1
|
Melocchi A, Schmittlein B, Sadhu S, Nayak S, Lares A, Uboldi M, Zema L, di Robilant BN, Feldman SA, Esensten JH. Automated manufacturing of cell therapies. J Control Release 2025; 381:113561. [PMID: 39993639 DOI: 10.1016/j.jconrel.2025.02.057] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Advanced therapy medicinal products (ATMPs), particularly genetically engineered cell-based therapies, are a major class of drugs with several high-profile Food and Drug Administration (FDA) approvals in the past decade. However, the high cost and limited production capacity of these drugs remain a barrier to access. These costs are primarily due to the complex manufacturing processes (often a single batch for a single patient), which increases personnel and facility expenses, and the challenges associated with tech-transfer from research and development stages to clinical-stage production. In order to scale up and scale out in a cost-effective way, automated solutions capable of multi-step manufacturing have been developed in academia and industry. The aim of the present article is to summarize the design approaches and key features of current multi-step automated systems for cell therapy manufacturing. For each system described in the literature, we will discuss different aspects in detail such as cell specificity, modularity, processing models, manufacturing locations, and integrated quality control. Our analysis highlights that developers need to balance competing needs in an environment where the biological, business, and technological factors are constantly evolving. Thus, designing engineering solutions that align with the pharmaceutical end-user is essential. Adopting a risk-based approach grounded in published data is the most effective strategy to evaluate existing and emerging automated systems.
Collapse
Affiliation(s)
- Alice Melocchi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy; Multiply Labs, San Francisco, CA, USA.
| | | | | | | | | | - Marco Uboldi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Lucia Zema
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | | | - Steven A Feldman
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan H Esensten
- Advanced Biotherapy Center (ABC), Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
2
|
Pashkina E, Blinova E, Bykova M, Aktanova A, Denisova V. Cell Therapy as a Way to Increase the Effectiveness of Hematopoietic Stem Cell Transplantation. Cells 2024; 13:2056. [PMID: 39768148 PMCID: PMC11675046 DOI: 10.3390/cells13242056] [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: 11/04/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a standard method for treating a number of pathologies, primarily blood diseases. Timely restoration of the immune system after HSCT is a critical factor associated with the development of complications such as relapses or secondary tumors and various infections, as well as the graft-versus-host reaction in allogeneic transplantation, which ultimately affects the survival of patients. Introduction into the recipient's body of immune system cells that are incapable of sensitization by recipient antigens during the period of immune reconstitution can increase the rate of restoration of the immune system, as well as reduce the risk of complications. This review presents the results of studies on cell therapy with various cell subpopulations of both bone marrow and mesenchymal origin during HSCT.
Collapse
Affiliation(s)
- Ekaterina Pashkina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Elena Blinova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| | - Maria Bykova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| | - Alina Aktanova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Vera Denisova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya st., 630099 Novosibirsk, Russia; (E.B.); (M.B.); (A.A.); (V.D.)
| |
Collapse
|
3
|
Braidotti S, Granzotto M, Curci D, Faganel Kotnik B, Maximova N. Advancing Allogeneic Hematopoietic Stem Cell Transplantation Outcomes through Immunotherapy: A Comprehensive Review of Optimizing Non-CAR Donor T-Lymphocyte Infusion Strategies. Biomedicines 2024; 12:1853. [PMID: 39200317 PMCID: PMC11351482 DOI: 10.3390/biomedicines12081853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Optimized use of prophylactic or therapeutic donor lymphocyte infusions (DLI) is aimed at improving clinical outcomes in patients with malignant and non-malignant hematological diseases who have undergone allogeneic hematopoietic stem cell transplantation (allo-HSCT). Memory T-lymphocytes (CD45RA-/CD45RO+) play a crucial role in immune reconstitution post-HSCT. The infusion of memory T cells is proven to be safe and effective in improving outcomes due to the enhanced reconstitution of immunity and increased protection against viremia, without exacerbating graft-versus-host disease (GVHD) risks. Studies indicate their persistence and efficacy in combating viral pathogens, suggesting a viable therapeutic avenue for patients. Conversely, using virus-specific T cells for viremia control presents challenges, such as regulatory hurdles, cost, and production time compared to CD45RA-memory T lymphocytes. Additionally, the modulation of regulatory T cells (Tregs) for therapeutic use has become an important area of investigation in GVHD, playing a pivotal role in immune tolerance modulation, potentially mitigating GVHD and reducing pharmacological immunosuppression requirements. Finally, donor T cell-mediated graft-versus-leukemia immune responses hold promise in curbing relapse rates post-HSCT, providing a multifaceted approach to therapeutic intervention in high-risk disease scenarios. This comprehensive review underscores the multifaceted roles of T lymphocytes in HSCT outcomes and identifies avenues for further research and clinical application.
Collapse
Affiliation(s)
- Stefania Braidotti
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Marilena Granzotto
- Azienda Sanitaria Universitaria Giuliano Isontina (ASU GI), 34125 Trieste, Italy;
| | - Debora Curci
- Advanced Translational Diagnostic Laboratory, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Barbara Faganel Kotnik
- Department of Hematology and Oncology, University Children’s Hospital, 1000 Ljubljana, Slovenia;
| | - Natalia Maximova
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| |
Collapse
|
4
|
Ekwe AP, Au R, Zhang P, McEnroe BA, Tan ML, Saldan A, Henden AS, Hutchins CJ, Henderson A, Mudie K, Kerr K, Fuery M, Kennedy GA, Hill GR, Tey SK. Clinical grade multiparametric cell sorting and gene-marking of regulatory T cells. Cytotherapy 2024; 26:719-728. [PMID: 38530690 DOI: 10.1016/j.jcyt.2024.02.023] [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: 07/28/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND AIMS Regulatory T cells (Tregs) are the main mediators of peripheral tolerance. Treg-directed therapy has shown promising results in preclinical studies of diverse immunopathologies. At present, the clinical applicability of adoptive Treg transfer is limited by difficulties in generating Tregs at sufficient cell dose and purity. METHODS We developed a Good Manufacturing Practice (GMP) compliant method based on closed-system multiparametric Fluorescence-Activated Cell Sorting (FACS) to purify Tregs, which are then expanded in vitro and gene-marked with a clinical grade retroviral vector to enable in vivo fate tracking. Following small-scale optimization, we conducted four clinical-scale processing runs. RESULTS We showed that Tregs could be enriched to 87- 92% purity following FACS-sorting, and expanded and transduced to yield clinically relevant cell dose of 136-732×106 gene-marked cells, sufficient for a cell dose of at least 2 × 106 cells/kg. The expanded Tregs were highly demethylated in the FOXP3 Treg-specific demethylated region (TSDR), consistent with bona fide natural Tregs. They were suppressive in vitro, but a small percentage could secrete proinflammatory cytokines, including interferon-γ and interleukin-17A. CONCLUSIONS This study demonstrated the feasibility of isolating, expanding and gene-marking Tregs in clinical scale, thus paving the way for future phase I trials that will advance knowledge about the in vivo fate of transferred Tregs and its relationship with concomitant Treg-directed pharmacotherapy and clinical response.
Collapse
Affiliation(s)
- Adaeze Precious Ekwe
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Kelvin Grove, Queensland, Australia
| | - Raymond Au
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ping Zhang
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Benjamin A McEnroe
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mei Ling Tan
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Alda Saldan
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andrea S Henden
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Cheryl J Hutchins
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Ashleigh Henderson
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Kari Mudie
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Keri Kerr
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Madonna Fuery
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Glen A Kennedy
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Siok-Keen Tey
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Kelvin Grove, Queensland, Australia; Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
5
|
Ullmann K, Manstein F, Triebert W, Kriedemann N, Franke A, Teske J, Mertens M, Lupanow V, Göhring G, Haase A, Martin U, Zweigerdt R. Matrix-free human pluripotent stem cell manufacturing by seed train approach and intermediate cryopreservation. Stem Cell Res Ther 2024; 15:89. [PMID: 38528578 DOI: 10.1186/s13287-024-03699-z] [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: 01/10/2024] [Accepted: 03/17/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs) have an enormous therapeutic potential, but large quantities of cells will need to be supplied by reliable, economically viable production processes. The suspension culture (three-dimensional; 3D) of hPSCs in stirred tank bioreactors (STBRs) has enormous potential for fuelling these cell demands. In this study, the efficient long-term matrix-free suspension culture of hPSC aggregates is shown. METHODS AND RESULTS STBR-controlled, chemical aggregate dissociation and optimized passage duration of 3 or 4 days promotes exponential hPSC proliferation, process efficiency and upscaling by a seed train approach. Intermediate high-density cryopreservation of suspension-derived hPSCs followed by direct STBR inoculation enabled complete omission of matrix-dependent 2D (two-dimensional) culture. Optimized 3D cultivation over 8 passages (32 days) cumulatively yielded ≈4.7 × 1015 cells, while maintaining hPSCs' pluripotency, differentiation potential and karyotype stability. Gene expression profiling reveals novel insights into the adaption of hPSCs to continuous 3D culture compared to conventional 2D controls. CONCLUSIONS Together, an entirely matrix-free, highly efficient, flexible and automation-friendly hPSC expansion strategy is demonstrated, facilitating the development of good manufacturing practice-compliant closed-system manufacturing in large scale.
Collapse
Affiliation(s)
- Kevin Ullmann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Felix Manstein
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Nils Kriedemann
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Annika Franke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jana Teske
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Mira Mertens
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Victoria Lupanow
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Gudrun Göhring
- Department of Human Genetics, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alexandra Haase
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic Transplantation and Vascular Surgery (HTTG), Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- REBIRTH Research Center for Translational and Regenerative Medicine, Hannover Medical School (MHH), Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
6
|
Eskandari SK, Daccache A, Azzi JR. Chimeric antigen receptor T reg therapy in transplantation. Trends Immunol 2024; 45:48-61. [PMID: 38123369 DOI: 10.1016/j.it.2023.11.005] [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: 10/09/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
In the quest for more precise and effective organ transplantation therapies, chimeric antigen receptor (CAR) regulatory T cell (Treg) therapies represent a potential cutting-edge advance. This review comprehensively analyses CAR Tregs and how they may address important drawbacks of polyclonal Tregs and conventional immunosuppressants. We examine a growing body of preclinical findings of CAR Treg therapy in transplantation, discuss CAR Treg design specifics, and explore established and attractive new targets in transplantation. In addition, we explore present impediments where future studies will be necessary to determine the efficacy of CAR Tregs in reshaping alloimmune responses and transplant microenvironments to reduce reliance on chemical immunosuppressants. Overall, ongoing studies and trials are crucial for understanding the full scope of CAR Treg therapy in transplantation.
Collapse
Affiliation(s)
- Siawosh K Eskandari
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Andrea Daccache
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Division of Bioscience Education and Research (UFR Biosciences), Claude Bernard University Lyon 1, Lyon, France
| | - Jamil R Azzi
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Baron KJ, Turnquist HR. Clinical Manufacturing of Regulatory T Cell Products For Adoptive Cell Therapy and Strategies to Improve Therapeutic Efficacy. Organogenesis 2023; 19:2164159. [PMID: 36681905 PMCID: PMC9870008 DOI: 10.1080/15476278.2022.2164159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Based on successes in preclinical animal transplant models, adoptive cell therapy (ACT) with regulatory T cells (Tregs) is a promising modality to induce allograft tolerance or reduce the use of immunosuppressive drugs to prevent rejection. Extensive work has been done in optimizing the best approach to manufacture Treg cell products for testing in transplant recipients. Collectively, clinical evaluations have demonstrated that large numbers of Tregs can be expanded ex vivo and infused safely. However, these trials have failed to induce robust drug-free tolerance and/or significantly reduce the level of immunosuppression needed to prevent solid organ transplant (SOTx) rejection. Improving Treg therapy effectiveness may require increasing Treg persistence or orchestrating Treg migration to secondary lymphatic tissues or places of inflammation. In this review, we describe current clinical Treg manufacturing methods used for clinical trials. We also highlight current strategies being implemented to improve delivered Treg ACT persistence and migration in preclinical studies.
Collapse
Affiliation(s)
- Kassandra J. Baron
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Infectious Disease and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Hēth R. Turnquist
- Departments of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA,CONTACT Hēth R. Turnquist Departments of Surgery, University of Pittsburgh School of Medicine, Thomas E. Starzl Transplantation Institute 200 Lothrop Street, BST W1542, PittsburghPA 15213, USA
| |
Collapse
|
8
|
Francis N, Braun M, Neagle S, Peiffer S, Bohn A, Rosenthal A, Olbrich T, Lollies S, Ilsmann K, Hauck C, Gerstmayer B, Weber S, Kirkpatrick A. Development of an automated manufacturing process for large-scale production of autologous T cell therapies. Mol Ther Methods Clin Dev 2023; 31:101114. [PMID: 37790245 PMCID: PMC10544074 DOI: 10.1016/j.omtm.2023.101114] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/13/2023] [Indexed: 10/05/2023]
Abstract
Engineered T cell therapies have shown significant clinical success. However, current manufacturing capabilities present a challenge in bringing these therapies to patients. Furthermore, the cost of development and manufacturing is still extremely high due to complexity of the manufacturing process. Increased automation can improve quality and reproducibility while reducing costs through minimizing hands-on operator time, allowing parallel manufacture of multiple products, and reducing the complexity of technology transfer. In this article, we describe the results of a strategic alliance between GSK and Miltenyi Biotec to develop a closed, automated manufacturing process using the CliniMACS Prodigy for autologous T cell therapy products that can deliver a high number of cells suitable for treating solid tumor indications and compatible with cryopreserved apheresis and drug product. We demonstrate the ability of the T cell Transduction - Large Scale process to deliver a significantly higher cell number than the existing process, achieving 1.5 × 1010 cells after 12 days of expansion, without affecting other product attributes. We demonstrate successful technology transfer of this robust process into three manufacturing facilities.
Collapse
Affiliation(s)
- Natalie Francis
- Cell & Gene Therapy, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Marion Braun
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Sarah Neagle
- Cell & Gene Therapy, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Sabine Peiffer
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Alexander Bohn
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Alexander Rosenthal
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Tanita Olbrich
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Sophia Lollies
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Keijo Ilsmann
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Carola Hauck
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Bernhard Gerstmayer
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Silvio Weber
- Cellular Therapy, Industrial Workflow Development, Miltenyi Biotec B.V. & Co. KG, Friedrich-Ebert-Str. 68, 51429 Bergisch Gladbach, Germany
| | - Aileen Kirkpatrick
- Cell & Gene Therapy, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| |
Collapse
|
9
|
Heo BY, Lee MW, Choi S, Jung Y, Pham TTD, Jang Y, Park JH, Kang S, Koh JS, Jo DY, Kwon J, Song IC. Autoimmune Limbic Encephalitis in Patients with Hematologic Malignancies after Haploidentical Hematopoietic Stem Cell Transplantation with Post-Transplant Cyclophosphamide. Cells 2023; 12:2049. [PMID: 37626859 PMCID: PMC10453524 DOI: 10.3390/cells12162049] [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: 07/13/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Autoimmune limbic encephalitis (LE) is a rare, but devastating complication of allogeneic hematopoietic stem cell transplantation (HSCT). There is currently limited evidence describing the risk factors, laboratory features, and underlying mechanisms of this neurologic adverse event. We retrospectively reviewed available clinical, imaging, and laboratory data from adult patients with hematological malignancies who underwent haploidentical HSCT with post-transplant cyclophosphamide (PTCy) at Chungnam National University Hospital from June 2016 to May 2020. Patients who developed LE were compared to those who did not based on clinical assessment, serum inflammatory biomarkers, and reconstitution of various T cell populations. Of 35 patients, 4 developed LE. There were no differences in patient demographics, donor demographics, or treatment conditions between patients that did and did not develop LE. Overall, patients with LE had worse clinical outcomes and overall survival than those without. In addition, they tended to have higher markers of systemic inflammation in the early post-transplant period, including fever, C-reactive protein (CRP), and cytokines. Remarkably, baseline interleukin-6 levels before HSCT were found to be higher in patients who developed LE than those who did not. In addition, analysis of T cell subsets showed impaired expansion of CD25+FOXP3+ regulatory T (Treg) cells in LE compared to non-LE patients despite appropriate reconstitution of the total CD4+ T cell population. Patients that developed LE within the first 30 days of HSCT were likely to have high serum IL-6 among other inflammatory cytokines coupled with suppression of regulatory T cell differentiation. Further work is needed on the mechanisms underlying impaired Treg expansion following HSCT and potential therapies.
Collapse
Affiliation(s)
- Bu Yeon Heo
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Myung-Won Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munwha-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Suyoung Choi
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Yunju Jung
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Thi Thuy Duong Pham
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Yunseon Jang
- Translational Immunology Institute, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Jung-Hyun Park
- Translational Immunology Institute, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Sora Kang
- Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munwha-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Jeong Suk Koh
- Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munwha-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Deog-Yeon Jo
- Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munwha-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Jaeyul Kwon
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Translational Immunology Institute, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Ik-Chan Song
- Department of Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, College of Medicine, Chungnam National University, 282 Munwha-ro, Jung-gu, Daejeon 35015, Republic of Korea
- Translational Immunology Institute, College of Medicine, Chungnam National University, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| |
Collapse
|
10
|
Bridgeman CJ, Shah SA, Oakes RS, Jewell CM. Dissecting regulatory T cell expansion using polymer microparticles presenting defined ratios of self-antigen and regulatory cues. Front Bioeng Biotechnol 2023; 11:1184938. [PMID: 37441198 PMCID: PMC10334287 DOI: 10.3389/fbioe.2023.1184938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Biomaterials allow for the precision control over the combination and release of cargo needed to engineer cell outcomes. These capabilities are particularly attractive as new candidate therapies to treat autoimmune diseases, conditions where dysfunctional immune cells create pathogenic tissue environments during attack of self-molecules termed self-antigens. Here we extend past studies showing combinations of a small molecule immunomodulator co-delivered with self-antigen induces antigen-specific regulatory T cells. In particular, we sought to elucidate how different ratios of these components loaded in degradable polymer particles shape the antigen presenting cell (APC) -T cell interactions that drive differentiation of T cells toward either inflammatory or regulatory phenotypes. Using rapamycin (rapa) as a modulatory cue and myelin self-peptide (myelin oligodendrocyte glycoprotein- MOG) - self-antigen attacked during multiple sclerosis (MS), we integrate these components into polymer particles over a range of ratios and concentrations without altering the physicochemical properties of the particles. Using primary cell co-cultures, we show that while all ratios of rapa:MOG significantly decreased expression of co-stimulation molecules on dendritic cells (DCs), these levels were insensitive to the specific ratio. During co-culture with primary T cell receptor transgenic T cells, we demonstrate that the ratio of rapa:MOG controls the expansion and differentiation of these cells. In particular, at shorter time points, higher ratios induce regulatory T cells most efficiently, while at longer time points the processes are not sensitive to the specific ratio. We also found corresponding changes in gene expression and inflammatory cytokine secretion during these times. The in vitro results in this study contribute to in vitro regulatory T cell expansion techniques, as well as provide insight into future studies to explore other modulatory effects of rapa such as induction of maintenance or survival cues.
Collapse
Affiliation(s)
- Christopher J. Bridgeman
- Fischell Department of Bioengineering, University of Maryland College Park, Baltimore, MD, United states
| | - Shrey A. Shah
- Fischell Department of Bioengineering, University of Maryland College Park, Baltimore, MD, United states
| | - Robert S. Oakes
- Fischell Department of Bioengineering, University of Maryland College Park, Baltimore, MD, United states
- United States Department of Veterans Affairs, Baltimore, MD, United states
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland College Park, Baltimore, MD, United states
- United States Department of Veterans Affairs, Baltimore, MD, United states
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, Baltimore, MD, United states
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, United states
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, United states
| |
Collapse
|
11
|
Zhang W, Chen Y, Zhao Z, Zheng H, Wang S, Liao Z, Sheng T, Zhao S, Hou W, Yu X, He F, Yu J, Zhang Y, Gu Z. Adoptive T reg therapy with metabolic intervention via perforated microneedles ameliorates psoriasis syndrome. SCIENCE ADVANCES 2023; 9:eadg6007. [PMID: 37196084 PMCID: PMC11803960 DOI: 10.1126/sciadv.adg6007] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023]
Abstract
Regulatory T (Treg) cells underlie multiple autoimmune disorders and potentialize an anti-inflammation treatment with adoptive cell therapy. However, systemic delivery of cellular therapeutics often lacks tissue targeting and accumulation for localized autoimmune diseases. Besides, the instability and plasticity of Treg cells also induce phenotype transition and functional loss, impeding clinical translation. Here, we developed a perforated microneedle (PMN) with favorable mechanical performance and a spacious encapsulation cavity to support cell survival, as well as tunable channels to facilitate cell migration for local Treg therapy of psoriasis. In addition, the enzyme-degradable microneedle matrix could release fatty acid in the hyperinflammatory area of psoriasis, enhancing the Treg suppressive functions via the fatty acid oxidation (FAO)-mediated metabolic intervention. Treg cells administered through PMN substantially ameliorated psoriasis syndrome with the assistance of fatty acid-mediated metabolic intervention in a psoriasis mouse model. This tailorable PMN could offer a transformative platform for local cell therapy to treat a variety of diseases.
Collapse
Affiliation(s)
- Wentao Zhang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingxin Chen
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Institute of Advanced Magnetic Materials and International Research Center for EM Metamaterials, College of Materials and Environmental Engineering, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Zhengjie Zhao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hanqi Zheng
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenqiang Wang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziyan Liao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Sheng
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sheng Zhao
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenhui Hou
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinmin Yu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fang He
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jicheng Yu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yuqi Zhang
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhen Gu
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
12
|
Amini L, Kaeda J, Fritsche E, Roemhild A, Kaiser D, Reinke P. Clinical adoptive regulatory T Cell therapy: State of the art, challenges, and prospective. Front Cell Dev Biol 2023; 10:1081644. [PMID: 36794233 PMCID: PMC9924129 DOI: 10.3389/fcell.2022.1081644] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/29/2022] [Indexed: 02/01/2023] Open
Abstract
Rejection of solid organ transplant and graft versus host disease (GvHD) continue to be challenging in post transplantation management. The introduction of calcineurin inhibitors dramatically improved recipients' short-term prognosis. However, long-term clinical outlook remains poor, moreover, the lifelong dependency on these toxic drugs leads to chronic deterioration of graft function, in particular the renal function, infections and de-novo malignancies. These observations led investigators to identify alternative therapeutic options to promote long-term graft survival, which could be used concomitantly, but preferably, replace pharmacologic immunosuppression as standard of care. Adoptive T cell (ATC) therapy has evolved as one of the most promising approaches in regenerative medicine in the recent years. A range of cell types with disparate immunoregulatory and regenerative properties are actively being investigated as potential therapeutic agents for specific transplant rejection, autoimmunity or injury-related indications. A significant body of data from preclinical models pointed to efficacy of cellular therapies. Significantly, early clinical trial observations have confirmed safety and tolerability, and yielded promising data in support of efficacy of the cellular therapeutics. The first class of these therapeutic agents commonly referred to as advanced therapy medicinal products have been approved and are now available for clinical use. Specifically, clinical trials have supported the utility of CD4+CD25+FOXP3+ regulatory T cells (Tregs) to minimize unwanted or overshooting immune responses and reduce the level of pharmacological immunosuppression in transplant recipients. Tregs are recognized as the principal orchestrators of maintaining peripheral tolerance, thereby blocking excessive immune responses and prevent autoimmunity. Here, we summarize rationale for the adoptive Treg therapy, challenges in manufacturing and clinical experiences with this novel living drug and outline future perspectives of its use in transplantation.
Collapse
Affiliation(s)
- Leila Amini
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany,Berlin Institute of Health—Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jaspal Kaeda
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Enrico Fritsche
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andy Roemhild
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Kaiser
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany,Berlin Institute of Health—Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany,*Correspondence: Petra Reinke,
| |
Collapse
|
13
|
Zhu X, Li Q, George V, Spanoudis C, Gilkes C, Shrestha N, Liu B, Kong L, You L, Echeverri C, Li L, Wang Z, Chaturvedi P, Muniz GJ, Egan JO, Rhode PR, Wong HC. A novel interleukin-2-based fusion molecule, HCW9302, differentially promotes regulatory T cell expansion to treat atherosclerosis in mice. Front Immunol 2023; 14:1114802. [PMID: 36761778 PMCID: PMC9907325 DOI: 10.3389/fimmu.2023.1114802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease caused by deposition of oxidative low-density lipoprotein (LDL) in the arterial intima which triggers the innate immune response through myeloid cells such as macrophages. Regulatory T cells (Tregs) play an important role in controlling the progression or regression of atherosclerosis by resolving macrophage-mediated inflammatory functions. Interleukin-2 (IL-2) signaling is essential for homeostasis of Tregs. Since recombinant IL-2 has an unfavorable pharmacokinetic profile limiting its therapeutic use, we constructed a fusion protein, designated HCW9302, containing two IL-2 domains linked by an extracellular tissue factor domain. We found that HCW9302 exhibited a longer serum half-life with an approximately 1000-fold higher affinity for the IL-2Rα than IL-2. HCW9302 could be administered to mice at a dosing range that expanded and activated Tregs but not CD4+ effector T cells. In an ApoE-/- mouse model, HCW9302 treatment curtailed the progression of atherosclerosis through Treg activation and expansion, M2 macrophage polarization and myeloid-derived suppressor cell induction. HCW9302 treatment also lessened inflammatory responses in the aorta. Thus, HCW9302 is a potential therapeutic agent to expand and activate Tregs for treatment of inflammatory and autoimmune diseases.
Collapse
|
14
|
Regulatory T Cells: Liquid and Living Precision Medicine for the Future of VCA. Transplantation 2023; 107:86-97. [PMID: 36210500 DOI: 10.1097/tp.0000000000004342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Transplant rejection remains a challenge especially in the field of vascularized composite allotransplantation (VCA). To blunt the alloreactive immune response' stable levels of maintenance immunosupression are required. However' the need for lifelong immunosuppression poses the risk of severe side effects, such as increased risk of infection, metabolic complications, and malignancies. To balance therapeutic efficacy and medication side effects, immunotolerance promoting immune cells (especially regulatory T cells [Treg]) have become of great scientific interest. This approach leverages immune system mechanisms that usually ensure immunotolerance toward self-antigens and prevent autoimmunopathies. Treg can be bioengineered to express a chimeric antigen receptor or a T-cell receptor. Such bioengineered Treg can target specific antigens and thereby reduce unwanted off-target effects. Treg have demonstrated beneficial clinical effects in solid organ transplantation and promising in vivo data in VCAs. In this review, we summarize the functional, phenotypic, and immunometabolic characteristics of Treg and outline recent advancements and current developments regarding Treg in the field of VCA and solid organ transplantation.
Collapse
|
15
|
Gotti E, Tettamanti S, Zaninelli S, Cuofano C, Cattaneo I, Rotiroti MC, Cribioli S, Alzani R, Rambaldi A, Introna M, Golay J. Optimization of therapeutic T cell expansion in G-Rex device and applicability to large-scale production for clinical use. Cytotherapy 2022; 24:334-343. [DOI: 10.1016/j.jcyt.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 02/02/2023]
|
16
|
Particles in Biopharmaceutical Formulations, Part 2: An Update on Analytical Techniques and Applications for Therapeutic Proteins, Viruses, Vaccines and Cells. J Pharm Sci 2021; 111:933-950. [PMID: 34919969 DOI: 10.1016/j.xphs.2021.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 11/21/2022]
Abstract
Particles in biopharmaceutical formulations remain a hot topic in drug product development. With new product classes emerging it is crucial to discriminate particulate active pharmaceutical ingredients from particulate impurities. Technical improvements, new analytical developments and emerging tools (e.g., machine learning tools) increase the amount of information generated for particles. For a proper interpretation and judgment of the generated data a thorough understanding of the measurement principle, suitable application fields and potential limitations and pitfalls is required. Our review provides a comprehensive overview of novel particle analysis techniques emerging in the last decade for particulate impurities in therapeutic protein formulations (protein-related, excipient-related and primary packaging material-related), as well as particulate biopharmaceutical formulations (virus particles, virus-like particles, lipid nanoparticles and cell-based medicinal products). In addition, we review the literature on applications, describe specific analytical approaches and illustrate advantages and drawbacks of currently available techniques for particulate biopharmaceutical formulations.
Collapse
|
17
|
Lentiviral Vectors for T Cell Engineering: Clinical Applications, Bioprocessing and Future Perspectives. Viruses 2021; 13:v13081528. [PMID: 34452392 PMCID: PMC8402758 DOI: 10.3390/v13081528] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/11/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
Lentiviral vectors have played a critical role in the emergence of gene-modified cell therapies, specifically T cell therapies. Tisagenlecleucel (Kymriah), axicabtagene ciloleucel (Yescarta) and most recently brexucabtagene autoleucel (Tecartus) are examples of T cell therapies which are now commercially available for distribution after successfully obtaining EMA and FDA approval for the treatment of blood cancers. All three therapies rely on retroviral vectors to transduce the therapeutic chimeric antigen receptor (CAR) into T lymphocytes. Although these innovations represent promising new therapeutic avenues, major obstacles remain in making them readily available tools for medical care. This article reviews the biological principles as well as the bioprocessing of lentiviral (LV) vectors and adoptive T cell therapy. Clinical and engineering successes, shortcomings and future opportunities are also discussed. The development of Good Manufacturing Practice (GMP)-compliant instruments, technologies and protocols will play an essential role in the development of LV-engineered T cell therapies.
Collapse
|
18
|
Grabarek AD, Jiskoot W, Hawe A, Pike-Overzet K, Menzen T. Forced degradation of cell-based medicinal products guided by flow imaging microscopy: Explorative studies with Jurkat cells. Eur J Pharm Biopharm 2021; 167:38-47. [PMID: 34274457 DOI: 10.1016/j.ejpb.2021.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/30/2021] [Accepted: 07/10/2021] [Indexed: 01/01/2023]
Abstract
Cell-based medicinal products (CBMPs) offer ground-breaking opportunities to treat diseases with limited or no therapeutic options. However, the intrinsic complexity of CBMPs results in great challenges with respect to analytical characterization and stability assessment. In our study, we submitted Jurkat cell suspensions to forced degradation studies mimicking conditions to which CBMPs might be exposed from procurement of cells to administration of the product. Flow imaging microscopy assisted by machine learning was applied for determination of cell viability and concentration, and quantification of debris particles. Additionally, orthogonal cell characterization techniques were used. Thawing of cells at 5 °C was detrimental to cell viability and resulted in high numbers of debris particles, in contrast to thawing at 37 °C or 20 °C which resulted in better stability. After freezing of cell suspensions at -18 °C in presence of dimethyl sulfoxide (DMSO), a DMSO concentration of 2.5% (v/v) showed low stabilizing properties, whereas 5% or 10% was protective. Horizontal shaking of cell suspensions did not affect cell viability, but led to a reduction in cell concentration. Fetal bovine serum (10% [v/v]) protected the cells during shaking. In conclusion, forced degradation studies with application of orthogonal analytical characterization methods allow for CBMP stability assessment and formulation screening.
Collapse
Affiliation(s)
- A D Grabarek
- Coriolis Pharma, Fraunhoferstraße 18 b, 82152 Martinsried, Germany; Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - W Jiskoot
- Coriolis Pharma, Fraunhoferstraße 18 b, 82152 Martinsried, Germany; Leiden Academic Centre for Drug Research, Leiden University, the Netherlands.
| | - A Hawe
- Leiden Academic Centre for Drug Research, Leiden University, the Netherlands
| | - K Pike-Overzet
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - T Menzen
- Leiden Academic Centre for Drug Research, Leiden University, the Netherlands.
| |
Collapse
|
19
|
Jones M, Nankervis B, Roballo KS, Pham H, Bushman J, Coeshott C. A Comparison of Automated Perfusion- and Manual Diffusion-Based Human Regulatory T Cell Expansion and Functionality Using a Soluble Activator Complex. Cell Transplant 2021; 29:963689720923578. [PMID: 32662685 PMCID: PMC7586259 DOI: 10.1177/0963689720923578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Absence or reduced frequency of human regulatory T cells (Tregs) can limit the control of inflammatory responses, autoimmunity, and the success of transplant engraftment. Clinical studies indicate that use of Tregs as immunotherapeutics would require billions of cells per dose. The Quantum® Cell Expansion System (Quantum system) is a hollow-fiber bioreactor that has previously been used to grow billions of functional T cells in a short timeframe, 8–9 d. Here we evaluated expansion of selected Tregs in the Quantum system using a soluble activator to compare the effects of automated perfusion with manual diffusion-based culture in flasks. Treg CD4+CD25+ cells from three healthy donors, isolated via column-free immunomagnetic negative/positive selection, were grown under static conditions and subsequently seeded into Quantum system bioreactors and into T225 control flasks in an identical culture volume of PRIME-XV XSFM medium with interleukin-2, for a 9-d expansion using a soluble anti-CD3/CD28/CD2 monoclonal antibody activator complex. Treg harvests from three parallel expansions produced a mean of 3.95 × 108 (range 1.92 × 108 to 5.58 × 108) Tregs in flasks (mean viability 71.3%) versus 7.00 × 109 (range 3.57 × 109 to 13.00 × 109) Tregs in the Quantum system (mean viability 91.8%), demonstrating a mean 17.7-fold increase in Treg yield for the Quantum system over that obtained in flasks. The two culture processes gave rise to cells with a memory Treg CD4+CD25+FoxP3+CD45RO+ phenotype of 93.7% for flasks versus 97.7% for the Quantum system. Tregs from the Quantum system demonstrated an 8-fold greater interleukin-10 stimulation index than cells from flask culture following restimulation. Quantum system–expanded Tregs proliferated, maintained their antigenic phenotype, and suppressed effector immune cells after cryopreservation. We conclude that an automated perfusion bioreactor can support the scale-up expansion of functional Tregs more efficiently than diffusion-based flask culture.
Collapse
Affiliation(s)
| | | | | | - Huong Pham
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Jared Bushman
- School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | | |
Collapse
|
20
|
Baeten P, Van Zeebroeck L, Kleinewietfeld M, Hellings N, Broux B. Improving the Efficacy of Regulatory T Cell Therapy. Clin Rev Allergy Immunol 2021; 62:363-381. [PMID: 34224053 PMCID: PMC8256646 DOI: 10.1007/s12016-021-08866-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 12/11/2022]
Abstract
Autoimmunity is caused by an unbalanced immune system, giving rise to a variety of organ-specific to system disorders. Patients with autoimmune diseases are commonly treated with broad-acting immunomodulatory drugs, with the risk of severe side effects. Regulatory T cells (Tregs) have the inherent capacity to induce peripheral tolerance as well as tissue regeneration and are therefore a prime candidate to use as cell therapy in patients with autoimmune disorders. (Pre)clinical studies using Treg therapy have already established safety and feasibility, and some show clinical benefits. However, Tregs are known to be functionally impaired in autoimmune diseases. Therefore, ex vivo manipulation to boost and stably maintain their suppressive function is necessary when considering autologous transplantation. Similar to autoimmunity, severe coronavirus disease 2019 (COVID-19) is characterized by an exaggerated immune reaction and altered Treg responses. In light of this, Treg-based therapies are currently under investigation to treat severe COVID-19. This review provides a detailed overview of the current progress and clinical challenges of Treg therapy for autoimmune and hyperinflammatory diseases, with a focus on recent successes of ex vivo Treg manipulation.
Collapse
Affiliation(s)
- Paulien Baeten
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Lauren Van Zeebroeck
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium.,VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Markus Kleinewietfeld
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium.,VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Niels Hellings
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Bieke Broux
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium. .,University MS Center, Campus Diepenbeek, Diepenbeek, Belgium. .,Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
21
|
Guo WW, Su XH, Wang MY, Han MZ, Feng XM, Jiang EL. Regulatory T Cells in GVHD Therapy. Front Immunol 2021; 12:697854. [PMID: 34220860 PMCID: PMC8250864 DOI: 10.3389/fimmu.2021.697854] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022] Open
Abstract
Graft versus host disease (GVHD) is a common complication and the leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Pharmacological immunosuppression used in GVHD prophylaxis and treatment lacks specificity and can increase the likelihood of infection and relapse. Regulatory T lymphocytes (Tregs) play a vital role in restraining excessive immune responses and inducing peripheral immune tolerance. In particular, clinical trials have demonstrated that Tregs can prevent and treat GVHD, without increasing the risk of relapse and infection. Hence, adoptive transfer of Tregs to control GVHD using their immunosuppressive properties represents a promising therapeutic approach. To optimally apply Tregs for control of GVHD, a thorough understanding of their biology is necessary. In this review, we describe the biological characteristics of Tregs, including how the stability of FOXP3 expression can be maintained. We will also discuss the mechanisms underlying Tregs-mediated modulation of GVHD and approaches to effectively increase Tregs’ numbers. Finally, we will examine the developing trends in the use of Tregs for clinical therapy.
Collapse
Affiliation(s)
- Wen-Wen Guo
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiu-Hua Su
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming-Yang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming-Zhe Han
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiao-Ming Feng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Er-Lie Jiang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
22
|
Lu J, Li P, Du X, Liu Y, Zhang B, Qi F. Regulatory T cells induce transplant immune tolerance. Transpl Immunol 2021; 67:101411. [PMID: 34020045 DOI: 10.1016/j.trim.2021.101411] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 01/03/2023]
Abstract
Organ transplantation is the preferred treatment option for end-stage organ failure. Although immunosuppressants are effective for preventing the occurrence of acute rejection, they also cause a series of side effects in transplant recipients. To improve the quality of patient survival, a new therapeutic strategy that has fewer side effects than current immunosuppressive regimens and can induce allograft immune tolerance and effectively prevent transplant rejection is needed. In this context, regulatory T cells (Tregs) are considered to be promising research targets. With the increasing understanding of the immunomodulatory role of Tregs, the use of Treg-based cellular therapies has shifted from prevention/treatment of autoimmune diseases to clinical trials for organ transplantation. This review describes the phenotype and in vitro expansion of Tregs and the mechanisms by which they exert immunomodulatory effects in transplantation immunity, highlights recent clinical trial data on Treg-based cellular therapies in transplantation, and describes future directions and limitations.
Collapse
Affiliation(s)
- Jian Lu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, China.
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| | - Xuezhi Du
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| | - Yanhong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| | - Baotong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China.
| |
Collapse
|
23
|
Garcia-Aponte OF, Herwig C, Kozma B. Lymphocyte expansion in bioreactors: upgrading adoptive cell therapy. J Biol Eng 2021; 15:13. [PMID: 33849630 PMCID: PMC8042697 DOI: 10.1186/s13036-021-00264-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/29/2021] [Indexed: 12/25/2022] Open
Abstract
Bioreactors are essential tools for the development of efficient and high-quality cell therapy products. However, their application is far from full potential, holding several challenges when reconciling the complex biology of the cells to be expanded with the need for a manufacturing process that is able to control cell growth and functionality towards therapy affordability and opportunity. In this review, we discuss and compare current bioreactor technologies by performing a systematic analysis of the published data on automated lymphocyte expansion for adoptive cell therapy. We propose a set of requirements for bioreactor design and identify trends on the applicability of these technologies, highlighting the specific challenges and major advancements for each one of the current approaches of expansion along with the opportunities that lie in process intensification. We conclude on the necessity to develop targeted solutions specially tailored for the specific stimulation, supplementation and micro-environmental needs of lymphocytes’ cultures, and the benefit of applying knowledge-based tools for process control and predictability.
Collapse
Affiliation(s)
- Oscar Fabian Garcia-Aponte
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria
| | - Christoph Herwig
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria.
| | - Bence Kozma
- Research Area Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorferstraße 1a, 1060, Vienna, Austria
| |
Collapse
|
24
|
Mukhatayev Z, Ostapchuk YO, Fang D, Le Poole IC. Engineered antigen-specific regulatory T cells for autoimmune skin conditions. Autoimmun Rev 2021; 20:102761. [PMID: 33476816 DOI: 10.1016/j.autrev.2021.102761] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/28/2020] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) are a subset of T cells responsible for the regulation of immune responses, thereby maintaining immune homeostasis and providing immune tolerance to both self and non-self-antigens. An increasing number of studies revealed Treg numbers and functions in a variety of autoimmune diseases. Treg deficiency can cause the development of several autoimmune skin diseases including vitiligo, alopecia areata, pemphigoid and pemphigus, psoriasis, and systemic sclerosis. Many clinical trials have been performed for autoimmune conditions using polyclonal Tregs, but efficiency can be significantly improved using antigen-specific Tregs engineered using T cell receptor (TCR) or chimeric antigen receptor (CAR) constructs. In this review, we systematically reviewed altered frequencies, impaired functions, and phenotypic features of Tregs in autoimmune skin conditions. We also summarized new advances in TCR and CAR based antigen-specific Tregs tested both in animal models and in clinics. The advantages and limitations of each approach were carefully discussed emphasizing possible clinical relevance to patients with autoimmune skin diseases. Moreover, we have reviewed potential approaches for engineering antigen-specific Tregs, and strategies for overcoming possible hurdles in clinical applications. Thereby, antigen-specific Tregs can be infused using autologous adoptive cell transfer to restore Treg numbers and to provide local immune tolerance for autoimmune skin disorders.
Collapse
Affiliation(s)
- Zhussipbek Mukhatayev
- Department of Dermatology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA; Department of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan; M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | | - Deyu Fang
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - I Caroline Le Poole
- Department of Dermatology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
25
|
Schwarze LI, Sonntag T, Wild S, Schmitz S, Uhde A, Fehse B. Automated production of CCR5-negative CD4 +-T cells in a GMP-compatible, clinical scale for treatment of HIV-positive patients. Gene Ther 2021; 28:572-587. [PMID: 33867524 PMCID: PMC8455337 DOI: 10.1038/s41434-021-00259-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 02/02/2023]
Abstract
Ex-vivo gene editing in T lymphocytes paves the way for novel concepts of immunotherapy. One of those strategies is directed at the protection of CD4+-T helper cells from HIV infection in HIV-positive individuals. To this end, we have developed and optimised a CCR5-targeting TALE nuclease, CCR5-Uco-hetTALEN, mediating high-efficiency knockout of C-C motif chemokine receptor 5 (CCR5), the HIV co-receptor essential during initial infection. Clinical translation of the knockout approach requires up-scaling of the manufacturing process to clinically relevant cell numbers in accordance with good manufacturing practice (GMP). Here we present a GMP-compatible mRNA electroporation protocol for the automated production of CCR5-edited CD4+-T cells in the closed CliniMACS Prodigy system. The automated process reliably produced high amounts of CCR5-edited CD4+-T cells (>1.5 × 109 cells with >60% CCR5 editing) within 12 days. Of note, about 40% of total large-scale produced cells showed a biallelic CCR5 editing, and between 25 and 42% of produced cells had a central memory T-cell phenotype. In conclusion, transfection of primary T cells with CCR5-Uco-hetTALEN mRNA is readily scalable for GMP-compatible production and hence suitable for application in HIV gene therapy.
Collapse
Affiliation(s)
- Lea Isabell Schwarze
- grid.13648.380000 0001 2180 3484Research Department Cell and Gene Therapy, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany ,grid.452463.2German Centre for Infection Research (DZIF), partner site, Hamburg, Germany
| | - Tanja Sonntag
- grid.13648.380000 0001 2180 3484Research Department Cell and Gene Therapy, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wild
- grid.59409.310000 0004 0552 5033Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Sabrina Schmitz
- grid.59409.310000 0004 0552 5033Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Almut Uhde
- grid.13648.380000 0001 2180 3484Research Department Cell and Gene Therapy, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- grid.13648.380000 0001 2180 3484Research Department Cell and Gene Therapy, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany ,grid.452463.2German Centre for Infection Research (DZIF), partner site, Hamburg, Germany
| |
Collapse
|
26
|
Fritsche E, Volk HD, Reinke P, Abou-El-Enein M. Toward an Optimized Process for Clinical Manufacturing of CAR-Treg Cell Therapy. Trends Biotechnol 2020; 38:1099-1112. [DOI: 10.1016/j.tibtech.2019.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/21/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
|
27
|
de Kivit S, Mensink M, Hoekstra AT, Berlin I, Derks RJE, Both D, Aslam MA, Amsen D, Berkers CR, Borst J. Stable human regulatory T cells switch to glycolysis following TNF receptor 2 costimulation. Nat Metab 2020; 2:1046-1061. [PMID: 32958937 DOI: 10.1038/s42255-020-00271-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 07/27/2020] [Indexed: 01/08/2023]
Abstract
Following activation, conventional T (Tconv) cells undergo an mTOR-driven glycolytic switch. Regulatory T (Treg) cells reportedly repress the mTOR pathway and avoid glycolysis. However, here we demonstrate that human thymus-derived Treg (tTreg) cells can become glycolytic in response to tumour necrosis factor receptor 2 (TNFR2) costimulation. This costimulus increases proliferation and induces a glycolytic switch in CD3-activated tTreg cells, but not in Tconv cells. Glycolysis in CD3-TNFR2-activated tTreg cells is driven by PI3-kinase-mTOR signalling and supports tTreg cell identity and suppressive function. In contrast to glycolytic Tconv cells, glycolytic tTreg cells do not show net lactate secretion and shuttle glucose-derived carbon into the tricarboxylic acid cycle. Ex vivo characterization of blood-derived TNFR2hiCD4+CD25hiCD127lo effector T cells, which were FOXP3+IKZF2+, revealed an increase in glucose consumption and intracellular lactate levels, thus identifying them as glycolytic tTreg cells. Our study links TNFR2 costimulation in human tTreg cells to metabolic remodelling, providing an additional avenue for drug targeting.
Collapse
Affiliation(s)
- Sander de Kivit
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mark Mensink
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
- Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anna T Hoekstra
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Ilana Berlin
- Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rico J E Derks
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Demi Both
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Muhammad A Aslam
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Derk Amsen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands.
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jannie Borst
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.
- Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
28
|
Ulbar F, Villanova I, Giancola R, Baldoni S, Guardalupi F, Fabi B, Olioso P, Capone A, Sola R, Ciardelli S, Del Papa B, Brattelli A, Ricciardi I, Taricani S, Sabbatinelli G, Iuliani O, Passeri C, Sportoletti P, Santarone S, Pierini A, Calabrese G, Falzetti F, Bonfini T, Accorsi P, Ruggeri L, Martelli MF, Velardi A, Di Ianni M. Clinical-Grade Expanded Regulatory T Cells Are Enriched with Highly Suppressive Cells Producing IL-10, Granzyme B, and IL-35. Biol Blood Marrow Transplant 2020; 26:2204-2210. [PMID: 32961369 DOI: 10.1016/j.bbmt.2020.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
Abstract
In the setting of T cell-depleted, full-haplotype mismatched transplantation, adoptive immunotherapy with regulatory T cells (Tregs) and conventional T cells (Tcons) can prevent graft-versus-host disease (GVHD) and improve post-transplantation immunologic reconstitution and is associated with a powerful graft-versus-leukemia effect. To improve the purity and the quantity of the infused Tregs, good manufacturing practices (GMP)-compatible expansion protocols are needed. Here we expanded Tregs using an automated, clinical-grade protocol. Cells were extensively characterized in vitro, and their efficiency was tested in vivo in a mouse model. Tregs were selected by CliniMacs (CD4+CD25+, 94.5 ± 6.3%; FoxP3+, 63.7 ± 11.5%; CD127+, 20 ± 3%; suppressive activity, 60 ± 7%), and an aliquot of 100 × 106 was expanded for 14 days using the CliniMACS Prodigy System, obtaining 684 ± 279 × 106 cells (CD4+CD25+, 99.6 ± 0.2%; FoxP3+, 82 ± 8%; CD127+, 1.1 ± 0.8%; suppressive activity, 75 ± 12%). CD39 and CTLA4 expression levels increased from 22.4 ± 12% to 58.1 ± 13.3% (P < .05) and from 20.4 ± 6.7% to 85.4 ± 9.8% (P < .01), respectively. TIM3 levels increased from .4 ± .05% to 29 ± 16% (P < .05). Memory Tregs were the prevalent population, whereas naive Tregs almost disappeared at the end of the culture. mRNA analysis displayed significant increases in CD39, IL-10, granzyme B, and IL-35 levels at the end of culture period (P < .05). Conversely, IFNγ expression decreased significantly by day +14. Expanded Tregs were sorted according to TIM3, CD39, and CD62L expression levels (purity >95%). When sorted populations were analyzed, TIM3+ cells showed significant increases in IL-10 and granzyme B (P < .01) .When expanded Tregs were infused in an NSG murine model, mice that received Tcons only died of GVHD, whereas mice that received both Tcons and Tregs survived without GVHD. GMP grade expanded cells that display phenotypic and functional Treg characteristics can be obtained using a fully automated system. Treg suppression is mediated by multiple overlapping mechanisms (eg, CTLA-4, CD39, IL-10, IL-35, TGF-β, granzyme B). TIM3+ cells emerge as a potentially highly suppressive population. © 2020 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Francesca Ulbar
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Ida Villanova
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | | | - Stefano Baldoni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Francesco Guardalupi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Bianca Fabi
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Paola Olioso
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Anita Capone
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Rosaria Sola
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Sara Ciardelli
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | | | - Ilda Ricciardi
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Stefano Taricani
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Giulia Sabbatinelli
- Department of Neurosciences, Imaging and Clinical Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Ornella Iuliani
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Cecilia Passeri
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Paolo Sportoletti
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Stella Santarone
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Antonio Pierini
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Giuseppe Calabrese
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy; Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Pescara, Italy
| | - Franca Falzetti
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Tiziana Bonfini
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Patrizia Accorsi
- Department of Oncology Hematology, Pescara Hospital, Pescara, Italy
| | - Loredana Ruggeri
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Massimo Fabrizio Martelli
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Department of Medicine, Division of Hematology and Clinical Immunology, University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy; Department of Oncology Hematology, Pescara Hospital, Pescara, Italy.
| |
Collapse
|
29
|
McBride DA, Kerr MD, Wai SL, Yee YY, Ogbonna DA, Shah NJ. Characterization of regulatory T cell expansion for manufacturing cellular immunotherapies. Biomater Sci 2020; 8:4186-4198. [PMID: 32441280 DOI: 10.1039/d0bm00622j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Regulatory T cells (Tregs) are critical mediators of peripheral immune tolerance. Tregs suppress immune activation against self-antigens and are the focus of cell-based therapies for autoimmune diseases. However, Tregs circulate at a very low frequency in blood, limiting the number of cells that can be isolated by leukapheresis. To effectively expand Tregsex vivo for cell therapy, we report the metabolic modulation of T cells using mono-(6-amino-6-deoxy)-β-cyclodextrin (βCD-NH2) encapsulated rapamycin (Rapa). Encapsulating Rapa in β-cyclodextrin increased its aqueous solubility ∼154-fold and maintained bioactivity for at least 30 days. βCD-NH2-Rapa complexes (CRCs) enriched the fraction of CD4+CD25+FoxP3+ mouse T (mT) cells and human T (hT) cells up to 6-fold and up to 2-fold respectively and suppressed the overall expansion of effector T cells by 5-fold in both species. Combining CRCs and transforming growth factor beta-1 (TGF-β1) synergistically promoted the expansion of CD4+CD25+FoxP3+ T cells. CRCs significantly reduced the fraction of pro-inflammatory interferon-gamma (IFN-γ) expressing CD4+ T cells, suppressing this Th1-associated cytokine while enhancing the fraction of IFN-γ- tumor necrosis factor-alpha (TNF-α) expressing CD4+ T cells. We developed a model using kinetic rate equations to describe the influence of the initial fraction of naïve T cells on the enrichment of Tregsin vitro. The model related the differences in the expansion kinetics of mT and hT cells to their susceptibility for immunophenotypic modulation. CRCs may be an effective and potent means for phenotypic modulation of T cells and the enrichment of Tregsin vitro. Our findings contribute to the development of experimental and analytical techniques for manufacturing Treg based immunotherapies.
Collapse
Affiliation(s)
- David A McBride
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | |
Collapse
|
30
|
El-Ayachi I, Washburn WK, Schenk AD. Recent Progress in Treg Biology and Transplant Therapeutics. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00278-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abstract
Purpose of Review
Regulatory T cell (Treg) biology continues to evolve at a rapid pace. The role of Tregs in solid organ transplantation offers a unique window into Treg ontogeny and function as well as limitless possibilities for clinical application. Here we review recent significant discoveries and key translational work.
Recent Findings
Advances in transplantation deepen understanding of Treg differentiation, expansion, transcription, co-stimulation, and signaling. T cell receptor (TCR) sequencing and single-cell analytics allow unprecedented insight into Treg repertoire diversity and phenotypic heterogeneity. Efforts to replace conventional immunosuppression with Treg adoptive immunotherapy are underway and coalescing around strategies to increase efficiency through development of donor-reactive Tregs.
Summary
Adoptive immunotherapy with Tregs is a leading tolerogenic strategy. Early clinical trials suggest that Treg infusion is safe and reports on efficacy will soon follow.
Collapse
|
31
|
Castella M, Caballero-Baños M, Ortiz-Maldonado V, González-Navarro EA, Suñé G, Antoñana-Vidósola A, Boronat A, Marzal B, Millán L, Martín-Antonio B, Cid J, Lozano M, García E, Tabera J, Trias E, Perpiña U, Canals JM, Baumann T, Benítez-Ribas D, Campo E, Yagüe J, Urbano-Ispizua Á, Rives S, Delgado J, Juan M. Point-Of-Care CAR T-Cell Production (ARI-0001) Using a Closed Semi-automatic Bioreactor: Experience From an Academic Phase I Clinical Trial. Front Immunol 2020; 11:482. [PMID: 32528460 PMCID: PMC7259426 DOI: 10.3389/fimmu.2020.00482] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
Development of semi-automated devices that can reduce the hands-on time and standardize the production of clinical-grade CAR T-cells, such as CliniMACS Prodigy from Miltenyi, is key to facilitate the development of CAR T-cell therapies, especially in academic institutions. However, the feasibility of manufacturing CAR T-cell products from heavily pre-treated patients with this system has not been demonstrated yet. Here we report and characterize the production of 28 CAR T-cell products in the context of a phase I clinical trial for CD19+ B-cell malignancies (NCT03144583). The system includes CD4-CD8 cell selection, lentiviral transduction and T-cell expansion using IL-7/IL-15. Twenty-seven out of 28 CAR T-cell products manufactured met the full list of specifications and were considered valid products. Ex vivo cell expansion lasted an average of 8.5 days and had a mean transduction rate of 30.6 ± 13.44%. All products obtained presented cytotoxic activity against CD19+ cells and were proficient in the secretion of pro-inflammatory cytokines. Expansion kinetics was slower in patient's cells compared to healthy donor's cells. However, product potency was comparable. CAR T-cell subset phenotype was highly variable among patients and largely determined by the initial product. TCM and TEM were the predominant T-cell phenotypes obtained. 38.7% of CAR T-cells obtained presented a TN or TCM phenotype, in average, which are the subsets capable of establishing a long-lasting T-cell memory in patients. An in-depth analysis to identify individual factors contributing to the optimal T-cell phenotype revealed that ex vivo cell expansion leads to reduced numbers of TN, TSCM, and TEFF cells, while TCM cells increase, both due to cell expansion and CAR-expression. Overall, our results show for the first time that clinical-grade production of CAR T-cells for heavily pre-treated patients using CliniMACS Prodigy system is feasible, and that the obtained products meet the current quality standards of the field. Reduced ex vivo expansion may yield CAR T-cell products with increased persistence in vivo.
Collapse
Affiliation(s)
- Maria Castella
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Blood and Tissue Bank (BST), Barcelona, Spain
| | - Miguel Caballero-Baños
- Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain.,Hospital Sant Joan de Déu, Barcelona, Spain
| | - Valentín Ortiz-Maldonado
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Guillermo Suñé
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Asier Antoñana-Vidósola
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anna Boronat
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Berta Marzal
- Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Lucía Millán
- Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Beatriz Martín-Antonio
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joan Cid
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Hemotherapy and Hemostasis, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Miquel Lozano
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Hemotherapy and Hemostasis, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Enric García
- Blood and Tissue Bank (BST), Barcelona, Spain.,Apheresis Unit, Hospital Sant Joan de Déu de Barcelona, Barcelona, Spain
| | - Jaime Tabera
- Blood and Tissue Bank (BST), Barcelona, Spain.,Unit of Advanced Therapies, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Esteve Trias
- Unit of Advanced Therapies, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Unai Perpiña
- Stem Cells and Regenerative Medicine Laboratory, Department of Biomedical Sciences, Production and Validation Center of Advanced Therapies (Creatio), Universitat de Barcelona, Barcelona, Spain
| | - Josep Ma Canals
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Stem Cells and Regenerative Medicine Laboratory, Department of Biomedical Sciences, Production and Validation Center of Advanced Therapies (Creatio), Universitat de Barcelona, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Tycho Baumann
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Daniel Benítez-Ribas
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Elías Campo
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain.,Department of Pathology, Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain.,Centro de Investigación Biomedical en Red de Cancer, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avancats, Barcelona, Spain
| | - Jordi Yagüe
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain.,Department of Biomedicine, School of Medicine, Josep Carreras Leukemia Research Institute, Universitat de Barcelona, Barcelona, Spain.,Immunotherapy Unit Blood and Tissue Bank-Hospital Clínic de Barcelona, Barcelona, Spain
| | - Susana Rives
- Department of Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, Barcelona, Spain.,Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Julio Delgado
- Department of Hematology, Institut Clínic de Malalties Hematològiques i Oncològiques, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomedical en Red de Cancer, Barcelona, Spain
| | - Manel Juan
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Blood and Tissue Bank (BST), Barcelona, Spain.,Department of Immunology, Centro de Diagnóstico Biomédico, Hospital Clínic de Barcelona, Barcelona, Spain.,Hospital Sant Joan de Déu, Barcelona, Spain.,Universitat de Barcelona, Barcelona, Spain.,Immunotherapy Unit Blood and Tissue Bank-Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Rangan S, Schulze HG, Vardaki MZ, Blades MW, Piret JM, Turner RFB. Applications of Raman spectroscopy in the development of cell therapies: state of the art and future perspectives. Analyst 2020; 145:2070-2105. [DOI: 10.1039/c9an01811e] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This comprehensive review article discusses current and future perspectives of Raman spectroscopy-based analyses of cell therapy processes and products.
Collapse
Affiliation(s)
- Shreyas Rangan
- Michael Smith Laboratories
- The University of British Columbia
- Vancouver
- Canada
- School of Biomedical Engineering
| | - H. Georg Schulze
- Michael Smith Laboratories
- The University of British Columbia
- Vancouver
- Canada
| | - Martha Z. Vardaki
- Michael Smith Laboratories
- The University of British Columbia
- Vancouver
- Canada
| | - Michael W. Blades
- Department of Chemistry
- The University of British Columbia
- Vancouver
- Canada
| | - James M. Piret
- Michael Smith Laboratories
- The University of British Columbia
- Vancouver
- Canada
- School of Biomedical Engineering
| | - Robin F. B. Turner
- Michael Smith Laboratories
- The University of British Columbia
- Vancouver
- Canada
- Department of Chemistry
| |
Collapse
|
33
|
Expansion processes for cell-based therapies. Biotechnol Adv 2019; 37:107455. [PMID: 31629791 DOI: 10.1016/j.biotechadv.2019.107455] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/08/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
Living cells are emerging as therapeutic entities for the treatment of patients affected with severe and chronic diseases where no conventional drug can provide a definitive cure. At the same time, the promise of cell-based therapies comes with several biological, regulatory, economic, logistical, safety and engineering challenges that need to be addressed before translating into clinical practice. Among the complex operations required for their manufacturing, cell expansion occupies a significant part of the entire process and largely determines the number, the phenotype and several other critical quality attributes of the final cell therapy products (CTPs). This review aims at characterizing the main culture systems and expansion processes used for CTP production, highlighting the need to implement scalable, cost-efficient technologies together with process optimization strategies to bridge the gap between basic scientific research and commercially available therapies.
Collapse
|
34
|
Flippe L, Bézie S, Anegon I, Guillonneau C. Future prospects for CD8 + regulatory T cells in immune tolerance. Immunol Rev 2019; 292:209-224. [PMID: 31593314 PMCID: PMC7027528 DOI: 10.1111/imr.12812] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD8+ Tregs have been long described and significant progresses have been made about their phenotype, their functional mechanisms, and their suppressive ability compared to conventional CD4+ Tregs. They are now at the dawn of their clinical use. In this review, we will summarize their phenotypic characteristics, their mechanisms of action, the similarities, differences and synergies between CD8+ and CD4+ Tregs, and we will discuss the biology, development and induction of CD8+ Tregs, their manufacturing for clinical use, considering open questions/uncertainties and future technically accessible improvements notably through genetic modifications.
Collapse
Affiliation(s)
- Léa Flippe
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|