1
|
Hsieh WC, Hsu TS, Wu KW, Lai MZ. Therapeutic application of regulatory T cell in osteoarthritis. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00083-0. [PMID: 40300967 DOI: 10.1016/j.jmii.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/07/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Regulatory T cells (Tregs) are the specific T cell population that suppress inflammatory immunity. Independent of their inhibitory activities, Tregs exhibit unique capacity to repair tissue damage. Rapid progresses are made in the processing and engineering of Tregs for clinical applications. Tregs have been used in the treatment of autoimmune diseases, transplantation rejection and graft-versus-host disease. Osteoarthritis is one of the major diseases that affect at least 600 million people worldwide. Osteoarthritis is characterized by physical erosion of cartilage, accompanied with chronic and low-grade inflammation. Tregs possess abilities to increase osteoclast differentiation and bone resorption, repair bone physical damage, and increase bone mass. Tregs are therefore candidate therapeutics for osteoarthritis for both inflammation resolution and tissue repairing. In this review, we will summarize the recent development in using Tregs in immunotherapy, and the potential of using Tregs in osteoarthritis.
Collapse
Affiliation(s)
- Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Kuan-Wen Wu
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ming-Zong Lai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
2
|
Ho QY, Hester J, Issa F. Regulatory cell therapy for kidney transplantation and autoimmune kidney diseases. Pediatr Nephrol 2025; 40:39-52. [PMID: 39278988 PMCID: PMC11584488 DOI: 10.1007/s00467-024-06514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 09/18/2024]
Abstract
Regulatory cell therapies, including regulatory T cells and mesenchymal stromal cells, have shown promise in early clinical trials for reducing immunosuppression burden in transplantation. While regulatory cell therapies may also offer potential for treating autoimmune kidney diseases, data remains sparse, limited mainly to preclinical studies. This review synthesises current literature on the application of regulatory cell therapies in these fields, highlighting the safety and efficacy shown in existing clinical trials. We discuss the need for further clinical validation, optimisation of clinical and immune monitoring protocols, and the challenges of manufacturing and quality control under Good Manufacturing Practice conditions, particularly for investigator-led trials. Additionally, we explore the potential for expanding clinical indications and the unique challenges posed in paediatric applications. Future directions include scaling up production, refining protocols to ensure consistent quality across manufacturing sites, and extending applications to other immune-mediated diseases.
Collapse
Affiliation(s)
- Quan Yao Ho
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK.
| |
Collapse
|
3
|
Chmiel J, Stasiak M, Skrzypkowska M, Samson L, Łuczkiewicz P, Trzonkowski P. Regulatory T lymphocytes as a treatment method for rheumatoid arthritis - Superiority of allogeneic to autologous cells. Heliyon 2024; 10:e36512. [PMID: 39319132 PMCID: PMC11419861 DOI: 10.1016/j.heliyon.2024.e36512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Cellular therapies utilizing regulatory T cells (Tregs) have flourished in the autoimmunity space as a new pillar of medicine. These cells have shown a great promise in the treatment of such devastating conditions as type 1 diabetes mellitus (T1DM), systemic lupus erythematosus (SLE) and graft versus host disease (GVHD). Novel treatment protocols, which utilize Tregs-mediated suppressive mechanisms, are based on the two main strategies: administration of immunomodulatory factors affecting Tregs or adoptive cell transfer (ACT). ACT involves extraction, in vitro expansion and subsequent administration of Tregs that could be either of autologous or allogeneic origin. Rheumatoid arthritis (RA) is another autoimmune candidate where this treatment approach is being considered. RA remains an especially challenging adversary since it is one of the most frequent and debilitating conditions among all autoaggressive disorders. Noteworthy, Tregs circulating in RA patients' blood have been proven defective and unable to suppress inflammation and joint destruction. With this knowledge, adoptive transfer of compromised autologous Tregs in the fledgling clinical trials involving RA patients should be reconsidered. In this article we hypothesize that incorporation of healthy donor allogeneic Tregs may provide more lucid and beneficial results.
Collapse
Affiliation(s)
- Joanna Chmiel
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Mariusz Stasiak
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Maria Skrzypkowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| | - Lucjan Samson
- University Clinical Centre in Gdańsk, Second Clinic of Orthopaedics and Kinetic Organ Traumatology, Poland
- Faculty of Medicine, Medical University of Gdańsk, Poland
| | | | - Piotr Trzonkowski
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Poland
| |
Collapse
|
4
|
Ekwe AP, Au R, Zhang P, McEnroe BA, Tan ML, Saldan A, Henden AS, Hutchins CJ, Henderson A, Mudie K, Kerr K, Fuery M, Kennedy GA, Hill GR, Tey SK. Clinical grade multiparametric cell sorting and gene-marking of regulatory T cells. Cytotherapy 2024; 26:719-728. [PMID: 38530690 DOI: 10.1016/j.jcyt.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND AIMS Regulatory T cells (Tregs) are the main mediators of peripheral tolerance. Treg-directed therapy has shown promising results in preclinical studies of diverse immunopathologies. At present, the clinical applicability of adoptive Treg transfer is limited by difficulties in generating Tregs at sufficient cell dose and purity. METHODS We developed a Good Manufacturing Practice (GMP) compliant method based on closed-system multiparametric Fluorescence-Activated Cell Sorting (FACS) to purify Tregs, which are then expanded in vitro and gene-marked with a clinical grade retroviral vector to enable in vivo fate tracking. Following small-scale optimization, we conducted four clinical-scale processing runs. RESULTS We showed that Tregs could be enriched to 87- 92% purity following FACS-sorting, and expanded and transduced to yield clinically relevant cell dose of 136-732×106 gene-marked cells, sufficient for a cell dose of at least 2 × 106 cells/kg. The expanded Tregs were highly demethylated in the FOXP3 Treg-specific demethylated region (TSDR), consistent with bona fide natural Tregs. They were suppressive in vitro, but a small percentage could secrete proinflammatory cytokines, including interferon-γ and interleukin-17A. CONCLUSIONS This study demonstrated the feasibility of isolating, expanding and gene-marking Tregs in clinical scale, thus paving the way for future phase I trials that will advance knowledge about the in vivo fate of transferred Tregs and its relationship with concomitant Treg-directed pharmacotherapy and clinical response.
Collapse
Affiliation(s)
- Adaeze Precious Ekwe
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Kelvin Grove, Queensland, Australia
| | - Raymond Au
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ping Zhang
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Benjamin A McEnroe
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mei Ling Tan
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Alda Saldan
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andrea S Henden
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Cheryl J Hutchins
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Ashleigh Henderson
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Kari Mudie
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Keri Kerr
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Madonna Fuery
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Glen A Kennedy
- Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Siok-Keen Tey
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Kelvin Grove, Queensland, Australia; Department of Haematology and Bone Marrow Transplantation, Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
5
|
Bender C, Wiedeman AE, Hu A, Ylescupidez A, Sietsema WK, Herold KC, Griffin KJ, Gitelman SE, Long SA. A phase 2 randomized trial with autologous polyclonal expanded regulatory T cells in children with new-onset type 1 diabetes. Sci Transl Med 2024; 16:eadn2404. [PMID: 38718135 DOI: 10.1126/scitranslmed.adn2404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/16/2024] [Indexed: 06/20/2024]
Abstract
CD4+CD25hiCD127lo/-FOXP3+ regulatory T cells (Tregs) play a key role in preventing autoimmunity. In autoimmune type 1 diabetes (T1D), adoptive transfer of autologous polyclonal Tregs has been shown to be safe in adults in phase 1 clinical trials. We explored factors contributing to efficacy of autologous polyclonal expanded Tregs (expTregs) in a randomized phase 2 multi-center, double-blind, clinical trial (Sanford/Lisata Therapeutics T-Rex phase 2 trial, ClinicalTrials.gov NCT02691247). One hundred ten treated children and adolescents with new-onset T1D were randomized 1:1:1 to high-dose (20 × 106 cells/kilogram) or low-dose (1 × 106 cells/kilogram) treatments or to matching placebo. Cytometry as well as bulk and single-cell RNA sequencing were performed on selected expTregs and peripheral blood samples from participants. The single doses of expTregs were safe but did not prevent decline in residual β cell function over 1 year compared to placebo (P = 0.94 low dose, P = 0.21 high dose), regardless of age or baseline C-peptide. ExpTregs were highly activated and suppressive in vitro. A transient increase of activated memory Tregs was detectable 1 week after infusion in the high-dose cohort, suggesting effective transfer of expTregs. However, the in vitro fold expansion of expTregs varied across participants, even when accounting for age, and lower fold expansion and its associated gene signature were linked with better C-peptide preservation regardless of Treg dose. These results suggest that a single dose of polyclonal expTregs does not alter progression in T1D; instead, Treg quality may be an important factor.
Collapse
Affiliation(s)
- Christine Bender
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Alice E Wiedeman
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Alex Hu
- Systems Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Alyssa Ylescupidez
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Kurt J Griffin
- Sanford Research, Sanford Health, and Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Stephen E Gitelman
- Department of Pediatrics, Diabetes Center, University of California at San Francisco, San Francisco, CA 94158, USA
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| |
Collapse
|
6
|
Ho P, Cahir-McFarland E, Fontenot JD, Lodie T, Nada A, Tang Q, Turka LA, Bluestone JA. Harnessing regulatory T cells to establish immune tolerance. Sci Transl Med 2024; 16:eadm8859. [PMID: 38478632 DOI: 10.1126/scitranslmed.adm8859] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/21/2024] [Indexed: 05/23/2025]
Abstract
Engineered regulatory T (Treg) cells have emerged as precision therapeutics aimed at inducing immune tolerance while reducing the risks associated with generalized immunosuppression. This Viewpoint highlights the opportunities and challenges for engineered Treg cell therapies in treating autoimmune and other inflammatory diseases.
Collapse
Affiliation(s)
- Patrick Ho
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Laurence A Turka
- Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
7
|
Dabiri H, Habibi-Anbouhi M, Ziaei V, Moghadasi Z, Sadeghizadeh M, Hajizadeh-Saffar E. Candidate Biomarkers for Targeting in Type 1 Diabetes; A Bioinformatic Analysis of Pancreatic Cell Surface Antigens. CELL JOURNAL 2024; 26:51-61. [PMID: 38351729 PMCID: PMC10864774 DOI: 10.22074/cellj.2023.1996297.1262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 08/27/2023] [Accepted: 10/24/2023] [Indexed: 02/18/2024]
Abstract
OBJECTIVE Type 1 diabetes (T1Ds) is an autoimmune disease in which the immune system invades and destroys insulin-producing cells. Nevertheless, at the time of diagnosis, about 30-40% of pancreatic beta cells are healthy and capable of producing insulin. Bi-specific antibodies, chimeric antigen receptor regulatory T cells (CAR-Treg cells), and labeled antibodies could be a new emerging option for the treatment or diagnosis of type I diabetic patients. The aim of the study is to choose appropriate cell surface antigens in the pancreas tissue for generating an antibody for type I diabetic patients. MATERIALS AND METHODS In this bioinformatics study, we extracted pancreas-specific proteins from two large databases; the Human Protein Atlas (HPA) and Genotype-Tissue Expression (GTEx) Portal. Pancreatic-enriched genes were chosen and narrowed down by Protter software for the investigation of accessible extracellular domains. The immunohistochemistry (IHC) data of the protein atlas database were used to evaluate the protein expression of selected antigens. We explored the function of candidate antigens by using the GeneCards database to evaluate the potential dysfunction or activation/hyperactivation of antigens after antibody binding. RESULTS The results showed 429 genes are highly expressed in the pancreas tissue. Also, eighteen genes encoded plasma membrane proteins that have high expression in the microarray (GEO) dataset. Our results introduced four structural proteins, including NPHS1, KIRREL2, GP2, and CUZD1, among all seventeen candidate proteins. CONCLUSION The presented antigens can potentially be used to produce specific pancreatic antibodies that guide CARTreg, bi-specific, or labeling molecules to the pancreas for treatment, detection, or other molecular targeted therapy scopes for type I diabetes.
Collapse
Affiliation(s)
- Hamed Dabiri
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Vahab Ziaei
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Moghadasi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Hadley P, Chen Y, Cline L, Han Z, Tang Q, Huang X, Desai T. Precise surface functionalization of PLGA particles for human T cell modulation. Nat Protoc 2023; 18:3289-3321. [PMID: 37853157 PMCID: PMC10775953 DOI: 10.1038/s41596-023-00887-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 07/05/2023] [Indexed: 10/20/2023]
Abstract
The biofunctionalization of synthetic materials has extensive utility for biomedical applications, but approaches to bioconjugation typically show insufficient efficiency and controllability. We recently developed an approach by building synthetic DNA scaffolds on biomaterial surfaces that enables the precise control of cargo density and ratio, thus improving the assembly and organization of functional cargos. We used this approach to show that the modulation and phenotypic adaptation of immune cells can be regulated using our precisely functionalized biomaterials. Here, we describe the three key procedures, including the fabrication of polymeric particles engrafted with short DNA scaffolds, the attachment of functional cargos with complementary DNA strands, and the surface assembly control and quantification. We also explain the critical checkpoints needed to ensure the overall quality and expected characteristics of the biological product. We provide additional experimental design considerations for modifying the approach by varying the material composition, size or cargo types. As an example, we cover the use of the protocol for human primary T cell activation and for the identification of parameters that affect ex vivo T cell manufacturing. The protocol requires users with diverse expertise ranging from synthetic materials to bioconjugation chemistry to immunology. The fabrication procedures and validation assays to design high-fidelity DNA-scaffolded biomaterials typically require 8 d.
Collapse
Affiliation(s)
- Pierce Hadley
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Yuanzhou Chen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, CA, USA
| | - Lariana Cline
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Zhiyuan Han
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Qizhi Tang
- Diabetes Center, University of California, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Xiao Huang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
- Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, CA, USA.
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| | - Tejal Desai
- UC Berkeley-UCSF Graduate Program in Bioengineering, UCSF Mission Bay Campus, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA.
- Diabetes Center, University of California, San Francisco, CA, USA.
- Cell Design Institute and Center for Synthetic Immunology, University of California, San Francisco, CA, USA.
- School of Engineering, Brown University, Providence, RI, USA.
| |
Collapse
|
9
|
Laukova M, Glatman Zaretsky A. Regulatory T cells as a therapeutic approach for inflammatory bowel disease. Eur J Immunol 2023; 53:e2250007. [PMID: 36562391 PMCID: PMC10107179 DOI: 10.1002/eji.202250007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/20/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
Foxp3+ T regulatory (Treg) cells suppress inflammation and are essential for maintaining tissue homeostasis. A growing appreciation of tissue-specific Treg functions has built interest in leveraging the endogenous suppressive mechanisms of these cells into cellular therapeutics in organ-specific diseases. Notably, Treg cells play a critical role in maintaining the intestinal environment. As a barrier site, the gut requires Treg cells to mediate interactions with the microbiota, support barrier integrity, and regulate the immune system. Without fully functional Treg cells, intestinal inflammation and microbial dysbiosis ensue. Thus, there is a particular interest in developing Treg cellular therapies for intestinal inflammatory disease, such as inflammatory bowel disease (IBD). This article reviews some of the critical pathways that are dysregulated in IBD, Treg cell mechanisms of suppression, and the efforts and approaches in the field to develop these cells as a cellular therapy for IBD.
Collapse
|
10
|
Bluestone JA, McKenzie BS, Beilke J, Ramsdell F. Opportunities for Treg cell therapy for the treatment of human disease. Front Immunol 2023; 14:1166135. [PMID: 37153574 PMCID: PMC10154599 DOI: 10.3389/fimmu.2023.1166135] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/22/2023] [Indexed: 05/09/2023] Open
Abstract
Regulatory T (Treg) cells are essential for maintaining peripheral tolerance, preventing autoimmunity, and limiting chronic inflammatory diseases. This small CD4+ T cell population can develop in the thymus and in the peripheral tissues of the immune system through the expression of an epigenetically stabilized transcription factor, FOXP3. Treg cells mediate their tolerogenic effects using multiple modes of action, including the production of inhibitory cytokines, cytokine starvation of T effector (e.g., IL-2), Teff suppression by metabolic disruption, and modulation of antigen-presenting cell maturation or function. These activities together result in the broad control of various immune cell subsets, leading to the suppression of cell activation/expansion and effector functions. Moreover, these cells can facilitate tissue repair to complement their suppressive effects. In recent years, there has been an effort to harness Treg cells as a new therapeutic approach to treat autoimmune and other immunological diseases and, importantly, to re-establish tolerance. Recent synthetic biological advances have enabled the cells to be genetically engineered to achieve tolerance and antigen-specific immune suppression by increasing their specific activity, stability, and efficacy. These cells are now being tested in clinical trials. In this review, we highlight both the advances and the challenges in this arena, focusing on the efforts to develop this new pillar of medicine to treat and cure a variety of diseases.
Collapse
|
11
|
Kaljanac M, Abken H. Do Treg Speed Up with CARs? Chimeric Antigen Receptor Treg Engineered to Induce Transplant Tolerance. Transplantation 2023; 107:74-85. [PMID: 36226849 PMCID: PMC9746345 DOI: 10.1097/tp.0000000000004316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Adoptive transfer of regulatory T cells (Treg) can induce transplant tolerance in preclinical models by suppressing alloantigen-directed inflammatory responses; clinical translation was so far hampered by the low abundance of Treg with allo-specificity in the peripheral blood. In this situation, ex vivo engineering of Treg with a T-cell receptor (TCR) or chimeric antigen receptor (CAR) provides a cell population with predefined specificity that can be amplified and administered to the patient. In contrast to TCR-engineered Treg, CAR Treg can be redirected toward a broad panel of targets in an HLA-unrestricted fashion' making these cells attractive to provide antigen-specific tolerance toward the transplanted organ. In preclinical models, CAR Treg accumulate and amplify at the targeted transplant, maintain their differentiated phenotype, and execute immune repression more vigorously than polyclonal Treg. With that, CAR Treg are providing hope in establishing allospecific, localized immune tolerance in the long term' and the first clinical trials administering CAR Treg for the treatment of transplant rejection are initiated. Here, we review the current platforms for developing and manufacturing alloantigen-specific CAR Treg and discuss the therapeutic potential and current hurdles in translating CAR Treg into clinical exploration.
Collapse
Affiliation(s)
- Marcell Kaljanac
- Division Genetic Immunotherapy, and Chair Genetic Immunotherapy, Leibniz Institute for Immunotherapy, University Regensburg, Regensburg, Germany
| | - Hinrich Abken
- Division Genetic Immunotherapy, and Chair Genetic Immunotherapy, Leibniz Institute for Immunotherapy, University Regensburg, Regensburg, Germany
| |
Collapse
|
12
|
MacDonald KN, Salim K, Levings MK. Manufacturing next-generation regulatory T-cell therapies. Curr Opin Biotechnol 2022; 78:102822. [PMID: 36332342 DOI: 10.1016/j.copbio.2022.102822] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/11/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022]
Abstract
Regulatory T-cell (Treg) therapy has shown promise in treating autoimmune diseases, transplant rejection, or graft-versus-host disease in early clinical trials. These trials have demonstrated that cell therapy using polyclonal Tregs is feasible and safe, however, the field has been limited by the lack of polyclonal cell specificity and consequent large cell numbers required, and the difficulty in generating autologous products for some patients. Thus, the field is moving toward 'next generation' Treg cell therapies that include genetic modification strategies to engineer specificity and/or modify function, as well as methods to generate Tregs in vitro. In this review, we describe how genetic modification of Tregs using viral transduction or gene editing may be incorporated into Treg manufacturing protocols. We also describe how Tregs may be generated via FOXP3 gene editing or overexpression, or by differentiation from pluripotent stem cells. The application of these various types of engineered Tregs is discussed.
Collapse
Affiliation(s)
- Katherine N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Salim
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
13
|
Tang Q, Leung J, Peng Y, Sanchez-Fueyo A, Lozano JJ, Lam A, Lee K, Greenland JR, Hellerstein M, Fitch M, Li KW, Esensten JH, Putnam AL, Lares A, Nguyen V, Liu W, Bridges ND, Odim J, Demetris AJ, Levitsky J, Taner T, Feng S. Selective decrease of donor-reactive T regs after liver transplantation limits T reg therapy for promoting allograft tolerance in humans. Sci Transl Med 2022; 14:eabo2628. [PMID: 36322627 PMCID: PMC11016119 DOI: 10.1126/scitranslmed.abo2628] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2024]
Abstract
Promoting immune tolerance to transplanted organs can minimize the amount of immunosuppressive drugs that patients need to take, reducing lifetime risks of mortality and morbidity. Regulatory T cells (Tregs) are essential for immune tolerance, and preclinical studies have shown their therapeutic efficacy in inducing transplantation tolerance. Here, we report the results of a phase 1/2 trial (ARTEMIS, NCT02474199) of autologous donor alloantigen-reactive Treg (darTreg) therapy in individuals 2 to 6 years after receiving a living donor liver transplant. The primary efficacy endpoint was calcineurin inhibitor dose reduction by 75% with stable liver function tests for at least 12 weeks. Among 10 individuals who initiated immunosuppression withdrawal, 1 experienced rejection before planned darTreg infusion, 5 received darTregs, and 4 were not infused because of failure to manufacture the minimal infusible dose of 100 × 106 cells. darTreg infusion was not associated with adverse events. Two darTreg-infused participants reached the primary endpoint, but an insufficient number of recipients were treated for assessing the efficacy of darTregs. Mechanistic studies revealed generalized Treg activation, senescence, and selective reduction of donor reactivity after liver transplantation. Overall, the ARTEMIS trial features a design concept for evaluating the efficacy of Treg therapy in transplantation. The mechanistic insight gained from the study may help guide the design of future trials.
Collapse
Affiliation(s)
- 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, San Francisco, CA 94158, USA
| | - Joey Leung
- 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
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, School of Immunology and Microbial Sciences, King’s College London University, London WC2R 2LS, UK
| | - Juan-Jose Lozano
- Bioinformatic Platform, Biomedical Research Center in Hepatic and Digestive Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alice Lam
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Karim Lee
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Medical Service, San Francisco VA Health Care System, San Francisco, CA 94121, USA
| | - Marc Hellerstein
- Nutrition Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mark Fitch
- Nutrition Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kelvin W. Li
- Nutrition Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jonathan H. Esensten
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
- Department of Lab Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Amy L. Putnam
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Angela Lares
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vinh Nguyen
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Weihong Liu
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nancy D. Bridges
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Jonah Odim
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Anthony J. Demetris
- Thomas E. Starzl Transplantation Institute and Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Josh Levitsky
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Timucin Taner
- Departments of Surgery and Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sandy Feng
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
14
|
Raugh A, Allard D, Bettini M. Nature vs. nurture: FOXP3, genetics, and tissue environment shape Treg function. Front Immunol 2022; 13:911151. [PMID: 36032083 PMCID: PMC9411801 DOI: 10.3389/fimmu.2022.911151] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/11/2022] [Indexed: 12/11/2022] Open
Abstract
The importance of regulatory T cells (Tregs) in preventing autoimmunity has been well established; however, the precise alterations in Treg function in autoimmune individuals and how underlying genetic associations impact the development and function of Tregs is still not well understood. Polygenetic susceptibly is a key driving factor in the development of autoimmunity, and many of the pathways implicated in genetic association studies point to a potential alteration or defect in regulatory T cell function. In this review transcriptomic control of Treg development and function is highlighted with a focus on how these pathways are altered during autoimmunity. In combination, observations from autoimmune mouse models and human patients now provide insights into epigenetic control of Treg function and stability. How tissue microenvironment influences Treg function, lineage stability, and functional plasticity is also explored. In conclusion, the current efficacy and future direction of Treg-based therapies for Type 1 Diabetes and other autoimmune diseases is discussed. In total, this review examines Treg function with focuses on genetic, epigenetic, and environmental mechanisms and how Treg functions are altered within the context of autoimmunity.
Collapse
Affiliation(s)
- Arielle Raugh
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, Houston, TX, United States
| | - Denise Allard
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
| | - Maria Bettini
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, United States
- *Correspondence: Maria Bettini,
| |
Collapse
|
15
|
Bernaldo-de-Quirós E, Pion M, Martínez-Bonet M, Correa-Rocha R. A New Generation of Cell Therapies Employing Regulatory T Cells (Treg) to Induce Immune Tolerance in Pediatric Transplantation. Front Pediatr 2022; 10:862807. [PMID: 35633970 PMCID: PMC9130702 DOI: 10.3389/fped.2022.862807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Kidney transplantation is the most common solid organ transplant and the preferred treatment for pediatric patients with end-stage renal disease, but it is still not a definitive solution due to immune graft rejection. Regulatory T cells (Treg) and their control over effector T cells is a crucial and intrinsic tolerance mechanism in limiting excessive immune responses. In the case of transplants, Treg are important for the survival of the transplanted organ, and their dysregulation could increase the risk of rejection in transplanted children. Chronic immunosuppression to prevent rejection, for which Treg are especially sensitive, have a detrimental effect on Treg counts, decreasing the Treg/T-effector balance. Cell therapy with Treg cells is a promising approach to restore this imbalance, promoting tolerance and thus increasing graft survival. However, the strategies used to date that employ peripheral blood as a Treg source have shown limited efficacy. Moreover, it is not possible to use this approach in pediatric patients due to the limited volume of blood that can be extracted from children. Here, we outline our innovative strategy that employs the thymus removed during pediatric cardiac surgeries as a source of therapeutic Treg that could make this therapy accessible to transplanted children. The advantageous properties and the massive amount of Treg cells obtained from pediatric thymic tissue (thyTreg) opens a new possibility for Treg therapies to prevent rejection in pediatric kidney transplants. We are recruiting patients in a clinical trial to prevent rejection in heart-transplanted children through the infusion of autologous thyTreg cells (NCT04924491). If its efficacy is confirmed, thyTreg therapy may establish a new paradigm in preventing organ rejection in pediatric transplants, and their allogeneic use would extend its application to other solid organ transplantation.
Collapse
Affiliation(s)
- Esther Bernaldo-de-Quirós
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marta Martínez-Bonet
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|