Review
Current status and perspectives of regulatory T cell-based therapy

https://doi.org/10.1016/j.jgg.2022.05.005Get rights and content

Abstract

The CD4+FOXP3+ regulatory T (Treg) cells are essential for maintaining immune homeostasis in healthy individuals. Results from clinical trials of Treg cell-based therapies in patients with graft versus host disease (GVHD), type 1 diabetes (T1D), liver transplantation, and kidney transplantation have demonstrated that adoptive transfer of Treg cells is emerging as a promising strategy to promote immune tolerance. Here we provide an overview of recent progresses and current challenges of Treg cell-based therapies. We summarize the completed and ongoing clinical trials with human Treg cells. Notably, a few of the chimeric antigen receptor (CAR)-Treg cell therapies are currently undergoing clinical trials. Meanwhile, we describe the new strategies for engineering Treg cells used in preclinical studies. Finally, we envision that the use of novel synthetic receptors, metabolic regulators, combined therapies, and in vivo generated antigen-specific or engineered Treg cells through the delivery of modified mRNA and CRISPR-based gene editing will further promote the advances of next-generation Treg cell therapies.

Introduction

The adaptive immune system can recognize and eliminate pathogens while maintaining the state of unresponsiveness toward self-tissues (Ferreira et al., 2019). This state is termed as immune tolerance, controlled by multiple regulators. Treg cells, a small subset of CD4+ T lymphocytes with immunosuppressive function, are identified as the main cell type responsible for T cell-mediated immune tolerance (Sakaguchi et al., 1995). In addition to maintaining immune tolerance, Treg cells also perform specialized functions in tissue homeostasis and repair (Panduro et al., 2016). Functional or numerical defects of Treg cells have been described in many autoimmune diseases (Miyara et al., 2011; Dominguez-Villar and Hafler, 2018). Adoptive transfer of Treg cells is emerging as an attractive therapeutic candidate, which can restore immune tolerance in autoimmune diseases and induce tolerance to allogeneic cells/tissues in transplantation. Completed clinical trials in patients with T1D (Bluestone et al., 2015; Dong et al., 2021), liver transplantation (Levitsky et al., 2009), kidney transplantation (Roemhild et al., 2020; Sawitzki et al., 2020), and GVHD (Brunstein et al., 2011; MacMillan et al., 2021; Pierini et al., 2021), have indicated the feasibility, safety, and efficacy of Treg cell therapies. More ongoing clinical trials will continue to validate their therapeutic potential.

Treg cells are characterized by the high expression of forkhead box protein 3 (FOXP3) and CD25. In addition to CD4+FOXP3+ Treg cells, CD4+FOXP3 type 1 Treg (Tr1) cells can also exert immunosuppressive function through the production of IL-10 and granzyme B, as well as through surface molecules (Song et al., 2021). Type 3 T helper (Th3) cells also mediate suppressive activity via secreting high levels of transforming growth factor-β (TGF-β) (Chen et al., 1994). CD4+ FOXP3+ Treg cells are the most frequently used in clinical trials of Treg cell therapies to restore or induce immune tolerance. In this Review, we focus on the therapeutic potential of CD4+FOXP3+ Treg cells. Here, we introduce the current knowledge regarding the biology of Treg cells. We review the cell sources and procedures of Treg cell therapies used in clinical trials. Meanwhile, we summarize new efforts in Treg cell therapies to improve efficacy and durability. These endeavors include transductions of TCR, TRuC, and CAR, and the use of metabolic regulators. Finally, we describe design strategies and perspectives of next-generation engineered Treg cells through the use of novel synthetic receptors, modified mRNA, and CRISPR-based gene editing.

Section snippets

Classification of Treg cells

Treg cells can develop in the thymus and periphery (Savage et al., 2020). The thymus is a key site where T cell tolerance is established. In the process of negative selection, thymocytes exhibiting overt reactivity to self-peptides, presented by major histocompatibility complex class (MHC) molecules, are eliminated either by clonal deletion or diverge into innate-like T cell lineages. However, some self-antigens reactive thymocytes differentiate into FOXP3-expressing Treg cells (thymic

Engineered Treg cells

Currently, several strategies (Raffin et al., 2020) are developed to generate Treg cells for adoptive immunotherapy. (1) Polyclonal Treg cells isolated from peripheral blood are frequently used in clinical trials and expanded in vitro using anti-CD3/CD28 beads plus high-dose IL-2 to generate numbers of Treg cells for adoptive immunotherapy. (2) Using APCs (from the donor) to specifically stimulate alloreactive Treg cells (from the recipient) in vitro is mainly used in GVHD prevention.

Perspectives

Although results from the completed clinical trials for GVHD, T1D, liver transplantation, and kidney transplantation have proved the efficacy and safety of Treg-based therapies, there are many challenges and unsolved problems. It is reported that CAR expression is often associated with tonic signaling, which can result in CAR-T cell dysfunction. CAR tonic signaling may be defined as excessively and sustained CAR-induced activation in a ligand-dependent or-independent manner. The impact of CAR

Conflict of interest

The authors declare no conflict of interests.

Acknowledgments

This research is supported by National Science Foundation for Distinguished Young Scholars (31525008); National Natural Science Foundation of China (32130041, 81830051, 31961133011); The National Key Research and Development Project (2019YFA0906102); Shanghai Collaborative Innovation Center of Cellular Homeostasis Regulation and Human Diseases; Shanghai Jiao Tong University (SJTU) - The Chinese University of Hong Kong (CUHK) Joint Research Collaboration Fund and the Fundamental Research Funds

References (142)

  • Z. Chen et al.

    The ubiquitin ligase Stub1 negatively modulates regulatory T cell suppressive activity by promoting degradation of the transcription factor Foxp3

    Immunity

    (2013)
  • I.E. Dijke et al.

    Discarded human thymus is a novel source of stable and long-lived therapeutic regulatory T cells

    Am. J. Transplant.

    (2016)
  • E. Elinav et al.

    Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice

    Gastroenterology

    (2008)
  • D. Friedmann-Morvinski et al.

    Redirected primary T cells harboring a chimeric receptor require costimulation for their antigen-specific activation

    Blood

    (2005)
  • L. Fultang et al.

    Metabolic engineering against the arginine microenvironment enhances CAR-T cell proliferation and therapeutic activity

    Blood

    (2020)
  • W.J. Grossman et al.

    Human T regulatory cells can use the perforin pathway to cause autologous target cell death

    Immunity

    (2004)
  • K.L. Hippen et al.

    Generation and large-scale expansion of human inducible regulatory T cells that suppress graft-versus-host disease

    Am. J. Transplant.

    (2011)
  • C.M. Hull et al.

    Generation of human islet-specific regulatory T cells by TCR gene transfer

    J. Autoimmun.

    (2017)
  • Y.C. Kim et al.

    Engineered antigen-specific human regulatory T cells: immunosuppression of FVIII-specific T- and B-cell responses

    Blood

    (2015)
  • Y.C. Kim et al.

    Engineered MBP-specific human Tregs ameliorate MOG-induced EAE through IL-2-triggered inhibition of effector T cells

    J. Autoimmun.

    (2018)
  • M.A. Kluger et al.

    Treg17 cells are programmed by Stat3 to suppress Th17 responses in systemic lupus

    Kidney Int.

    (2016)
  • D. Kolodin et al.

    Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice

    Cell Metabol.

    (2015)
  • J. Levitsky et al.

    Immunoregulatory profiles in liver transplant recipients on different immunosuppressive agents

    Hum. Immunol.

    (2009)
  • A.N. Macintyre et al.

    The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function

    Cell Metabol.

    (2014)
  • M.L. MacMillan et al.

    First-in-human phase 1 trial of induced regulatory T cells for graft-versus-host disease prophylaxis in HLA-matched siblings

    Blood Adv

    (2021)
  • M. Miyara et al.

    Human FoxP3+ regulatory T cells in systemic autoimmune diseases

    Autoimmun. Rev.

    (2011)
  • L. Morsut et al.

    Engineering customized cell sensing and response behaviors using synthetic Notch receptors.

    Cell

    (2016)
  • M. Noval Rivas et al.

    Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy

    Immunity

    (2015)
  • F. Noyan et al.

    Prevention of allograft rejection by use of regulatory T cells with an MHC-specific chimeric antigen receptor

    Am. J. Transplant.

    (2017)
  • N. Ohkura et al.

    Development and maintenance of regulatory T cells

    Immunity

    (2013)
  • A. Pierini et al.

    Haploidentical age-adapted myeloablative transplant and regulatory and effector T cells for acute myeloid leukemia

    Blood Adv.

    (2021)
  • B.R. Powell et al.

    An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy

    J. Pediatr.

    (1982)
  • J. Rana et al.

    CAR- and TRuC-redirected regulatory T cells differ in capacity to control adaptive immunity to FVIII

    Mol. Ther.

    (2021)
  • B. Akkaya et al.

    Regulatory T cells mediate specific suppression by depleting peptide-MHC class II from dendritic cells

    Nat. Immunol.

    (2019)
  • N. Ali et al.

    Regulatory T cells in skin facilitate epithelial stem cell differentiation

    Cell

    (2017)
  • B. Allard et al.

    The adenosine pathway in immuno-oncology

    Nat. Rev. Clin. Oncol.

    (2020)
  • A. Arvey et al.

    Genetic and epigenetic variation in the lineage specification of regulatory T cells

    Elife

    (2015)
  • M. Ayyoub et al.

    Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the TH17 lineage-specific transcription factor RORγt

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • M. Battaglia et al.

    Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients

    J. Immunol.

    (2006)
  • C.L. Bennett et al.

    The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3

    Nat. Genet.

    (2001)
  • J.A. Bluestone et al.

    Type 1 diabetes immunotherapy using polyclonal regulatory T cells

    Sci. Transl. Med.

    (2015)
  • M. Bonelli et al.

    Quantitative and qualitative deficiencies of regulatory T cells in patients with systemic lupus erythematosus (SLE)

    Int. Immunol.

    (2008)
  • M.E. Brunkow et al.

    Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse

    Nat. Genet.

    (2001)
  • T.A. Chatila et al.

    JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome

    J. Clin. Invest.

    (2000)
  • Y. Chen et al.

    Regulatory T cell clones induced by oral tolerance: suppression of autoimmune encephalomyelitis

    Science

    (1994)
  • T. Chinen et al.

    An essential role for the IL-2 receptor in Treg cell function

    Nat. Immunol.

    (2016)
  • L.W. Collison et al.

    The inhibitory cytokine IL-35 contributes to regulatory T-cell function

    Nature

    (2007)
  • J.L. Coombes et al.

    A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism

    J. Exp. Med.

    (2007)
  • N.A.J. Dawson et al.

    Functional effects of chimeric antigen receptor co-receptor signaling domains in human regulatory T cells

    Sci. Transl. Med.

    (2020)
  • S. Deaglio et al.

    Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression

    J. Exp. Med.

    (2007)
  • Cited by (12)

    View all citing articles on Scopus
    View full text