natural infections with RSV provide limited protection against reinfection because of inefficient immunological responses that do not induce long-term memory. RSV natural infection has been shown to induce unbalanced immune response. The effective clearance of RSV is known to require the induction of a balanced Th1/Th2 immune response, which involves the induction of cytotoxic T lymphocytes (CTL). In our previous study, recombinant AIK-C measles vaccine strains MVAIK/RSV/F and MVAIK/RSV/G were developed, which expressed the RSV fusion (F) protein or glycoprotein (G). These recombinant viruses elicited antibody responses against RSV in cotton rats, and no infectious virus was recovered, but small amounts of infiltration of inflammatory cells were observed in the lungs following RSV challenge. In the present study, recombinant AIK-C measles vaccine strains MVAIK/RSV/M2-1 and MVAIK/RSV/NP were developed, expressing RSV M2-1 or Nucleoprotein (NP), respectively. These viruses exhibited temperature-sensitivity (ts), which was derived from AIK-C, and expressed respective RSV antigens. The intramuscular inoculation of cotton rats with the recombinant measles virus led to the induction of CD8(+) IFN-γ(+) cells. No infectious virus was recovered from a lung homogenate following the challenge. A Histological examination of the lungs revealed a significant reduction in inflammatory reactions without alveolar damage. These results support the recombinant measles viruses being effective vaccine candidates against RSV that induce RSV-specific CTL responses with or without the development of an antibody response.
Collapse
Affiliation(s)
- Yoshiaki Yamaji
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan
| | - Tetsuo Nakayama
- Laboratory of Viral Infection I, Kitasato Institute for Life Sciences, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
18
|
Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood 2014; 124:628-37. [PMID: 24855206 DOI: 10.1182/blood-2013-12-547349] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.
Collapse
|
19
|
Goéré D, Flament C, Rusakiewicz S, Poirier-Colame V, Kepp O, Martins I, Pesquet J, Eggermont A, Elias D, Chaput N, Zitvogel L. Potent Immunomodulatory Effects of the Trifunctional Antibody Catumaxomab. Cancer Res 2013; 73:4663-73. [DOI: 10.1158/0008-5472.can-12-4460] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|