101
|
Wagner JC, Ronin E, Ho P, Peng Y, Tang Q. Anti-HLA-A2-CAR Tregs prolong vascularized mouse heterotopic heart allograft survival. Am J Transplant 2022; 22:2237-2245. [PMID: 35434896 PMCID: PMC9427704 DOI: 10.1111/ajt.17063] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/22/2022] [Accepted: 04/12/2022] [Indexed: 01/25/2023]
Abstract
Alloantigen-specific regulatory T cell (Treg) therapy is a promising approach for suppressing alloimmune responses and minimizing immunosuppression after solid organ transplantation. Chimeric antigen receptor (CAR) targeting donor alloantigens can confer donor reactivity to Tregs. However, CAR Treg therapy has not been evaluated in vascularized transplant or multi-MHC mismatched models. Here, we evaluated the ability of CAR Tregs targeting HLA-A2 (A2-CAR) to prolong the survival of heterotopic heart transplants in mice. After verifying the in vitro activation, proliferation, and enhanced suppressive function of A2-CAR Tregs in the presence of A2-antigen, we analyzed the in vivo function of Tregs in C57BL/6 (B6) mice receiving A2-expressing heart allografts. A2-CAR Treg infusion increased the median survival of grafts from B6.HLA-A2 transgenic donors from 23 to 99 days, whereas median survival with polyclonal Treg infusion was 35 days. In a more stringent model of haplo-mismatched hearts from BALB/cxB6.HLA-A2 F1 donors, A2-CAR Tregs slightly increased median graft survival from 11 to 14 days, which was further extended to >100 days when combined with a 9-day course of rapamycin treatment. These findings demonstrate the efficacy of CAR Tregs, alone or in combination with immunosuppressive agents, toward protecting vascularized grafts in fully immunocompetent recipients.
Collapse
Affiliation(s)
- Johanna C. Wagner
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Emilie Ronin
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Patrick Ho
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yani Peng
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
- Gladstone-UCSF Institute of Genomic Immunology, 513 Parnassus Ave, San Francisco, CA 94143, USA
| |
Collapse
|
102
|
Riet T, Chmielewski M. Regulatory CAR-T cells in autoimmune diseases: Progress and current challenges. Front Immunol 2022; 13:934343. [PMID: 36032080 PMCID: PMC9399761 DOI: 10.3389/fimmu.2022.934343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
CAR (Chimeric Antigen Receptor) T-cell therapy has revolutionized the field of oncology in recent years. This innovative shift in cancer treatment also provides the opportunity to improve therapies for many patients suffering from various autoimmune diseases. Recent studies have confirmed the therapeutic suppressive potential of regulatory T cells (Tregs) to modulate immune response in autoimmune diseases. However, the polyclonal character of regulatory T cells and their unknown TCR specificity impaired their therapeutic potency in clinical implementation. Genetical engineering of these immune modulating cells to express antigen-specific receptors and using them therapeutically is a logical step on the way to overcome present limitations of the Treg strategy for the treatment of autoimmune diseases. Encouraging preclinical studies successfully demonstrated immune modulating properties of CAR Tregs in various mouse models. Still, there are many concerns about targeted Treg therapies relating to CAR target selectivity, suppressive functions, phenotype stability and safety aspects. Here, we summarize recent developments in CAR design, Treg biology and future strategies and perspectives in CAR Treg immunotherapy aiming at clinical translation.
Collapse
|
103
|
Preclinical assessment of antigen-specific chimeric antigen receptor regulatory T cells for use in solid organ transplantation. Gene Ther 2022; 30:309-322. [PMID: 35931871 PMCID: PMC10113151 DOI: 10.1038/s41434-022-00358-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022]
Abstract
A primary goal in transplantation medicine is the induction of a tolerogenic environment for prevention of transplant rejection without the need for long-term pharmacological immunosuppression. Generation of alloantigen-specific regulatory T cells (Tregs) by transduction with chimeric antigen receptors (CARs) is a promising strategy to achieve this goal. This publication reports the preclinical characterization of Tregs (TR101) transduced with a human leukocyte antigen (HLA)-A*02 CAR lentiviral vector (TX200) designated to induce immunosuppression of allograft-specific effector T cells in HLA-A*02-negative recipients of HLA-A*02-positive transplants. In vitro results demonstrated specificity, immunosuppressive function, and safety of TX200-TR101. In NOD scid gamma (NSG) mice, TX200-TR101 prevented graft-versus-host disease (GvHD) in a xenogeneic GvHD model and TX200-TR101 Tregs localized to human HLA-A*02-positive skin transplants in a transplant model. TX200-TR101 persisted over the entire duration of a 3-month study in humanized HLA-A*02 NSG mice and remained stable, without switching to a proinflammatory phenotype. Concomitant tacrolimus did not impair TX200-TR101 Treg survival or their ability to inhibit peripheral blood mononuclear cell (PBMC) engraftment. These data demonstrate that TX200-TR101 is specific, stable, efficacious, and safe in preclinical models, and provide the basis for a first-in-human study.
Collapse
|
104
|
Hippen KL, Hefazi M, Larson JH, Blazar BR. Emerging translational strategies and challenges for enhancing regulatory T cell therapy for graft-versus-host disease. Front Immunol 2022; 13:926550. [PMID: 35967386 PMCID: PMC9366169 DOI: 10.3389/fimmu.2022.926550] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 02/03/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for many types of cancer. Genetic disparities between donor and host can result in immune-mediated attack of host tissues, known as graft versus host disease (GVHD), a major cause of morbidity and mortality following HSCT. Regulatory CD4+ T cells (Tregs) are a rare cell type crucial for immune system homeostasis, limiting the activation and differentiation of effector T cells (Teff) that are self-reactive or stimulated by foreign antigen exposure. Adoptive cell therapy (ACT) with Treg has demonstrated, first in murine models and now in patients, that prophylactic Treg infusion can also suppress GVHD. While clinical trials have demonstrated Treg reduce severe GVHD occurrence, several impediments remain, including Treg variability and practical need for individualized Treg production for each patient. Additionally, there are challenges in the use of in vitro expansion techniques and in achieving in vivo Treg persistence in context of both immune suppressive drugs and in lymphoreplete patients being treated for GVHD. This review will focus on 3 main translational approaches taken to improve the efficacy of tTreg ACT in GVHD prophylaxis and development of treatment options, following HSCT: genetic modification, manipulating TCR and cytokine signaling, and Treg production protocols. In vitro expansion for Treg ACT presents a multitude of approaches for gene modification to improve efficacy, including: antigen specificity, tissue targeting, deletion of negative regulators/exhaustion markers, resistance to immunosuppressive drugs common in GVHD treatment. Such expansion is particularly important in patients without significant lymphopenia that can drive Treg expansion, enabling a favorable Treg:Teff ratio in vivo. Several potential therapeutics have also been identified that enhance tTreg stability or persistence/expansion following ACT that target specific pathways, including: DNA/histone methylation status, TCR/co-stimulation signaling, and IL-2/STAT5 signaling. Finally, this review will discuss improvements in Treg production related to tissue source, Treg subsets, therapeutic approaches to increase Treg suppression and stability during tTreg expansion, and potential for storing large numbers of Treg from a single production run to be used as an off-the-shelf infusion product capable of treating multiple recipients.
Collapse
Affiliation(s)
- Keli L. Hippen
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Jemma H. Larson
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| | - Bruce R. Blazar
- University of Minnesota Cancer Center and the Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, Minneapolis, MN, United States
| |
Collapse
|
105
|
Bednar KJ, Lee JH, Ort T. Tregs in Autoimmunity: Insights Into Intrinsic Brake Mechanism Driving Pathogenesis and Immune Homeostasis. Front Immunol 2022; 13:932485. [PMID: 35844555 PMCID: PMC9280893 DOI: 10.3389/fimmu.2022.932485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
CD4+CD25highFoxp3+ regulatory T-cells (Tregs) are functionally characterized for their ability to suppress the activation of multiple immune cell types and are indispensable for maintaining immune homeostasis and tolerance. Disruption of this intrinsic brake system assessed by loss of suppressive capacity, cell numbers, and Foxp3 expression, leads to uncontrolled immune responses and tissue damage. The conversion of Tregs to a pathogenic pro-inflammatory phenotype is widely observed in immune mediated diseases. However, the molecular mechanisms that underpin the control of Treg stability and suppressive capacity are incompletely understood. This review summarizes the concepts of Treg cell stability and Treg cell plasticity highlighting underlying mechanisms including translational and epigenetic regulators that may enable translation to new therapeutic strategies. Our enhanced understanding of molecular mechanism controlling Tregs will have important implications into immune homeostasis and therapeutic potential for the treatment of immune-mediated diseases.
Collapse
|
106
|
Lam AJ, Haque M, Ward-Hartstonge KA, Uday P, Wardell CM, Gillies JK, Speck M, Mojibian M, Klein Geltink RI, Levings MK. PTEN is required for human Treg suppression of costimulation in vitro. Eur J Immunol 2022; 52:1482-1497. [PMID: 35746855 DOI: 10.1002/eji.202249888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/07/2022]
Abstract
Regulatory T cell (Treg) therapy is under clinical investigation for the treatment of transplant rejection, autoimmune disease, and graft-versus-host disease. With the advent of genome editing, attention has turned to reinforcing Treg function for therapeutic benefit. A hallmark of Tregs is dampened activation of PI3K-AKT signalling, of which PTEN is a major negative regulator. Loss-of-function studies of PTEN, however, have not conclusively shown a requirement for PTEN in upholding Treg function and stability. Using CRISPR-based genome editing in human Tregs, we show that PTEN ablation does not cause a global defect in Treg function and stability; rather, it selectively blocks their ability to suppress antigen-presenting cells. PTEN-KO Tregs exhibit elevated glycolytic activity, upregulate FOXP3, maintain a Treg phenotype, and have no discernable defects in lineage stability. Functionally, PTEN is dispensable for human Treg-mediated inhibition of T cell activity in vitro and in vivo, but is required for suppression of costimulatory molecule expression by antigen-presenting cells. These data are the first to define a role for a signalling pathway in controlling a subset of human Treg activity. Moreover, they point to the functional necessity of PTEN-regulated PI3K-AKT activity for optimal human Treg function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Avery J Lam
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Manjurul Haque
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Kirsten A Ward-Hartstonge
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Prakruti Uday
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Jana K Gillies
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Madeleine Speck
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Majid Mojibian
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Ramon I Klein Geltink
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.,Department of Molecular Oncology, BC Cancer Research, Vancouver, BC, V5Z 1L3, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada
| |
Collapse
|
107
|
Gille I, Claas FHJ, Haasnoot GW, Heemskerk MHM, Heidt S. Chimeric Antigen Receptor (CAR) Regulatory T-Cells in Solid Organ Transplantation. Front Immunol 2022; 13:874157. [PMID: 35720402 PMCID: PMC9204347 DOI: 10.3389/fimmu.2022.874157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Solid organ transplantation is the treatment of choice for various end-stage diseases, but requires the continuous need for immunosuppression to prevent allograft rejection. This comes with serious side effects including increased infection rates and development of malignancies. Thus, there is a clinical need to promote transplantation tolerance to prevent organ rejection with minimal or no immunosuppressive treatment. Polyclonal regulatory T-cells (Tregs) are a potential tool to induce transplantation tolerance, but lack specificity and therefore require administration of high doses. Redirecting Tregs towards mismatched donor HLA molecules by modifying these cells with chimeric antigen receptors (CAR) would render Tregs far more effective at preventing allograft rejection. Several studies on HLA-A2 specific CAR Tregs have demonstrated that these cells are highly antigen-specific and show a superior homing capacity to HLA-A2+ allografts compared to polyclonal Tregs. HLA-A2 CAR Tregs have been shown to prolong survival of HLA-A2+ allografts in several pre-clinical humanized mouse models. Although promising, concerns about safety and stability need to be addressed. In this review the current research, obstacles of CAR Treg therapy, and its potential future in solid organ transplantation will be discussed.
Collapse
Affiliation(s)
- Ilse Gille
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Department of Hematology, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Eurotransplant Reference Laboratory, Leiden University Medical Center, Leiden, Netherlands
| | - Geert W Haasnoot
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Eurotransplant Reference Laboratory, Leiden University Medical Center, Leiden, Netherlands
| | | | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands.,Eurotransplant Reference Laboratory, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
108
|
Kath J, Du W, Pruene A, Braun T, Thommandru B, Turk R, Sturgeon ML, Kurgan GL, Amini L, Stein M, Zittel T, Martini S, Ostendorf L, Wilhelm A, Akyüz L, Rehm A, Höpken UE, Pruß A, Künkele A, Jacobi AM, Volk HD, Schmueck-Henneresse M, Stripecke R, Reinke P, Wagner DL. Pharmacological interventions enhance virus-free generation of TRAC-replaced CAR T cells. Mol Ther Methods Clin Dev 2022; 25:311-330. [PMID: 35573047 PMCID: PMC9062427 DOI: 10.1016/j.omtm.2022.03.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptor (CAR) redirected T cells are potent therapeutic options against hematological malignancies. The current dominant manufacturing approach for CAR T cells depends on retroviral transduction. With the advent of gene editing, insertion of a CD19-CAR into the T cell receptor (TCR) alpha constant (TRAC) locus using adeno-associated viruses for gene transfer was demonstrated, and these CD19-CAR T cells showed improved functionality over their retrovirally transduced counterparts. However, clinical-grade production of viruses is complex and associated with extensive costs. Here, we optimized a virus-free genome-editing method for efficient CAR insertion into the TRAC locus of primary human T cells via nuclease-assisted homology-directed repair (HDR) using CRISPR-Cas and double-stranded template DNA (dsDNA). We evaluated DNA-sensor inhibition and HDR enhancement as two pharmacological interventions to improve cell viability and relative CAR knockin rates, respectively. While the toxicity of transfected dsDNA was not fully prevented, the combination of both interventions significantly increased CAR knockin rates and CAR T cell yield. Resulting TRAC-replaced CD19-CAR T cells showed antigen-specific cytotoxicity and cytokine production in vitro and slowed leukemia progression in a xenograft mouse model. Amplicon sequencing did not reveal significant indel formation at potential off-target sites with or without exposure to DNA-repair-modulating small molecules. With TRAC-integrated CAR+ T cell frequencies exceeding 50%, this study opens new perspectives to exploit pharmacological interventions to improve non-viral gene editing in T cells.
Collapse
Affiliation(s)
- Jonas Kath
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Weijie Du
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Alina Pruene
- Regenerative Immune Therapies Applied, Clinics of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Region, Germany
| | - Tobias Braun
- Regenerative Immune Therapies Applied, Clinics of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Region, Germany
| | | | - Rolf Turk
- Integrated DNA Technologies, Inc., Coralville, IA 52241, USA
| | | | - Gavin L. Kurgan
- Integrated DNA Technologies, Inc., Coralville, IA 52241, USA
| | - Leila Amini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Maik Stein
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tatiana Zittel
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Stefania Martini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Lennard Ostendorf
- Department of Nephrology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
| | | | | | - Armin Rehm
- Department of Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine (MDC), 13125 Berlin, Germany
| | - Uta E. Höpken
- Department of Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine (MDC), 13125 Berlin, Germany
| | - Axel Pruß
- Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Annette Künkele
- Department of Pediatric Oncology and Hematology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
| | | | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Michael Schmueck-Henneresse
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Renata Stripecke
- Regenerative Immune Therapies Applied, Clinics of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Hannover-Braunschweig Region, Germany
- Clinic I for Internal Medicine, Cancer Center Cologne Essen, University Hospital Cologne, Cologne, Germany
| | - Petra Reinke
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dimitrios L. Wagner
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany
- Corresponding author Dimitrios Laurin Wagner, MD, PhD, Berlin Center for Advanced Therapies (BeCAT) BIH Center for Regenerative Therapies (BCRT) Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
109
|
Ellis GI, Coker KE, Winn DW, Deng MZ, Shukla D, Bhoj V, Milone MC, Wang W, Liu C, Naji A, Duran-Struuck R, Riley JL. Trafficking and persistence of alloantigen-specific chimeric antigen receptor regulatory T cells in Cynomolgus macaque. Cell Rep Med 2022; 3:100614. [PMID: 35551746 PMCID: PMC9133392 DOI: 10.1016/j.xcrm.2022.100614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/16/2022] [Accepted: 03/29/2022] [Indexed: 01/13/2023]
Abstract
Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models.
Collapse
Affiliation(s)
- Gavin I Ellis
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly E Coker
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Delaine W Winn
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Mosha Z Deng
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Divanshu Shukla
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Vijay Bhoj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
110
|
Qu G, Chen J, Li Y, Yuan Y, Liang R, Li B. Current status and perspectives of regulatory T cell-based therapy. J Genet Genomics 2022; 49:599-611. [DOI: 10.1016/j.jgg.2022.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/08/2022] [Accepted: 05/18/2022] [Indexed: 02/08/2023]
|
111
|
Krovi SH, Kuchroo VK. Activation pathways that drive CD4 + T cells to break tolerance in autoimmune diseases . Immunol Rev 2022; 307:161-190. [PMID: 35142369 PMCID: PMC9255211 DOI: 10.1111/imr.13071] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022]
Abstract
Autoimmune diseases are characterized by dysfunctional immune systems that misrecognize self as non-self and cause tissue destruction. Several cell types have been implicated in triggering and sustaining disease. Due to a strong association of major histocompatibility complex II (MHC-II) proteins with various autoimmune diseases, CD4+ T lymphocytes have been thoroughly investigated for their roles in dictating disease course. CD4+ T cell activation is a coordinated process that requires three distinct signals: Signal 1, which is mediated by antigen recognition on MHC-II molecules; Signal 2, which boosts signal 1 in a costimulatory manner; and Signal 3, which helps to differentiate the activated cells into functionally relevant subsets. These signals are disrupted during autoimmunity and prompt CD4+ T cells to break tolerance. Herein, we review our current understanding of how each of the three signals plays a role in three different autoimmune diseases and highlight the genetic polymorphisms that predispose individuals to autoimmunity. We also discuss the drawbacks of existing therapies and how they can be addressed to achieve lasting tolerance in patients.
Collapse
Affiliation(s)
- Sai Harsha Krovi
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
112
|
Bolivar-Wagers S, Larson JH, Jin S, Blazar BR. Cytolytic CD4 + and CD8 + Regulatory T-Cells and Implications for Developing Immunotherapies to Combat Graft-Versus-Host Disease. Front Immunol 2022; 13:864748. [PMID: 35493508 PMCID: PMC9040077 DOI: 10.3389/fimmu.2022.864748] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T-cells (Treg) are critical for the maintenance of immune homeostasis and tolerance induction. While the immunosuppressive mechanisms of Treg have been extensively investigated for decades, the mechanisms responsible for Treg cytotoxicity and their therapeutic potential in regulating immune responses have been incompletely explored and exploited. Conventional cytotoxic T effector cells (Teffs) are known to be important for adaptive immune responses, particularly in the settings of viral infections and cancer. CD4+ and CD8+ Treg subsets may also share similar cytotoxic properties with conventional Teffs. Cytotoxic effector Treg (cyTreg) are a heterogeneous population in the periphery that retain the capacity to suppress T-cell proliferation and activation, induce cellular apoptosis, and migrate to tissues to ensure immune homeostasis. The latter can occur through several cytolytic mechanisms, including the Granzyme/Perforin and Fas/FasL signaling pathways. This review focuses on the current knowledge and recent advances in our understanding of cyTreg and their potential application in the treatment of human disease, particularly Graft-versus-Host Disease (GVHD).
Collapse
Affiliation(s)
| | | | | | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
113
|
Lapp MM, Lin G, Komin A, Andrews L, Knudson M, Mossman L, Raimondi G, Arciero JC. Modeling the Potential of Treg-Based Therapies for Transplant Rejection: Effect of Dose, Timing, and Accumulation Site. Transpl Int 2022; 35:10297. [PMID: 35479106 PMCID: PMC9035492 DOI: 10.3389/ti.2022.10297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/17/2022] [Indexed: 02/04/2023]
Abstract
Introduction: The adoptive transfer of regulatory T cells (Tregs) has emerged as a method to promote graft tolerance. Clinical trials have demonstrated the safety of adoptive transfer and are now assessing their therapeutic efficacy. Strategies that generate large numbers of antigen specific Tregs are even more efficacious. However, the combinations of factors that influence the outcome of adoptive transfer are too numerous to be tested experimentally. Here, mathematical modeling is used to predict the most impactful treatment scenarios. Methods: We adapted our mathematical model of murine heart transplant rejection to simulate Treg adoptive transfer and to correlate therapeutic efficacy with Treg dose and timing, frequency of administration, and distribution of injected cells. Results: The model predicts that Tregs directly accumulating to the graft are more protective than Tregs localizing to draining lymph nodes. Inhibiting antigen-presenting cell maturation and effector functions at the graft site was more effective at modulating rejection than inhibition of T cell activation in lymphoid tissues. These complex dynamics define non-intuitive relationships between graft survival and timing and frequency of adoptive transfer. Conclusion: This work provides the framework for better understanding the impact of Treg adoptive transfer and will guide experimental design to improve interventions.
Collapse
Affiliation(s)
- Maya M. Lapp
- Department of Mathematics, The College of Wooster, Wooster, OH, United States
| | - Guang Lin
- Department of Mathematics, Purdue University, West Lafayette, IN, United States
| | - Alexander Komin
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Leah Andrews
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Mei Knudson
- Department of Mathematics, Carleton College, Northfield, MN, United States
| | - Lauren Mossman
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| | - Julia C. Arciero
- Department of Mathematical Sciences, Indiana University-Purdue University of Indianapolis, Indianapolis, IN, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| |
Collapse
|
114
|
Hossian AKMN, Hackett CS, Brentjens RJ, Rafiq S. Multipurposing CARs: Same engine, different vehicles. Mol Ther 2022; 30:1381-1395. [PMID: 35151842 PMCID: PMC9077369 DOI: 10.1016/j.ymthe.2022.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αβ T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Christopher S Hackett
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA.
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
115
|
Mamo T, Hippen KL, MacMillan ML, Brunstein CG, Miller JS, Wagner JE, Blazar BR, McKenna DH. Regulatory T cells: A review of manufacturing and clinical utility. Transfusion 2022; 62:904-915. [PMID: 35015309 PMCID: PMC8986575 DOI: 10.1111/trf.16797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Tewodros Mamo
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Keli L. Hippen
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Margaret L. MacMillan
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Claudio G. Brunstein
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Jeffrey S. Miller
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | - John E. Wagner
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Bruce R. Blazar
- Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - David H. McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
116
|
Schreeb K, Culme-Seymour E, Ridha E, Dumont C, Atkinson G, Hsu B, Reinke P. Study Design: HLA-A*02-Chimeric Antigen Receptor Regulatory T Cells in Renal Transplantation. Kidney Int Rep 2022; 7:1258-1267. [PMID: 35694562 PMCID: PMC9174048 DOI: 10.1016/j.ekir.2022.03.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/04/2022] [Accepted: 03/28/2022] [Indexed: 11/21/2022] Open
Abstract
Introduction Cell therapy with regulatory T cells (Tregs) in solid organ transplantation is a promising approach for the prevention of graft rejection and induction of immunologic tolerance. Previous clinical studies have demonstrated the safety of Tregs in renal transplant recipients. Antigen-specific Tregs, such as chimeric antigen receptor (CAR)-Tregs, are expected to be more efficacious than polyclonal Tregs in homing to the target antigen. We have developed an autologous cell therapy (TX200-TR101) where a human leukocyte antigen (HLA) class I molecule A∗02 (HLA-A∗02)-CAR is introduced into autologous naive Tregs from a patient with HLA-A∗02-negative end-stage renal disease (ESRD) awaiting an HLA-A∗02-positive donor kidney. Methods This article describes the design of the STEADFAST study, a first-in-human, phase I/IIa, multicenter, open-label, single-ascending dose, dose-ranging study to assess TX200-TR101 in living-donor renal transplant recipients. Up to 15 transplant recipients will receive TX200-TR101 and will be followed up for a total of 84 weeks post-transplant, alongside a control cohort of up to 6 transplant recipients. All transplant recipients will receive a standard of care immunosuppressive regimen, with the intent of intensified tapering of the regimen in the TX200-TR101 cohort. Results The primary end point is the incidence and severity of treatment-emergent adverse events (AEs) within 28 days post–TX200-TR101 infusion. Other end points include additional safety parameters, clinical and renal outcome parameters, and the evaluation of biomarkers. Conclusion The STEADFAST study represents the next frontier in adoptive cell therapies. TX200-TR101 holds great potential to prevent immune-mediated graft rejection and induce immunologic tolerance after HLA-A∗02-mismatched renal transplantation.
Collapse
|
117
|
Cremoni M, Massa F, Sicard A. Overcoming barriers to widespread use of CAR-Treg therapy in organ transplant recipients. HLA 2022; 99:565-572. [PMID: 35233971 DOI: 10.1111/tan.14591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 02/27/2022] [Indexed: 11/30/2022]
Abstract
Preventing allograft rejection has been the main challenge of transplantation medicine since the discovery of immune responses against foreign HLA molecules in the mid-20th century. Prevention of rejection currently relies on immunosuppressive drugs, which lack antigen specificity and therefore increase the risk for infections and cancers. Adoptive cell therapy with donor-reactive regulatory T cells (Tregs) has progressively emerged as a promising approach to reduce the need for pan-immunosuppressive drugs and minimize morbidity and mortality in solid-organ transplant recipients. Chimeric antigen receptor (CAR) technology has recently been used successfully to generate Tregs specific for donor HLA molecules and overcome the limitations of Tregs enrichment protocols based on repetitive stimulations with alloantigens. While this novel approach opens new possibilities to make Tregs therapy more feasible, it also creates additional challenges. It is essential to determine which source of therapeutic Tregs, CAR constructs, target alloantigens, safety strategies, patients and immunosuppressive regimens are optimal for the success of CAR Treg therapy. Here, we discuss unmet needs and strategies to bring donor-specific CAR Treg therapy to the clinic and make it as accessible as possible.
Collapse
Affiliation(s)
- Marion Cremoni
- Department of Nephrology, Dialysis, Transplantation, Nice University Hospital, Nice, France.,Clinical Research Unit, University Côte d'Azur (UR2CA), Nice, France
| | - Filippo Massa
- Department of Nephrology, Dialysis, Transplantation, Nice University Hospital, Nice, France.,Laboratory of Molecular Physio Medicine (LP2M), University Côte d'Azur, Nice, France
| | - Antoine Sicard
- Department of Nephrology, Dialysis, Transplantation, Nice University Hospital, Nice, France.,Laboratory of Molecular Physio Medicine (LP2M), University Côte d'Azur, Nice, France
| |
Collapse
|
118
|
Khosravi-Maharlooei M, Madley R, Borsotti C, Ferreira LMR, Sharp RC, Brehm MA, Greiner DL, Parent AV, Anderson MS, Sykes M, Creusot RJ. Modeling human T1D-associated autoimmune processes. Mol Metab 2022; 56:101417. [PMID: 34902607 PMCID: PMC8739876 DOI: 10.1016/j.molmet.2021.101417] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by impaired immune tolerance to β-cell antigens and progressive destruction of insulin-producing β-cells. Animal models have provided valuable insights for understanding the etiology and pathogenesis of this disease, but they fall short of reflecting the extensive heterogeneity of the disease in humans, which is contributed by various combinations of risk gene alleles and unique environmental factors. Collectively, these factors have been used to define subgroups of patients, termed endotypes, with distinct predominating disease characteristics. SCOPE OF REVIEW Here, we review the gaps filled by these models in understanding the intricate involvement and regulation of the immune system in human T1D pathogenesis. We describe the various models developed so far and the scientific questions that have been addressed using them. Finally, we discuss the limitations of these models, primarily ascribed to hosting a human immune system (HIS) in a xenogeneic recipient, and what remains to be done to improve their physiological relevance. MAJOR CONCLUSIONS To understand the role of genetic and environmental factors or evaluate immune-modifying therapies in humans, it is critical to develop and apply models in which human cells can be manipulated and their functions studied under conditions that recapitulate as closely as possible the physiological conditions of the human body. While microphysiological systems and living tissue slices provide some of these conditions, HIS mice enable more extensive analyses using in vivo systems.
Collapse
Affiliation(s)
- Mohsen Khosravi-Maharlooei
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Rachel Madley
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Chiara Borsotti
- Department of Health Sciences, Histology laboratory, Università del Piemonte Orientale, Novara, Italy
| | - Leonardo M R Ferreira
- Departments of Microbiology & Immunology, and Regenerative Medicine & Cell Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Robert C Sharp
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michael A Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dale L Greiner
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Audrey V Parent
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Mark S Anderson
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Remi J Creusot
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
119
|
Aghajanian H, Rurik JG, Epstein JA. CAR-based therapies: opportunities for immuno-medicine beyond cancer. Nat Metab 2022; 4:163-169. [PMID: 35228742 PMCID: PMC9947862 DOI: 10.1038/s42255-022-00537-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/20/2022] [Indexed: 02/01/2023]
Abstract
One of the most exciting new therapies for cancer involves the use of autologous T cells that are engineered to recognize and destroy cancerous cells. Patients with previously untreatable B cell leukaemias and lymphomas have been cured, and efforts are underway to extend this success to other tumours. Here, we discuss recent studies and emerging research aimed to extend this approach beyond oncology in areas such as cardiometabolic disorders, autoimmunity, fibrosis and senescence. We also summarize new technologies that may help to reduce the cost and increase access to related forms of immunotherapy.
Collapse
Affiliation(s)
- Haig Aghajanian
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Joel G. Rurik
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Jonathan A. Epstein
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
120
|
Radic M, Neeli I, Marion T. Prospects for CAR T cell immunotherapy in autoimmune diseases: clues from Lupus. Expert Opin Biol Ther 2022; 22:499-507. [PMID: 35089116 DOI: 10.1080/14712598.2022.2026921] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Medicine stands at the threshold of a new era heralded by the vast potential of cell engineering. Like advances made possible by genetic engineering, current prospects for purposeful control of cell functions through cell engineering may bring breakthroughs in the treatment of previously intractable diseases. AREAS COVERED Engineering of cytotoxic T cells for expression of chimeric antigen receptors (CARs) instructs them to attack and destroy malignant cells and thus provides an exciting new approach in oncology. A decade of practical experience and first-in-human trials encourage the search for new and broader uses of CAR technology, including in autoimmune diseases. EXPERT OPINION Systemic lupus erythematosus is an example of a broader category of autoimmune diseases, for which cell engineering will provide a powerful new therapeutic approach. This article describes different types of CAR T cell strategies that will provide new treatment options for patients with autoimmune diseases and replace conventional therapies.
Collapse
Affiliation(s)
- Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN (USA)
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN (USA)
| | - Tony Marion
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN (USA)
| |
Collapse
|
121
|
Arjomandnejad M, Kopec AL, Keeler AM. CAR-T Regulatory (CAR-Treg) Cells: Engineering and Applications. Biomedicines 2022; 10:287. [PMID: 35203496 PMCID: PMC8869296 DOI: 10.3390/biomedicines10020287] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Regulatory T cells are critical for maintaining immune tolerance. Recent studies have confirmed their therapeutic suppressive potential to modulate immune responses in organ transplant and autoimmune diseases. However, the unknown and nonspecific antigen recognition of polyclonal Tregs has impaired their therapeutic potency in initial clinical findings. To address this limitation, antigen specificity can be conferred to Tregs by engineering the expression of transgenic T-cell receptor (TCR) or chimeric antigen receptor (CAR). In contrast to TCR Tregs, CAR Tregs are major histocompatibility complex (MHC) independent and less dependent on interleukin-2 (IL-2). Furthermore, CAR Tregs maintain Treg phenotype and function, home to the target tissue and show enhanced suppressive efficacy compared to polyclonal Tregs. Additional development of engineered CAR Tregs is needed to increase Tregs' suppressive function and stability, prevent CAR Treg exhaustion, and assess their safety profile. Further understanding of Tregs therapeutic potential will be necessary before moving to broader clinical applications. Here, we summarize recent studies utilizing CAR Tregs in modulating immune responses in autoimmune diseases, transplantation, and gene therapy and future clinical applications.
Collapse
Affiliation(s)
- Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (M.A.); (A.L.K.)
| | - Acadia L. Kopec
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (M.A.); (A.L.K.)
| | - Allison M. Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA; (M.A.); (A.L.K.)
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
122
|
Boardman DA, Levings MK. Emerging strategies for treating autoimmune disorders with genetically modified Treg cells. J Allergy Clin Immunol 2022; 149:1-11. [PMID: 34998473 DOI: 10.1016/j.jaci.2021.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022]
Abstract
Gene editing of living cells is a cornerstone of present-day medical research that has enabled scientists to address fundamental biologic questions and identify novel strategies to treat diseases. The ability to manipulate adoptive cell therapy products has revolutionized cancer immunotherapy and promises similar results for the treatment of autoimmune diseases, inflammatory disorders, and transplant rejection. Clinical trials have recently deemed polyclonal regulatory T (Treg) cell therapy to be a safe therapeutic option, but questions remain regarding the efficacy of this approach. In this review, we discuss how gene editing technologies are being applied to transform the future of Treg cell therapy, focusing on the preclinical strategies that are currently being investigated to enhance the efficacy, function, and survival of human Treg cells. We explore approaches that may be used to generate immunoregulatory cells ex vivo, detail emerging strategies that are being used to modify these cells (such as using chimeric antigen receptors to confer antigen specificity), and outline concepts that have been explored to repurpose conventional T cells to target and destroy autoreactive and alloreactive lymphocytes. We also describe the key hurdles that currently hinder the clinical adoption of Treg cell therapy and propose potential future avenues of research for this field.
Collapse
Affiliation(s)
- Dominic A Boardman
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
123
|
Lee SK, Han J, Piao H, Shin N, Jang JY, Yan JJ, Kim H, Chung J, Yang J. Anti-C4d chimeric antigen receptor regulatory T cells suppressed allograft rejection in ABO-incompatible heart transplantation. Genes Dis 2022; 9:1-4. [PMID: 35005103 PMCID: PMC8720693 DOI: 10.1016/j.gendis.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/27/2021] [Accepted: 07/09/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Sun-Kyung Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea.,Department of Medicine, Graduate School, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jerome Han
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Honglin Piao
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea.,Department of Medicine, Graduate School, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Nara Shin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Joon Young Jang
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji-Jing Yan
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Hyori Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jaeseok Yang
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
124
|
Koehler N, Buhler L, Egger B, Gonelle-Gispert C. Multipotent Mesenchymal Stromal Cells Interact and Support Islet of Langerhans Viability and Function. Front Endocrinol (Lausanne) 2022; 13:822191. [PMID: 35222280 PMCID: PMC8864309 DOI: 10.3389/fendo.2022.822191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a widespread disease, affecting approximately 41.5 million people worldwide. It is generally treated with exogenous insulin, maintaining physiological blood glucose levels but also leading to long-term therapeutic complications. Pancreatic islet cell transplantation offers a potential alternative treatment to insulin injections. Shortage of human organ donors has raised the interest for porcine islet xenotransplantation. Neonatal porcine islets are highly available, can proliferate and mature in vitro as well as after transplantation in vivo. Despite promising preclinical results, delayed insulin secretion caused by immaturity and immunogenicity of the neonatal porcine islets remains a challenge for their clinical application. Multipotent mesenchymal stromal cells (MSCs) are known to have pro-angiogenic, anti-inflammatory and immunomodulatory effects. The current state of research emphasizes the great potential of co-culture and co-transplantation of islet cells with MSCs. Studies have shown enhanced islet proliferation and maturation, insulin secretion and graft survival, resulting in an improved graft outcome. This review summarizes the immunomodulatory and anti-inflammatory properties of MSC in the context of islet transplantation.
Collapse
Affiliation(s)
- Naomi Koehler
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Leo Buhler
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Bernhard Egger
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Carmen Gonelle-Gispert,
| |
Collapse
|
125
|
Rickert CG, Markmann JF. Transplantation in the Age of Precision Medicine: The Emerging Field of Treg Therapy. Semin Nephrol 2022; 42:76-85. [DOI: 10.1016/j.semnephrol.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
126
|
Arjomandnejad M, Sylvia K, Blackwood M, Nixon T, Tang Q, Muhuri M, Gruntman AM, Gao G, Flotte TR, Keeler AM. Modulating immune responses to AAV by expanded polyclonal T-regs and capsid specific chimeric antigen receptor T-regulatory cells. Mol Ther Methods Clin Dev 2021; 23:490-506. [PMID: 34853797 PMCID: PMC8605179 DOI: 10.1016/j.omtm.2021.10.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022]
Abstract
Immune responses to adeno-associated virus (AAV) capsids limit the therapeutic potential of AAV gene therapy. Herein, we model clinical immune responses by generating AAV capsid-specific chimeric antigen receptor (AAV-CAR) T cells. We then modulate immune responses to AAV capsid with AAV-CAR regulatory T cells (Tregs). AAV-CAR Tregs in vitro display phenotypical Treg surface marker expression, and functional suppression of effector T cell proliferation and cytotoxicity. In mouse models, AAV-CAR Tregs mediated continued transgene expression from an immunogenic capsid, despite antibody responses, produced immunosuppressive cytokines, and decreased tissue inflammation. AAV-CAR Tregs are also able to bystander suppress immune responses to immunogenic transgenes similarly mediating continued transgene expression, producing immunosuppressive cytokines, and reducing tissue infiltration. Taken together, AAV-CAR T cells and AAV-CAR Tregs are directed and powerful immunosuppressive tools to model and modulate immune responses to AAV capsids and transgenes in the local environment.
Collapse
Affiliation(s)
- Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Katelyn Sylvia
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Meghan Blackwood
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Thomas Nixon
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Qiushi Tang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Alisha M Gruntman
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Clinical Sciences, Cummings School of Veterinary Medicine at Tufts University, Grafton, MA 01536, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Allison M Keeler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA.,NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
127
|
CAR Treg: A new approach in the treatment of autoimmune diseases. Int Immunopharmacol 2021; 102:108409. [PMID: 34863655 DOI: 10.1016/j.intimp.2021.108409] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 12/22/2022]
Abstract
Regulatory T cells (Tregs) have the role of regulating self-tolerance, and suppressing immune responses. Defects in Treg function and number can lead to in loss of tolerance or autoimmune disease. To treat or control autoimmune diseases, one of the options is to develop immune tolerance for Tregs cell therapy, which includes promotion and activation. Recently, cell-based treatment as a promising approach to increase cells function and number has been developed. Cell therapy by chimeric T antigen receptor (CAR-T) cells has shown significant efficacy in the treatment of leukemia, which has led researchers to use CAR-T cells in other diseases like autoimmune diseases. Here, we describe the existing treatments for autoimmune diseases and the available treatments based on Treg, their benefits and restrictions for implementation in clinical trials. We also discussed potential solutions to overcome these limitations. It seems novel designs of CARs to be new hope for autoimmune diseases and expected to be a potential cure option in a wide array of disease in the future. Therefore, it is very important to address this issue and increase information about it.
Collapse
|
128
|
Advances in engineering and synthetic biology toward improved therapeutic immune cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
129
|
The effect of the cystic fibrosis care center on outcomes after lung transplantation for cystic fibrosis. J Heart Lung Transplant 2021; 41:300-307. [PMID: 34930671 DOI: 10.1016/j.healun.2021.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/02/2021] [Accepted: 11/14/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The purpose of this study was to evaluate outcomes in people with cystic fibrosis (CF) who underwent lung transplant (LT) at a transplant center with an accredited Cystic Fibrosis Care Center (CFCC) in the United States. METHODS We reviewed the Scientific Registry of Transplant Recipients for all adult patients with CF who received a first-time LT from 2005 to 2018. The primary outcome was graft failure. Unadjusted Kaplan-Meier analysis and adjusted multilevel Cox proportional hazards models were used to evaluate outcomes in CF patients undergoing lung transplantation at a CFCC. RESULTS 2,573 patients with CF underwent a first time LT during the study period. Of the 68 lung transplantation centers, 50 were CFCCs (73.5%). After adjustment for potential confounders, patients who underwent lung transplantation at a hospital with an accredited CFCC had a 33% reduction in risk of death or re-transplantation compared to those transplanted at a hospital without an accredited CFCC (HR: 0.67, 95% CI: 0.56-0.82, p < 0.001). CONCLUSIONS People with CF who undergo LT at a transplant center with a CFCC have improved graft survival and decreased need for re-transplantation compared to those who undergo LT at a non-CFCC, independent of volume.
Collapse
|
130
|
Lamarthée B, Marchal A, Charbonnier S, Blein T, Leon J, Martin E, Rabaux L, Vogt K, Titeux M, Delville M, Vinçon H, Six E, Pallet N, Michonneau D, Anglicheau D, Legendre C, Taupin JL, Nemazanyy I, Sawitzki B, Latour S, Cavazzana M, André I, Zuber J. Transient mTOR inhibition rescues 4-1BB CAR-Tregs from tonic signal-induced dysfunction. Nat Commun 2021; 12:6446. [PMID: 34750385 PMCID: PMC8575891 DOI: 10.1038/s41467-021-26844-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 10/25/2021] [Indexed: 12/30/2022] Open
Abstract
The use of chimeric antigen receptor (CAR)-engineered regulatory T cells (Tregs) has emerged as a promising strategy to promote immune tolerance. However, in conventional T cells (Tconvs), CAR expression is often associated with tonic signaling, which can induce CAR-T cell dysfunction. The extent and effects of CAR tonic signaling vary greatly according to the expression intensity and intrinsic properties of the CAR. Here, we show that the 4-1BB CSD-associated tonic signal yields a more dramatic effect in CAR-Tregs than in CAR-Tconvs with respect to activation and proliferation. Compared to CD28 CAR-Tregs, 4-1BB CAR-Tregs exhibit decreased lineage stability and reduced in vivo suppressive capacities. Transient exposure of 4-1BB CAR-Tregs to a Treg stabilizing cocktail, including an mTOR inhibitor and vitamin C, during ex vivo expansion sharply improves their in vivo function and expansion after adoptive transfer. This study demonstrates that the negative effects of 4-1BB tonic signaling in Tregs can be mitigated by transient mTOR inhibition.
Collapse
MESH Headings
- Animals
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- Graft vs Host Disease/immunology
- Graft vs Host Disease/therapy
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Humans
- Immunosuppressive Agents/pharmacology
- Immunotherapy, Adoptive/methods
- Jurkat Cells
- Male
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Sirolimus/pharmacology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/immunology
- TOR Serine-Threonine Kinases/metabolism
- Transplantation, Heterologous
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Mice
Collapse
Affiliation(s)
- Baptiste Lamarthée
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Armance Marchal
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Soëli Charbonnier
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Tifanie Blein
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Juliette Leon
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Emmanuel Martin
- Lymphocyte activation and susceptibility to EBV, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Lucas Rabaux
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Katrin Vogt
- Department of Immunology, Charité University Hospital, Berlin, Germany
| | - Matthias Titeux
- Maladie génétique cutanée, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Marianne Delville
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
- Université de Paris, Paris, France
- Service de Biothérapie et Thérapie Génique Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Hélène Vinçon
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Emmanuelle Six
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Nicolas Pallet
- Université de Paris, INSERM U1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| | | | - Dany Anglicheau
- Université de Paris, Paris, France
- Service de Transplantation rénale adulte, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Christophe Legendre
- Université de Paris, Paris, France
- Service de Transplantation rénale adulte, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Jean-Luc Taupin
- Université de Paris, Paris, France
- Laboratoire d'immunologie et histocompatibilité, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Ivan Nemazanyy
- Plateforme de Métabolique, Structure Fédérative de Recherche, Necker, INSERM US24/CNRS UMS, 3633, Paris, France
| | - Birgit Sawitzki
- Department of Immunology, Charité University Hospital, Berlin, Germany
| | - Sylvain Latour
- Lymphocyte activation and susceptibility to EBV, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Marina Cavazzana
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
- Université de Paris, Paris, France
- Service de Biothérapie et Thérapie Génique Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Isabelle André
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France
| | - Julien Zuber
- Laboratoire de lymphohématopoïèse humaine, INSERM UMR 1163, IHU IMAGINE, Paris, France.
- Université de Paris, Paris, France.
- Service de Transplantation rénale adulte, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France.
| |
Collapse
|
131
|
McNerney MP, Doiron KE, Ng TL, Chang TZ, Silver PA. Theranostic cells: emerging clinical applications of synthetic biology. Nat Rev Genet 2021; 22:730-746. [PMID: 34234299 PMCID: PMC8261392 DOI: 10.1038/s41576-021-00383-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Synthetic biology seeks to redesign biological systems to perform novel functions in a predictable manner. Recent advances in bacterial and mammalian cell engineering include the development of cells that function in biological samples or within the body as minimally invasive diagnostics or theranostics for the real-time regulation of complex diseased states. Ex vivo and in vivo cell-based biosensors and therapeutics have been developed to target a wide range of diseases including cancer, microbiome dysbiosis and autoimmune and metabolic diseases. While probiotic therapies have advanced to clinical trials, chimeric antigen receptor (CAR) T cell therapies have received regulatory approval, exemplifying the clinical potential of cellular therapies. This Review discusses preclinical and clinical applications of bacterial and mammalian sensing and drug delivery platforms as well as the underlying biological designs that could enable new classes of cell diagnostics and therapeutics. Additionally, we describe challenges that must be overcome for more rapid and safer clinical use of engineered systems.
Collapse
Affiliation(s)
- Monica P McNerney
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Kailyn E Doiron
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tai L Ng
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Timothy Z Chang
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Pamela A Silver
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW Current immunosuppressive regimens used in kidney transplantation are sometimes ineffective and carry significant risks of morbidity and mortality. Cellular therapies are a promising alternative to prolong graft survival while minimizing treatment toxicity. We review the recently published breakthrough studies using cell therapies in kidney transplantation. RECENT FINDINGS The reviewed phase I and II trials showed that cell therapies are feasible and safe in kidney transplantation, sometimes associated with less infectious complications than traditional regimens. Regulatory T cells and macrophages were added to the induction regimen, allowing for lower immunosuppressive drug doses without higher rejection risk. Regulatory T cells are also a treatment for subclinical rejection on the 6 months biopsy. Other strategies, like bone marrow-derived mesenchymal cells, genetically modified regulatory T cells, and chimerism-based tolerance are also really promising. In addition, to improve graft tolerance, cell therapy could be used to prevent or treat viral infection after transplantation. SUMMARY Emerging data underline that cell therapy is a feasible and safe treatment in kidney transplantation. Although the evidence points to a benefit for transplant recipients, studies with standardized protocols, representative control groups, and longer follow-up are needed to answer the question definitively and guide future research.
Collapse
Affiliation(s)
- Simon Leclerc
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Caroline Lamarche
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
133
|
Muller YD, Ferreira LMR, Ronin E, Ho P, Nguyen V, Faleo G, Zhou Y, Lee K, Leung KK, Skartsis N, Kaul AM, Mulder A, Claas FHJ, Wells JA, Bluestone JA, Tang Q. Precision Engineering of an Anti-HLA-A2 Chimeric Antigen Receptor in Regulatory T Cells for Transplant Immune Tolerance. Front Immunol 2021; 12:686439. [PMID: 34616392 PMCID: PMC8488356 DOI: 10.3389/fimmu.2021.686439] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Infusion of regulatory T cells (Tregs) engineered with a chimeric antigen receptor (CAR) targeting donor-derived human leukocyte antigen (HLA) is a promising strategy to promote transplant tolerance. Here, we describe an anti-HLA-A2 CAR (A2-CAR) generated by grafting the complementarity-determining regions (CDRs) of a human monoclonal anti-HLA-A2 antibody into the framework regions of the Herceptin 4D5 single-chain variable fragment and fusing it with a CD28-ζ signaling domain. The CDR-grafted A2-CAR maintained the specificity of the original antibody. We then generated HLA-A2 mono-specific human CAR Tregs either by deleting the endogenous T-cell receptor (TCR) via CRISPR/Cas9 and introducing the A2-CAR using lentiviral transduction or by directly integrating the CAR construct into the TCR alpha constant locus using homology-directed repair. These A2-CAR+TCRdeficient human Tregs maintained both Treg phenotype and function in vitro. Moreover, they selectively accumulated in HLA-A2-expressing islets transplanted from either HLA-A2 transgenic mice or deceased human donors. A2-CAR+TCRdeficient Tregs did not impair the function of these HLA-A2+ islets, whereas similarly engineered A2-CAR+TCRdeficientCD4+ conventional T cells rejected the islets in less than 2 weeks. A2-CAR+TCRdeficient Tregs delayed graft-versus-host disease only in the presence of HLA-A2, expressed either by co-transferred peripheral blood mononuclear cells or by the recipient mice. Altogether, we demonstrate that genome-engineered mono-antigen-specific A2-CAR Tregs localize to HLA-A2-expressing grafts and exhibit antigen-dependent in vivo suppression, independent of TCR expression. These approaches may be applied towards developing precision Treg cell therapies for transplant tolerance.
Collapse
Affiliation(s)
- Yannick D Muller
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - Leonardo M R Ferreira
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States.,Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, United States
| | - Emilie Ronin
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - Patrick Ho
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States.,Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, United States
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - Gaetano Faleo
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| | - Yu Zhou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, Zuckerberg San Francisco General Hospital and Trauma Center, San Francisco, CA, United States
| | - Karim Lee
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, United States
| | - Nikolaos Skartsis
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Anupurna M Kaul
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Arend Mulder
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States.,Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
134
|
Cox JR, Blazeck J. Protein engineering: a driving force toward synthetic immunology. Trends Biotechnol 2021; 40:509-521. [PMID: 34627648 DOI: 10.1016/j.tibtech.2021.09.005] [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: 07/09/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/29/2022]
Abstract
The full application of the diverse toolkit of protein engineering has made it easier to control the immune system. In particular, synthetic cytokine variants and engineered immune receptor platforms have shown promise for the treatment of various indications with dysregulated immune function, particularly cancer. Here, we review recent advances in the control of immune cell signaling and therapeutic potency that have employed protein engineering strategies. We further discuss how safety concerns are driving the design of immunotherapeutics toward 'user-defined' control or requiring multiple distinct inputs before a functional response, highlighting emergent control strategies employed for chimeric antigen receptor (CAR) engineering.
Collapse
Affiliation(s)
- John R Cox
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst St. NW, Atlanta, GA 30332, USA
| | - John Blazeck
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst St. NW, Atlanta, GA 30332, USA.
| |
Collapse
|
135
|
Jiang H, Fu D, Bidgoli A, Paczesny S. T Cell Subsets in Graft Versus Host Disease and Graft Versus Tumor. Front Immunol 2021; 12:761448. [PMID: 34675938 PMCID: PMC8525316 DOI: 10.3389/fimmu.2021.761448] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an essential therapeutic modality for patients with hematological malignancies and other blood disorders. Unfortunately, acute graft-versus-host disease (aGVHD) remains a major source of morbidity and mortality following allo-HCT, which limits its use in a broader spectrum of patients. Chronic graft-versus-host disease (cGVHD) also remains the most common long-term complication of allo-HCT, occurring in reportedly 30-70% of patients surviving more than 100 days. Chronic GVHD is also the leading cause of non-relapse mortality (NRM) occurring more than 2 years after HCT for malignant disease. Graft versus tumor (GVT) is a major component of the overall beneficial effects of allogeneic HCT in the treatment of hematological malignancies. Better understanding of GVHD pathogenesis is important to identify new therapeutic targets for GVHD prevention and therapy. Emerging data suggest opposing roles for different T cell subsets, e.g., IFN-γ producing CD4+ and CD8+ T cells (Th1 and Tc1), IL-4 producing T cells (Th2 and Tc2), IL-17 producing T cells (Th17 and Tc17), IL-9 producing T cells (Th9 and Tc9), IL-22 producing T cells (Th22), T follicular helper cells (Tfh), regulatory T-cells (Treg) and tissue resident memory T cells (Trm) in GVHD and GVT etiology. In this review, we first summarize the general description of the cytokine signals that promote the differentiation of T cell subsets and the roles of these T cell subsets in the pathogenesis of GVHD. Next, we extensively explore preclinical findings of T cell subsets in both GVHD/GVT animal models and humans. Finally, we address recent findings about the roles of T-cell subsets in clinical GVHD and current strategies to modulate T-cell differentiation for treating and preventing GVHD in patients. Further exploring and outlining the immune biology of T-cell differentiation in GVHD that will provide more therapeutic options for maintaining success of allo-HCT.
Collapse
Affiliation(s)
| | | | | | - Sophie Paczesny
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
136
|
Mohseni YR, Saleem A, Tung SL, Dudreuilh C, Lang C, Peng Q, Volpe A, Adigbli G, Cross A, Hester J, Farzaneh F, Scotta C, Lechler RI, Issa F, Fruhwirth GO, Lombardi G. Chimeric antigen receptor-modified human regulatory T cells that constitutively express IL-10 maintain their phenotype and are potently suppressive. Eur J Immunol 2021; 51:2522-2530. [PMID: 34320225 PMCID: PMC8581768 DOI: 10.1002/eji.202048934] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 04/30/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022]
Abstract
Clinical trials of Treg therapy in transplantation are currently entering phases IIa and IIb, with the majority of these employing polyclonal Treg populations that harbor a broad specificity. Enhancing Treg specificity is possible with the use of chimeric antigen receptors (CARs), which can be customized to respond to a specific human leukocyte antigen (HLA). In this study, we build on our previous work in the development of HLA-A2 CAR-Tregs by further equipping cells with the constitutive expression of interleukin 10 (IL-10) and an imaging reporter as additional payloads. Cells were engineered to express combinations of these domains and assessed for phenotype and function. Cells expressing the full construct maintained a stable phenotype after transduction, were specifically activated by HLA-A2, and suppressed alloresponses potently. The addition of IL-10 provided an additional advantage to suppressive capacity. This study therefore provides an important proof-of-principle for this cell engineering approach for next-generation Treg therapy in transplantation.
Collapse
Affiliation(s)
- Yasmin R. Mohseni
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Adeel Saleem
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
- Imaging Therapies and Cancer GroupComprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College LondonLondonUK
- Department of Haematology and Precision MedicineKings College HospitalLondonUK
| | - Sim L. Tung
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Caroline Dudreuilh
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Cameron Lang
- Imaging Therapies and Cancer GroupComprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College LondonLondonUK
| | - Qi Peng
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Alessia Volpe
- Imaging Therapies and Cancer GroupComprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College LondonLondonUK
| | - George Adigbli
- Transplantation Research & Immunology Group, Nuffield Department of Surgical SciencesUniversity of Oxford, Oxford, UK
| | - Amy Cross
- Transplantation Research & Immunology Group, Nuffield Department of Surgical SciencesUniversity of Oxford, Oxford, UK
| | - Joanna Hester
- Transplantation Research & Immunology Group, Nuffield Department of Surgical SciencesUniversity of Oxford, Oxford, UK
| | - Farzin Farzaneh
- Department of Haematological MedicineSchool of Cancer and Pharmaceutical Studies, King's College LondonLondonUK
| | - Cristiano Scotta
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Robert I. Lechler
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| | - Fadi Issa
- Transplantation Research & Immunology Group, Nuffield Department of Surgical SciencesUniversity of Oxford, Oxford, UK
| | - Gilbert O. Fruhwirth
- Imaging Therapies and Cancer GroupComprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College LondonLondonUK
| | - Giovanna Lombardi
- MRC Centre for Transplantation ImmunologySchool of Immunology and Microbial Sciences, King's College LondonLondonUK
| |
Collapse
|
137
|
Parsons RF, Baquerizo A, Kirchner VA, Malek S, Desai CS, Schenk A, Finger EB, Brennan TV, Parekh KR, MacConmara M, Brayman K, Fair J, Wertheim JA. Challenges, highlights, and opportunities in cellular transplantation: A white paper of the current landscape. Am J Transplant 2021; 21:3225-3238. [PMID: 34212485 DOI: 10.1111/ajt.16740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023]
Abstract
Although cellular transplantation remains a relatively small field compared to solid organ transplantation, the prospects for advancement in basic science and clinical care remain bountiful. In this review, notable historical events and the current landscape of the field of cellular transplantation are reviewed with an emphasis on islets (allo- and xeno-), hepatocytes (including bioartificial liver), adoptive regulatory immunotherapy, and stem cells (SCs, specifically endogenous organ-specific and mesenchymal). Also, the nascent but rapidly evolving field of three-dimensional bioprinting is highlighted, including its major processing steps and latest achievements. To reach its full potential where cellular transplants are a more viable alternative than solid organ transplants, fundamental change in how the field is regulated and advanced is needed. Greater public and private investment in the development of cellular transplantation is required. Furthermore, consistent with the call of multiple national transplant societies for allo-islet transplants, the oversight of cellular transplants should mirror that of solid organ transplants and not be classified under the unsustainable, outdated model that requires licensing as a drug with the Food and Drug Administration. Cellular transplantation has the potential to bring profound benefit through progress in bioengineering and regenerative medicine, limiting immunosuppression-related toxicity, and providing markedly reduced surgical morbidity.
Collapse
Affiliation(s)
- Ronald F Parsons
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia
| | - Angeles Baquerizo
- Scripps Center for Cell and Organ Transplantation, La Jolla, California
| | - Varvara A Kirchner
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Sayeed Malek
- Division of Transplant Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chirag S Desai
- Division of Transplantation, Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Austin Schenk
- Division of Transplantation, Department of Surgery, Ohio State University, Columbus, Ohio
| | - Erik B Finger
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Todd V Brennan
- Department of Surgery, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kalpaj R Parekh
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Malcolm MacConmara
- Division of Surgical Transplantation, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kenneth Brayman
- Division of Transplantation, Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Jeffrey Fair
- Division of Transplant Surgery, Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Jason A Wertheim
- Departments of Surgery and Biomedical Engineering, University of Arizona Health Sciences, Tucson, Arizona
| | | |
Collapse
|
138
|
Safinia N, Vaikunthanathan T, Lechler RI, Sanchez‐Fueyo A, Lombardi G. Advances in Liver Transplantation: where are we in the pursuit of transplantation tolerance? Eur J Immunol 2021; 51:2373-2386. [PMID: 34375446 PMCID: PMC10015994 DOI: 10.1002/eji.202048875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/07/2021] [Accepted: 07/23/2021] [Indexed: 12/22/2022]
Abstract
Liver transplantation is the ultimate treatment option for end-stage liver disease. Breakthroughs in surgical practice and immunosuppression have seen considerable advancements in survival after transplantation. However, the intricate management of immunosuppressive regimens, balancing desired immunological quiescence while minimizing toxicity has proven challenging. Diminishing improvements in long-term morbidity and mortality have been inextricably linked with the protracted use of these medications. As such, there is now enormous interest to devise protocols that will allow us to minimize or completely withdraw immunosuppressants after transplantation. Immunosuppression withdrawal trials have proved the reality of tolerance following liver transplantation, however, without intervention will only occur after several years at the risk of potential cumulative immunosuppression-related morbidity. Focus has now been directed at accelerating this phenomenon through tolerance-inducing strategies. In this regard, efforts have seen the use of regulatory cell immunotherapy. Here we focus particularly on regulatory T cells, discussing preclinical data that propagated several clinical trials of adoptive cell therapy in liver transplantation. Furthermore, we describe efforts to further optimize the specificity and survival of regulatory cell therapy guided by concurrent immunomonitoring studies and the development of novel technologies including chimeric antigen receptors and co-administration of low-dose IL-2.
Collapse
Affiliation(s)
- Niloufar Safinia
- Division of Transplantation Immunology & Mucosal BiologyKing's College LondonLondonUK
| | | | - Robert Ian Lechler
- Division of Transplantation Immunology & Mucosal BiologyKing's College LondonLondonUK
| | | | - Giovanna Lombardi
- Division of Transplantation Immunology & Mucosal BiologyKing's College LondonLondonUK
| |
Collapse
|
139
|
Scott DW. Driving CARs to BARs: The Winding Road to Specific Regulatory T Cells for Tolerance. Front Immunol 2021; 12:742719. [PMID: 34552599 PMCID: PMC8450509 DOI: 10.3389/fimmu.2021.742719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) transduced T cells have significantly improved cancer immunotherapy. Similarly, engineering regulatory T cells (Treg) with specific receptors to endow specificity and increase efficacy of Tregs holds great promise for therapy of a variety of adverse immune responses. In this review, we focus on our approaches using retroviral transduction of specific T-cell receptors, single chain variable fragments (scFv) or antigen in models of monogenic diseases, autoimmunity and allergy. The advantages of each of these for different targets diseases are discussed as well as their potential for clinical translation.
Collapse
Affiliation(s)
- David W Scott
- Department of Medicine (MED), Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
140
|
Maanaoui M, Kerr-Conte J. Pushing the boundaries of organs before it's too late: pre-emptive regeneration. Transpl Int 2021; 34:1761-1769. [PMID: 34532871 DOI: 10.1111/tri.13969] [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: 04/13/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 11/28/2022]
Abstract
Solid organ transplantation is marked by accelerated aging and inexorable fibrosis. It is crucial to promote strategies to attenuate, or to reverse, damage before organ failure. Hence, the objective of this article is to provide insight into strategies, which aim to regenerate or rejuvenate the transplanted organs. Cell therapy with mesenchymal stromal cells is currently under investigation because of their antifibrotic properties. Their ability to promote mitochondrial biogenesis, and to transfer mitochondria to wounded cells, is another approach to boost the organ regeneration. Other teams have investigated bioengineered organs, which consists of decellularization of the damaged organ followed by recellularization. Lastly, the development of CAR-T cell-based technologies may revolutionize the field of transplantation, as recent preclinical studies showed that CAR-T cells could efficiently clear senescent cells from an organ and reverse fibrosis. Ultimately, these cutting-edge strategies may bring the holy grail of a pre-emptive regenerated organ closer to reality.
Collapse
Affiliation(s)
- Mehdi Maanaoui
- Department of Nephrology, CHU Lille, Lille, France.,Inserm, CHU Lille, Institut Pasteur Lille, U1190 - EGID, Univ. Lille, Lille, France
| | - Julie Kerr-Conte
- Inserm, CHU Lille, Institut Pasteur Lille, U1190 - EGID, Univ. Lille, Lille, France
| |
Collapse
|
141
|
Hu M, Rogers NM, Li J, Zhang GY, Wang YM, Shaw K, O'Connell PJ, Alexander SI. Antigen Specific Regulatory T Cells in Kidney Transplantation and Other Tolerance Settings. Front Immunol 2021; 12:717594. [PMID: 34512640 PMCID: PMC8428972 DOI: 10.3389/fimmu.2021.717594] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Kidney transplantation is the most common solid organ transplant and the best current therapy for end-stage kidney failure. However, with standard immunosuppression, most transplants develop chronic dysfunction or fail, much of which is due to chronic immune injury. Tregs are a subset of T cells involved in limiting immune activation and preventing autoimmune disease. These cells offer the potential to provide tolerance or to allow reduction in immunosuppression in kidney transplants. The importance of Tregs in kidney transplantation has been shown in a number of seminal mouse and animal studies, including those with T cell receptors (TCRs) transgenic Tregs (TCR-Tregs) or Chimeric Antigen Receptor (CAR) Tregs (CAR-Tregs) showing that specificity increases the potency of Treg function. Here we outline the animal and human studies and clinical trials directed at using Tregs in kidney transplantation and other tolerance settings and the various modifications to enhance allo-specific Treg function in vivo and in vitro.
Collapse
Affiliation(s)
- Min Hu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Jennifer Li
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Geoff Y Zhang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Karli Shaw
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW, Australia
| |
Collapse
|
142
|
Hefazi M, Bolivar-Wagers S, Blazar BR. Regulatory T Cell Therapy of Graft-versus-Host Disease: Advances and Challenges. Int J Mol Sci 2021; 22:9676. [PMID: 34575843 PMCID: PMC8469916 DOI: 10.3390/ijms22189676] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
Graft-versus-host disease (GVHD) is the leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Immunomodulation using regulatory T cells (Tregs) offers an exciting option to prevent and/or treat GVHD as these cells naturally function to maintain immune homeostasis, can induce tolerance following HSCT, and have a tissue reparative function. Studies to date have established a clinical safety profile for polyclonal Tregs. Functional enhancement through genetic engineering offers the possibility of improved potency, specificity, and persistence. In this review, we provide the most up to date preclinical and clinical data on Treg cell therapy with a particular focus on GVHD. We discuss the different Treg subtypes and highlight the pharmacological and genetic approaches under investigation to enhance the application of Tregs in allo-HSCT. Lastly, we discuss the remaining challenges for optimal clinical translation and provide insights as to future directions of the field.
Collapse
Affiliation(s)
- Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Sara Bolivar-Wagers
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA;
| |
Collapse
|
143
|
Miyamoto E, Takahagi A, Ohsumi A, Martinu T, Hwang D, Boonstra KM, Joe B, Umana JM, Bei KF, Vosoughi D, Liu M, Cypel M, Keshavjee S, Juvet SC. Ex vivo delivery of regulatory T cells for control of alloimmune priming in the donor lung. Eur Respir J 2021; 59:13993003.00798-2021. [PMID: 34475226 DOI: 10.1183/13993003.00798-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/17/2021] [Indexed: 11/05/2022]
Abstract
Survival after lung transplantation (LTx) is hampered by uncontrolled inflammation and alloimmunity. Regulatory T cells (Tregs) are being studied as a cellular therapy in solid organ transplantation. Whether these systemically administered Tregs can function at the appropriate location and time is an important concern. We hypothesized that in vitro expanded, recipient-derived Tregs can be delivered to donor lungs prior to LTx via ex vivo lung perfusion (EVLP), maintaining their immunomodulatory ability.In a rat model, Wistar Kyoto (WKy) CD4+CD25high Tregs were expanded in vitro prior to EVLP. Expanded Tregs were administered to Fisher 344 (F344) donor lungs during EVLP; left lungs were transplanted into WKy recipients. Treg localisation and function post-transplant were assessed. In a proof-of-concept experiment, cryopreserved expanded human CD4+CD25+CD127low Tregs were thawed and injected into discarded human lungs during EVLP.Rat Tregs entered the lung parenchyma and retained suppressive function. Expanded Tregs had no adverse effect on donor lung physiology during EVLP; lung water as measured by wet-to-dry weight ratio was reduced by Treg therapy. The administered cells remained in the graft at 3 days post-transplant where they reduced activation of intragraft effector CD4+ T cells; these effects were diminished by day 7. Human Tregs entered the lung parenchyma during EVLP where they expressed key immunoregulatory molecules (CTLA4+, 4-1BB+, CD39+, and CD15s+).Pre-transplant Treg administration can inhibit alloimmunity within the lung allograft at early time points post- transplant. Our organ-directed approach has potential for clinical translation.
Collapse
Affiliation(s)
- Ei Miyamoto
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Takahagi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Ohsumi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hwang
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Kristen M Boonstra
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Betty Joe
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Juan Mauricio Umana
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ke F Bei
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Vosoughi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Stephen C Juvet
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
144
|
Singh RP, Bischoff DS, Hahn BH. CD8 + T regulatory cells in lupus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:147-156. [PMID: 35880241 PMCID: PMC9242525 DOI: 10.2478/rir-2021-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/23/2021] [Indexed: 04/11/2023]
Abstract
T regulatory cells (Tregs) have a key role in the maintenance of immune homeostasis and the regulation of immune tolerance by preventing the inflammation and suppressing the autoimmune responses. Numerical and functional deficits of these cells have been reported in systemic lupus erythematosus (SLE) patients and mouse models of SLE, where their imbalance and dysregulated activities have been reported to significantly influence the disease pathogenesis, progression and outcomes. Most studies in SLE have focused on CD4+ Tregs and it has become clear that a critical role in the control of immune tolerance after the breakdown of self-tolerance is provided by CD8+ Tregs. Here we review the role, cellular and molecular phenotypes, and mechanisms of action of CD8+ Tregs in SLE, including ways to induce these cells for immunotherapeutic modulation in SLE.
Collapse
Affiliation(s)
- Ram P. Singh
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
| | - David S. Bischoff
- Research Service, Veteran Administration Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bevra H. Hahn
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
145
|
Pilat N, Lefsihane K, Brouard S, Kotsch K, Falk C, Steiner R, Thaunat O, Fusil F, Montserrat N, Amarelli C, Casiraghi F. T- and B-cell therapy in solid organ transplantation: current evidence and future expectations. Transpl Int 2021; 34:1594-1606. [PMID: 34448274 DOI: 10.1111/tri.13972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/08/2021] [Indexed: 01/13/2023]
Abstract
Cell therapy has emerged as an attractive therapeutic option in organ transplantation. During the last decade, the therapeutic potency of Treg immunotherapy has been shown in various preclinical animal models and safety was demonstrated in first clinical trials. However, there are still critical open questions regarding specificity, survival, and migration to the target tissue so the best Treg population for infusion into patients is still under debate. Recent advances in CAR technology hold the promise for Treg-functional superiority. Another exciting strategy is the generation of B-cell antibody receptor (BAR) Treg/cytotoxic T cells to specifically regulate or deplete alloreactive memory B cells. Finally, B cells are also capable of immune regulation, making them promising candidates for immunomodulatory therapeutic strategies. This article summarizes available literature on cell-based innovative therapeutic approaches aiming at modulating alloimmune response for transplantation. Crucial areas of investigation that need a joined effort of the transplant community for moving the field toward successful achievement of tolerance are highlighted.
Collapse
Affiliation(s)
- Nina Pilat
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Katia Lefsihane
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Sophie Brouard
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Katja Kotsch
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Department for General and Visceral Surgery, Berlin Institute of Health, Berlin, Germany
| | - Christine Falk
- Institute of Transplant Immunology, Hannover Medical School, MHH, Hannover, Germany
| | - Romy Steiner
- Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Olivier Thaunat
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.,Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Floriane Fusil
- International Center of Infectiology Research (CIRI), French Institute of Health and Medical Research (INSERM) Unit 1111, Claude Bernard University Lyon I, National Center for Scientific Research (CNRS) Mixed University Unit (UMR) 5308, Ecole Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Cristiano Amarelli
- Department of Cardiac Surgery and Transplants Monaldi, A.O. dei Colli, Naples, Italy
| | | |
Collapse
|
146
|
Jin X, Xu Q, Pu C, Zhu K, Lu C, Jiang Y, Xiao L, Han Y, Lu L. Therapeutic efficacy of anti-CD19 CAR-T cells in a mouse model of systemic lupus erythematosus. Cell Mol Immunol 2021; 18:1896-1903. [PMID: 32472023 PMCID: PMC8322088 DOI: 10.1038/s41423-020-0472-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/09/2020] [Indexed: 11/10/2022] Open
Abstract
Dysregulated B-cell activation plays pivotal roles in systemic lupus erythematosus (SLE), which makes B-cell depletion a potential strategy for SLE treatment. The clinical success of anti-CD19 CAR-T cells in treating B-cell malignancies has attracted the attention of researchers. In this study, we aimed to investigate the feasibility of applying anti-CD19 CAR-T cell therapy to SLE treatment in a mouse disease model. We constructed murine anti-CD19 CARs with either CD28 or 4-1BB as the intracellular costimulatory motif and evaluated the therapeutic function of the corresponding CAR-T cells by infusing them into MRL-lpr mice. Furthermore, anti-CD19 CAR-T cells were transferred to MRL-lpr mice before the onset of disease to determine their role in SLE prevention. According to our observations, compared with antibody treatment, the adoptive transfer of our anti-CD19 CAR-T cells showed a more sustained B-cell-depletion effect in MRL-lpr mice. The transfer of syngeneic anti-CD19 CAR-T cells not only prevented disease pathogenesis before the onset of disease symptoms but also displayed therapeutic benefits at a later stage after disease progression. We also tried to optimize the treatment strategy and found that compared with CAR-T cells with the CD28 costimulatory motif, CAR-T cells with the 4-1BB costimulatory motif showed better therapeutic efficiency without cell enrichment. Taken together, these results show that anti-CD19 CAR-T cell therapy was effective in the prevention and treatment of a murine model of SLE, indicating its potential for clinical use in patients.
Collapse
Affiliation(s)
- Xuexiao Jin
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, 314400, PR China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China
| | - Qin Xu
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China
| | - Chengfei Pu
- Innovative Cellular Therapeutics Co., Ltd., Shanghai, 201203, PR China
| | - Kaixiang Zhu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, 314400, PR China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Cheng Lu
- Innovative Cellular Therapeutics Co., Ltd., Shanghai, 201203, PR China
| | - Yu Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Lei Xiao
- Innovative Cellular Therapeutics Co., Ltd., Shanghai, 201203, PR China
| | - Yongmei Han
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China.
| | - Linrong Lu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining, 314400, PR China.
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, PR China.
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, PR China.
| |
Collapse
|
147
|
Abstract
Technological advancements in blood glucose monitoring and therapeutic insulin administration have improved the quality of life for people with type 1 diabetes. However, these efforts fall short of replicating the exquisite metabolic control provided by native islets. We examine the integrated advancements in islet cell replacement and immunomodulatory therapies that are coalescing to enable the restoration of endogenous glucose regulation. We highlight advances in stem cell biology and graft site design, which offer innovative sources of cellular material and improved engraftment. We also cover cutting-edge approaches for preventing allograft rejection and recurrent autoimmunity. These insights reflect a growing understanding of type 1 diabetes etiology, β cell biology, and biomaterial design, together highlighting therapeutic opportunities to durably replace the β cells destroyed in type 1 diabetes.
Collapse
Affiliation(s)
- Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, and Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- University of Florida Diabetes Institute, University of Florida, Gainesville, FL 32610, USA
| | - Holger A Russ
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cherie L Stabler
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
148
|
Fung VCW, Rosado-Sánchez I, Levings MK. Transduction of Human T Cell Subsets with Lentivirus. Methods Mol Biol 2021; 2285:227-254. [PMID: 33928557 DOI: 10.1007/978-1-0716-1311-5_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Lentivirus-mediated gene transfer is an efficient method to introduce a variety of transgenes to human T cells. Here we describe a protocol to transduce human CD4+, CD8+, or CD4+ regulatory T cells. To illustrate the method, we use transduction with lentivirus encoding an HLA-A2-specific chimeric antigen receptor (CAR) and a transduction marker as an example. Methods to isolate, transduce, purify, and expand CD4+ and CD8+ T cells as well as regulatory T cells are provided. We also describe how to carry out cytotoxicity or suppression assays to assess the function of the resulting CAR T cell or CAR regulatory T cells, respectively.
Collapse
Affiliation(s)
- Vivian C W Fung
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Isaac Rosado-Sánchez
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada. .,BC Children's Hospital Research Institute, Vancouver, BC, Canada. .,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
149
|
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: 21] [Impact Index Per Article: 5.3] [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
|
150
|
Thomson AW, Sasaki K, Ezzelarab MB. Non-human Primate Regulatory T Cells and Their Assessment as Cellular Therapeutics in Preclinical Transplantation Models. Front Cell Dev Biol 2021; 9:666959. [PMID: 34211972 PMCID: PMC8239398 DOI: 10.3389/fcell.2021.666959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
Non-human primates (NHP) are an important resource for addressing key issues regarding the immunobiology of regulatory T cells (Treg), their in vivo manipulation and the translation of adoptive Treg therapy to clinical application. In addition to their phenotypic and functional characterization, particularly in cynomolgus and rhesus macaques, NHP Treg have been isolated and expanded successfully ex vivo. Their numbers can be enhanced in vivo by administration of IL-2 and other cytokines. Both polyclonal and donor antigen (Ag) alloreactive NHP Treg have been expanded ex vivo and their potential to improve long-term outcomes in organ transplantation assessed following their adoptive transfer in combination with various cytoreductive, immunosuppressive and "Treg permissive" agents. In addition, important insights have been gained into the in vivo fate/biodistribution, functional stability, replicative capacity and longevity of adoptively-transferred Treg in monkeys. We discuss current knowledge of NHP Treg immunobiology, methods for their in vivo expansion and functional validation, and results obtained testing their safety and efficacy in organ and pancreatic islet transplantation models. We compare and contrast results obtained in NHP and mice and also consider prospects for future, clinically relevant studies in NHP aimed at improved understanding of Treg biology, and innovative approaches to promote and evaluate their therapeutic potential.
Collapse
Affiliation(s)
- Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kazuki Sasaki
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Mohamed B. Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|