Introduction

Regulatory T (Treg) cells constitute ~2–5% of the adult peripheral blood CD4+ T cell repertoire1. Whereas the majority of immune system cells function to promote inflammation to fight pathogens and cancers, Treg cells keep immunity in check to maintain homeostasis and prevent pathology. Dysregulation of Treg cell function is linked with many diseases and the failure of these cells to control antigen-specific effector T cells can lead to autoimmune diseases, allergies and various other hyperinflammatory diseases. When a patient receives a transplant, Treg cells prevent alloreactive effector T cells from attacking the foreign tissue. In contrast, tumours enhance the Treg cell response so that anticancer immunity is suppressed. Therapies focused on Treg cells thus aim to either enhance their function (in the case of autoimmunity, allergy, transplantation and other hyperinflammatory diseases) or hinder it (in patients with cancer). Establishing an appropriate balance between Treg cells and effector T cells is thus critical to human health and underlies the broad clinical potential for Treg cell-based therapies.

Treg cells are characterized by constitutively high expression of FOXP3 and CD25 (the IL-2 receptor α-chain; IL-2Rα). As their lineage-defining transcription factor, FOXP3 is essential for Treg cell development. The absence of functional FOXP3 from birth results in deficient Treg cell function and a disease characterized by widespread autoimmunity and allergy, known as immune dysregulation, polyendocrinopathy, enteropathy and X-linked (IPEX) syndrome2,3. FOXP3 has both transcriptional activation and repression functions; for example, it can simultaneously promote CD25 and suppress IL2 transcription, resulting in the expression of CD25 (ref. 4) and reliance on environmental IL-2 (refs. 5,6,7,8), which are both characteristic of Treg cells.

Like all other T cells, Treg cells express a T cell receptor (TCR), which allows them to detect antigens presented by major histocompatibility complex II (MHC II) on antigen-presenting cells (APCs; commonly dendritic cells or macrophages). Upon recognition of their cognate antigen–MHC on an APC, Treg cell immune modulation can be broadly categorized into three main phases (Fig. 1). First, interactions with APCs reduce their capacity to activate effector T cells. Second, activation-stimulated release of cytokines and metabolites by Treg cells diminishes the pro-inflammatory activity of surrounding immune cells. Third, this re-shaped environment favours the expansion and de novo development of new Treg cell populations, thereby promoting tolerance. Treg cell-focused therapies attempt to target one or more of these modes of action to either enhance cell function (in the case of autoimmunity, allergy or transplantation) or to suppress it (in the case of cancer). An additional therapeutic goal is to change the abundance of Treg cells to re-establish their healthy balance with effector T cells.

Fig. 1: Three phases of Treg cell-mediated immune suppression.
figure 1

a, In phase 1, regulatory T (Treg) cells suppress the ability of antigen-presenting cells (APCs) to present antigen to and co-stimulate effector T (Teff) cells. The T cell receptor (TCR) of a Treg cell forms a tight immune synapse with the antigen–major histocompatibility complex (MHC) complex displayed on the APC, physically blocking effector T cell access to the same antigen. Additionally, Treg cell CTLA4 preferentially binds to CD80 and CD86 on the APC, blocking effector T cell access to co-stimulatory signalling through the lower affinity receptor, CD28. Upon retreat from the APC, the Treg cell removes co-stimulatory proteins and antigen–MHC complexes, thereby further abrogating effector T cell activation. These Treg cell–APC interactions are thought to occur predominantly in secondary lymphoid organs (such as lymph nodes) but might also occur in tissues. b, In phase 2, Treg cells release a variety of regulatory cytokines into their microenvironment to suppress inflammation. Most notable are IL-10 and TGFβ, which suppress APCs and promote FOXP3 expression, respectively. TGFβ is cleaved from its latent form into its active form by various mechanisms, notably integrin αvβ8 on Treg cells. Treg cells express high levels of CD25, which has a higher affinity for IL-2 than the dimeric IL-2 receptor expressed by conventional T cells. Therefore, Treg cells behave as an ‘IL-2 sink’, restricting the amount available for conventional T cells. Treg cells also express the ectonucleotidases CD39 and CD73, which work together to convert ATP to adenosine, which has anti-inflammatory properties. c, In phase 3, Treg cells mediate ‘infectious tolerance’ by inducing the expansion of existing Treg cells and/or by converting effector T cells into Treg cells, establishing a long-lasting tolerogenic balance.

In this Review, we first provide a brief overview of the development and function of Treg cells, then outline the challenges associated with their use as a biomarker of immune function and discuss therapeutic approaches to boost endogenous Treg cells. We next discuss Treg cellular therapy as an exciting prospect for the treatment of autoimmunity and to induce transplant tolerance, and describe how the detrimental effects of Treg cells can be targeted to fight cancer. We highlight new, more precise Treg cell-targeting approaches that are beginning clinical testing. Although we focus on FOXP3+ Treg cells, we acknowledge that other types of suppressive T cells, such as IL-10-producing type 1 Treg cells9 and CD8+ Treg cells10, also have important roles in immune homeostasis. We review knowledge derived from the study of human Treg cells but refer to selected studies in model systems using mice or non-human primates.

The biology of human Treg cells

Treg cell development in the thymus and periphery

Treg cells arise via two major developmental pathways: selection in the thymus to generate thymic Treg (tTreg) cells (sometimes also referred to as natural Treg cells) or differentiation in the periphery from conventional T cells to generate peripheral Treg (pTreg) cells. In the thymus, Treg cell differentiation is driven by the pattern and strength of TCR and cytokine signalling11,12, with a general consensus that Treg cells arise from cells bearing ‘intermediate’ affinity TCRs with hydrophobic antigen–MHC-binding regions that bind self-antigens13. These cells with intermediate affinity TCRs bifurcate into two populations: cells receiving continuous antigen signalling develop into autoreactive, IL-2-producing effector T cells, whereas less continuous TCR signalling, for example, interrupted by TGFβ-mediated inhibition, results in cells expressing FOXP3 (ref. 14). The development of these FOXP3+ Treg cells is then locked in by IL-2 produced by their surrounding autoreactive counterparts14,15. Studies in mice suggest that most Treg cells bear TCRs that recognize common tissue antigens, rather than highly restricted tissue-specific antigens, resulting in tissue-agnostic migration patterns of the cells16. Although FOXP3 is necessary for tTreg development, ongoing expression in fully committed Treg cells might not be essential because knockout of FOXP3 does not significantly disrupt their transcriptome or methylome17. Accordingly, transcription factors other than FOXP3 (for example, TCF1 and SATB1) also have major roles in tTreg cell development, and collectively multiple proteins contribute to lineage-specific epigenetic organization18,19,20.

The development of pTreg cells has been best characterized in intestinal tissues, where cells differentiate to induce tolerance to dietary antigens and commensal microbiota. Recently, three studies reported the role of a new type of intestinal APC21,22,23, termed RORγt+ APCs, which share features of both dendritic and epithelial cells. These APCs express integrins αvβ8 and αvβ3, which convert latent TGFβ to its active form. TGFβ then converts conventional T cells into FOXP3+ Treg cells, which proceed to restrain gut-resident T cells. In humans, RORγt+ APCs are enriched in mesenteric lymph nodes23 and might be dysfunctional in inflammatory bowel disease21. Interestingly, although FOXP3 expression in pTreg cells is essential for suppression of a transcriptional programme associated with TH17 cells, commitment to the pTreg lineage is reported to be FOXP3 independent24. Nevertheless, consistent evidence that people with genetic mutations in FOXP3 suffer from colitis25 indicates that this transcription factor has a non-redundant role in maintaining intestinal tolerance.

Whether or not tTreg and pTreg cells are distinguishable in humans has been debated for many years. In mice, surface expression of neuropilin 1 (NRP1) is often used to define tTreg cells as it is not expressed by pTreg cells26 but, in humans, this receptor does not effectively identify Treg cellular origin27. Alternatively, expression of the transcription factor HELIOS (encoded by the IKZF2 gene) has been proposed as a characteristic of tTreg cells and, in humans, blood-derived T cells co-expressing high levels of FOXP3 and HELIOS are enriched with stable, highly suppressive Treg cells28.

Treg lineage stability

The stability of the Treg cell lineage and how the biology of thymic versus peripherally derived cells differs are key questions in this field. Understanding Treg cell stability is fundamental for addressing the risk of their conversion into pathogenic effector T cells as well as the longevity of therapeutic effects. The best marker of Treg cell stability is the level of CpG DNA methylation at the Treg cell-specific demethylated region (TSDR), a non-coding DNA sequence element located in the first intron of FOXP3 (ref. 29). This locus is stably demethylated in Treg cells but not in activated conventional T cells29. Of note, since FOXP3 is on the X chromosome, in females one allele is methylated due to X-inactivation, making it important to account for sex during the analysis. In the context of inflammation, some studies in mice show stable FOXP3 expression and lineage stability30, whereas others report loss of FOXP3 and gain of an effector T cell phenotype31,32. These contrasting results likely arise from differences in the lineage-tracing systems used and because FOXP3 is transiently expressed in activated effector T cells that could be present in putative Treg cell populations33.

Similar concerns have also been raised about the stability of human Treg cells, particularly in the context of Treg cell therapy products34 (discussed below). Patients with IPEX have autoreactive, TSDR-demethylated effector T cells that resemble destabilized Treg cells35. These destabilized Treg cells do not emerge in the presence of wild-type FOXP3+ Treg cells, showing that FOXP3 is important for Treg cell stability and that functional Treg cells dominantly assert tolerance in a heterogeneous population of unstable progenitors35. A consideration is that it is difficult to attribute changes in phenotype or function to true lineage instability versus effects driven by heterogeneous cell populations that include conventional T cells. Nevertheless, numerous studies in humans found that Treg cells resist lineage instability and maintain suppressive function in the presence of inflammatory cytokines, including IL-6, TNF or IL-12 (refs. 36,37). In addition, multiple preclinical models of adoptive Treg cell therapy have not revealed significant loss of tolerogenic properties38,39,40.

HELIOS expression is also associated with lineage stability, potentially via its ability to suppress IL-2, IFNγ and TNF production, as demonstrated in patients with biallelic or dominant negative IKZF2 mutations41,42. Indeed, ex vivo culture of human FOXP3+ HELIOS+ Treg cells in pro-inflammatory cytokines does not destabilize their phenotype and rather enhances their proliferation36,37. However, knockout of IKZF2 in human Treg cells does not change cell phenotype or function, suggesting that HELIOS might be a Treg stability marker but its ongoing expression is not required for fully functional cells28.

Overall, the conflicting reports regarding Treg cell stability are likely attributed to different Treg cell enrichment and tracking protocols used between studies as well as to differences between mice and humans. To date, there are no reports of a Treg cell product losing its suppressive capacity in patients, suggesting that current clinical Treg cell isolation protocols derive functionally stable products.

Mechanisms of action

Multiple mechanisms are associated with the suppression and/or promotion of tissue repair by Treg cells (Box 1). Gene knockout studies in mice support mechanistic roles for IL-2 consumption43 and the expression of CTLA4 (ref. 44), IL-10 (ref. 45) and/or TGFβ46. Below, we summarize some of the best-characterized mechanisms of action grouped into three phases (Fig. 1). More comprehensive summaries of additional mechanisms are found in recent reviews47,48. Precisely which mechanisms control immune homeostasis in different tissues and disease contexts, especially in humans, remains undefined.

Phase 1: APC modulation

Treg cells inhibit activation of effector T cells by physically blocking their access to cognate antigens and depleting co-stimulatory proteins from APCs, which primarily reside in secondary lymphoid organs, including lymph nodes (Fig. 1a). Live microscopy reveals that Treg cells form tight bonds with APCs that are presenting their cognate antigen, thereby blocking effector T cells with the same antigen specificity from accessing the APC and preventing T cell activation49. Furthermore, when Treg cells pull away from APCs they remove the antigen–MHC complex from the APC surface, further preventing effector T cell activation49. Beyond these effects on antigen presentation, Treg cells limit the ability of APCs to provide co-stimulatory signals (via CD80 and CD86) to effector T cells by expressing high levels of the coinhibitory receptor CTLA4, which outcompetes the effector T cell co-stimulatory receptor CD28 for binding to CD80 and CD86 and depletes them from the APC surface50,51. As a result, Treg cells from people with genetic mutations in CTLA4 have decreased suppressive capacity, contributing to their autoimmune phenotypes52. Moreover, combined blockade of CD28 and CTLA4 with the CTLA4–Ig fusion protein inhibits the therapeutic effect of Treg cells in a humanized mouse model of skin transplantation53.

Phase 2: microenvironment regulation

Treg cells produce multiple anti-inflammatory cytokines and metabolites, most notably TGFβ and IL-10 (Fig. 1b). TGFβ is produced by many T cells but often remains in its latent form, tethered to the cell membrane. However, Treg cells express αvβ8 integrin, which can release TGFβ from its inhibitory complex into its active form54. Activated autocrine TGFβ then maintains and promotes FOXP3 expression46,55. Treg cells also produce IL-10, which suppresses production of innate, pro-inflammatory cytokines (such as TNF, IL-6 and IL-1β) and APC function45,56. Furthermore, Treg cells produce IL-35 (Ebi3–p35 heterodimer), which diminishes inflammation and promotes Treg cell function57. An additional anti-inflammatory mechanism involves expression of the ectonucleotidases CD39 and CD73, which work together to convert extracellular ATP into immunosuppressive adenosine58,59, contributing to Treg cell suppression.

Not only do Treg cells produce anti-inflammatory mediators, they also behave as an ‘IL-2 sink’. In Treg cells, the IL-2Rα subunit (CD25) complexed with IL-2Rβ and IL-2Rγ subunits form a trimeric receptor with 100-fold higher affinity for IL-2 than the dimeric IL-2Rβ–IL-2Rγ form that is typically expressed by effector T cells60. Therefore, Treg cells deplete IL-2 from their environment and deprive conventional T cells of this essential cytokine. Experiments in mice with a Treg cell-specific deletion of CD25 revealed that Treg cell-mediated IL-2 deprivation was primarily required for suppression of CD8+ T cells43, whereas the effect on CD4+ T cells was nuanced. Specifically, only CD4+ T cells with weak TCR signalling and low IL-2 production are suppressed via this mechanism61. A therapeutic consideration is that combining Treg cell adoptive transfer with low-dose IL-2 therapy might be deleterious62 because provision of the exogenous cytokine could override the IL-2 sink effect.

Phase 3: infectious tolerance

‘Infectious tolerance’ refers to the establishment of independent, long-lasting tolerance in response to a treatment63 (Fig. 1c). The concept is that enhanced Treg cell function leads to a permanent change in the balance of Treg versus effector T cells, favouring tolerance induction. An additional angle is that enhancing Treg cells with one antigen specificity can induce infectious tolerance to a distinct antigen via a process known as ‘linked suppression’, which is mediated by local APC modulation. The notion of infectious tolerance is crucial for Treg cell therapy because it raises the possibility that a transient treatment could lead to long-lasting tolerogenic effects. Although the mechanisms by which tTreg and/or pTreg cells mediate infectious tolerance remain to be determined, the process is likely TGFβ dependent64,65 and enhanced in the absence of CD28 signalling66. In humans, it is difficult to study infectious tolerance in vivo, and therefore evidence is from studies of autoimmunity and transplantation in mice67,68,69. With more sophisticated cell tracing and gene-editing tools emerging, more research to fully understand this process is warranted.

Assessing human Treg cells in clinical studies

Identifying Treg cells

Perhaps one of the biggest challenges to the study of human Treg cells is the difficulty in tracking their numbers and function in health versus disease, or in response to therapy, due to the lack of a single definitive phenotypic marker (Fig. 2). Human Treg cells are commonly identified as CD25high, CD127 and FOXP3+ (ref. 70); however, these proteins are technically difficult to measure via flow cytometry as they have gradients of expression rather than clear positive and negative populations (Fig. 2a). Moreover, this pattern of expression is also characteristic of activated effector T cells. Thus, multiple phenotypic markers need to be assessed to increase confidence in accurate Treg cell identification. Simple approaches include co-staining for HELIOS and/or CD45RA70. CD45RA is particularly useful in separating resting (CD45RA+FOXP3low) or activated (CD45RAFOXP3high) Treg cells from conventional T cells expressing low levels of FOXP3 (CD45RAFOXP3low)71. More complex approaches, such as ascertaining the absence of IL-2 production33 and/or using gene signatures, can further help identify Treg cells. For example, nanoString or RNA sequencing has been used to identify and track changes in the transcriptomic signature of Treg cells in people with type 1 diabetes mellitus (T1DM)72 or heart transplant recipients73 (Fig. 2b).

Fig. 2: Tracking Treg cells in clinical studies.
figure 2

a, Human regulatory T (Treg) cells can be identified based on their surface expression of CD4+CD25highCD127low, intracellular expression of FOXP3 and HELIOS, and lack of IL-2 production (inhibited by FOXP3 and HELIOS). b, Stable Treg cells have demethylated DNA at the CNS2 region of FOXP3 (the Treg cell-specific demethylation region (TSDR)) and a characteristic gene signature comprised of a combination of high and low gene expression compared to effector T (Teff) cells. c, Treg cell function can be evaluated ex vivo by measuring reduced expression of co-stimulatory molecules on co-cultured antigen-presenting cells (APCs) and/or the ability to reduce Teff cell proliferation and IL-2 production. d, Antigen-specific Treg cells can be tracked with fluorescently labelled multimers or antigen-induced marker assays to quantify antigen-stimulated expression of activation proteins, including CD134 and CD137.

Measuring Treg cell antigen specificity

Treg cell therapy is thought to be most effective if it enriches the function of disease-relevant, antigen-specific cells. While TCR sequencing provides information on clonality changes, the use of multimers and/or activation-induced marker (AIM) assays can quantify antigen-specific Treg cells (Fig. 2d). Multimers are complexes of antigen–MHC conjugates that bind to antigen-specific Treg cells so they can be detected by flow cytometry. Multimers have been used to track Treg cells in T1DM, revealing that people with protective MHC haplotypes have an increased frequency of islet-specific Treg cells74. However, multimer-based methods are limited by manufacturing difficulties as well as limited knowledge of relevant antigens. Moreover, this technology is highly influenced by cell freezing75, making its use in human studies challenging.

As an alternative, many groups are turning towards AIM assays in which whole proteins or mixtures of peptides are added to blood or peripheral blood mononuclear cells, resulting in rapid upregulation of antigen-stimulated surface proteins. Antigen-specific Treg cells can be identified using a variety of markers, including CD25, CD134 (OX40) and CD137 (4-1BB), and are distinguished from conventional T cells based on the absence of CD154 (CD40L) expression76,77,78,79. With AIM assays growing in popularity76, we predict that they will increasingly be incorporated into immune monitoring for antigen-specific Treg cells.

Functional assays

Treg cells are functionally defined by their suppressive capability; their study was revolutionized by Thornton and Shevach who developed an in vitro suppression assay, which, in its original form, tested for the ability of Treg cells to prevent proliferation and IL-2 production by stimulated effector CD4+ T cells80 (Fig. 2c). Although many variations on this assay have since been developed and the assay is considered the ‘gold standard’ to assess Treg cell function, it is limited by two major caveats. First, the mechanisms of suppression measured in this assay and the assay’s physiological relevance are unclear. Experiments testing roles for TGFβ, IL-10, CTLA4 and CD39–CD73 in suppression generally show no, or only partial, effects. Second, activated effector T cells can also exhibit suppressor-like activity via unknown mechanisms81. An alternate functional assay that is gaining popularity is to co-culture Treg cells with APCs and assess the subsequent downregulation of co-stimulatory markers on the latter82 (Fig. 2c). The development of improved in vitro assays where Treg cell mechanisms of action are defined and reflect relevant in vivo effects is urgently needed. Systems such as organoid-based co-cultures83, live tissue slices84 and organs on chips83 could offer new approaches to these old issues.

Boosting endogenous Treg cells

Given that Treg cells are essential for maintaining immune homeostasis and that most hyperinflammatory diseases can, in part, be ascribed to insufficient Treg cell efficacy, there is significant interest in devising ways to increase Treg cell activity. In this section, we focus on strategies to enhance Treg cells in vivo either by harnessing natural mechanisms or by pharmacological approaches.

Harnessing natural mechanisms

Allergen immunotherapy

Allergen immunotherapy (AIT) for foreign antigens, including food and aero-antigens85,86, seeks to reverse or prevent allergic symptoms mediated by antigen-specific T helper 2 (TH2) cell immunity and enhance Treg cells by repeated, low-dose allergen exposure to restore tolerance (Table 1). AIT is particularly effective for peanut allergy, as shown in the large PALISADE trial87, where 67.2% of treated patients, compared to only 4% of placebo-treated patients, could eat peanut protein without symptoms at the end of the trial. Although the effects of AIT are presumed to be at least partly due to the induction of Treg cells, consistent evidence supporting this mechanism in humans is lacking, possibly because few studies tracked changes in antigen-specific Treg cells or considered changes in ratios of Treg cells to allergen-specific TH2 cells (Table 1). For example, the PALISADE trial did not find increased peanut-specific circulating Treg cells, although there was a decrease in peanut-specific TH2 cells87, potentially resulting in a beneficial balance between these cells. Indeed, a study of Treg cells in birch allergy found that ratios of antigen-specific Treg to TH2 cells best correlated with clinical phenotypes77. On the other hand, another study of peanut AIT did find an increase in circulating antigen-specific Treg cells using assays that showed decreased TSDR methylation and enhanced in vitro suppression88. Administration of peanut AIT in combination with omalizumab, an anti-IgE antibody that reduces allergen-triggered inflammation, also led to enhanced antigen-specific in vitro suppression by Treg cells89. Of note, continued efficacy of AIT seems to be reliant on consistent antigen exposure, suggesting that, if Treg cell-mediated tolerance is enhanced, it might not be permanently re-set for life, contrary to the theory of infectious tolerance.

Table 1 Selected clinical studies reporting effects of oral allergen-specific immunotherapy on antigen-specific Treg cells

Limitations of AIT include significant risks, side effects and patient compliance; therefore, various modifications of AIT are being explored. For example, a short regimen of allergen peptides modified to be less inflammatory increases the proportion of activated Treg cells and decreases grass allergy symptoms90. Similarly, less immunogenic mannan-coupled ‘allergoids’ that are taken up by dendritic cells and induce Treg cells91 are effective in treating patients with dust mite allergy92.

Microbiome and environment

Treg cells are strongly influenced by the microbiome and also control immune homeostasis to commensal bacteria. Therefore, there is significant interest in developing microbiota or other environmental interventions to enhance Treg cell function. As an example, allergic infants lacking a series of Clostridiales species had fewer Treg cells in their blood than healthy controls93. When these bacteria species were introduced to allergy-prone mice, there was an increase in protective Treg cells. The reverse approach is also possible, with Treg cell-promoting therapies facilitating beneficial microbiome changes. In mice, faecal microbiota transplant from animals previously treated with low-dose IL-2 to expand Treg cells offered increased protection from colitis and T1DM compared to controls94. Similarly, in a small number of patients with autoimmune disease, treatment with low-dose IL-2 promoted a Treg cell-permissive intestinal microbiome94.

Vitamin D, obtained from the diet or generated in the epidermis upon ultraviolet B light exposure, also influences Treg cells. Daylight influences the number of circulating Treg cells in healthy people, and a study conducted in the northern hemisphere showed that peak Treg cells in July and August correlated with the highest blood vitamin D levels95. A systematic review of the impact of oral vitamin D supplementation concluded that it increases the number of circulating Treg cells relative to placebo and enhances their suppressive function96. A study of more than 25,000 healthy people showed supplementation with 2,000 IU of vitamin D per day resulted in a 22% drop in the incidence of autoimmune disease compared to placebo after 5 years97. Although these studies remain correlative, they suggest that manipulation of micronutrients could be a promising approach to enhance Treg cell function.

Diet and exercise

A so-called Western lifestyle consisting of high-fat, salt and caloric intake might be a causative factor driving rising rates of autoimmunity. High levels of salt inhibit Treg cells, which might be due to effects on mitochondrial respiration98. In terms of obesity, adipose tissue in overweight mice and humans is characterized by low-grade inflammation and decreased Treg cell frequency and function relative to lean individuals99,100,101. Lean individuals fed a high-fat diet for 2 weeks had decreased Treg cell frequencies and elevated markers of inflammation in their subcutaneous fat, relative to baseline100. In a pilot trial that studied the effects of diet and the microbiome in people with multiple sclerosis, 2 weeks of intermittent fasting led to microbiome changes that were similar to those seen in mice following the same diet and to a small but significant increase in proportions of circulating Treg cells102.

In mice, exercise promotes Treg cell function by inducing expansion of the muscle Treg cell compartment, thereby limiting the production of IFNγ, an inflammatory cytokine that impairs exercise-induced performance enhancement103. Therapeutic boosting of Treg cells in mice improves insulin sensitivity in obesity99 and muscle regenerative capacity104,105. At least some of the beneficial effects of exercise on Treg cells are mediated by IL-6 released from muscle during exercise104, challenging the concept that IL-6 might destabilize Treg cells and aligning with human Treg cell-based studies showing beneficial effects of inflammatory cytokines36. Further investigation into how Treg cells promote muscle recovery might reveal relevant mechanisms that could be harnessed to treat diseases such as sarcopenia, sterile muscle injury and tocilizumab-associated muscle weakness.

Pharmacological approaches

Drawbacks to the non-pharmacological interventions discussed above include poor compliance and highly heterogeneous, non-Treg cell-specific effects. Thus, a significant area of growth is in developing traditional drug-based approaches to boost Treg cells in vivo.

Low-dose IL-2

IL-2 is a critical cytokine that is indispensable for Treg cell function and survival. Although Treg cells are unable to produce their own IL-2, they express the high-affinity trimeric IL-2 receptor (including CD25)4; therefore, multiple clinical trials have tested the effect of administering low doses of IL-2 to promote Treg cell expansion106. Treg cells with their high-affinity IL-2 receptor should outcompete CD25 effector T cells for IL-2 administered at low doses. Although low-dose IL-2 therapy is well tolerated, evidence for its efficacy is mixed. In trials for chronic graft-versus-host disease (GVHD)107 and hepatitis C virus-induced vasculitis108, more than half of patients showed improved symptoms after IL-2 therapy. Similarly, the TRANSREG study, which enroled patients with 11 different autoimmune diseases, reported decreased disease activity109. Other studies treating rheumatoid arthritis110, systemic lupus erythematosus (SLE)111,112 and T1DM113 also reported beneficial effects. In transplantation, low-dose IL-2 administered to two face transplant recipients resulted in enhanced Treg cell frequency in the skin graft and higher suppressive capacity114.

On the other hand, several studies found no benefit or detrimental effects of low-dose IL-2. The LITE trial, which aimed to decrease immunosuppression in liver allograft recipients, was halted due to a potentially increased risk of rejection with no evidence for alloreactive Treg expansion115. In the TILT study, autologous polyclonal Treg cells plus low-dose IL-2 were combined to treat new-onset T1DM but showed no metabolic benefit and increased circulation of pro-inflammatory cells62.

These heterogeneous results might be partly explained by the widely varying dosing regimens between studies, both in terms of timing and amount of cytokine. Given the short half-life and potential negative feedback pathways of the cytokine, small differences in these parameters could have major effects on outcomes116. Another consideration is that low-dose IL-2 could also expand other, non-suppressive immune cells leading to deleterious impacts. It consistently causes an increase in circulating CD56bright natural killer cells and eosinophils, although these appear transient and not harmful117. However, a study found that low-dose IL-2 increases inflammatory granzyme B+ lymphocytes and clonally expanded CD8+ T cells62, potentially fuelling hyperinflammation. Overall, the lack of a true Treg-specific effect and undesirable pharmacodynamics makes low-dose IL-2 a challenging strategy to boost endogenous Treg cells.

Modified IL-2 for Treg selective effects

Given the challenges with natural IL-2, strategies to modify IL-2 to increase its specificity for Treg cells and extend its half-life are being pursued with three general approaches. The first is to mutate the IL-2 protein so it is more selective for CD25, resulting in so-called ‘muteins’. Mutant forms of IL-2 are often combined with a second approach of adding a half-life-extending moiety. Efavaleukin alfa (previously AMG 592) is a mutated form of IL-2 with decreased binding to the IL-2Rβ subunit and increased reliance on the IL-2Rα subunit (CD25) of the IL-2R; it is also fused to an immunoglobulin Fc domain to extend its half-life. This drug was in clinical trials for ulcerative colitis, but was terminated due to meeting a prespecified futility rule (NCT04987307). An earlier trial of efavaleukin alfa in SLE (NCT03451422) showed Treg cell expansion with minimal off-target effects on conventional T cells and natural killer cells118, but clinical development was also halted due to a likelihood of it being ineffective. Similarly, development of other Fc-fusion IL-2 muteins was stopped by Roche and Bristol Myers Squibb due to a lack of efficacy in ulcerative colitis and psoriasis, respectively. Merck, Cugene and Xencor also performed trials with Fc-fusion IL-2 muteins in ulcerative colitis (NCT04924114) or healthy volunteers (NCT05328557, NCT04857866), with results pending.

Exemplifying a second approach to modify IL-2, rezpegaldesleukin (NKTR-358) consists of wild-type IL-2 conjugated to polyethylene glycol to extend its half-life. Compared to unmodified IL-2, it has improved Treg cell selectivity, a longer half-life and consistently expands Treg cells, with only a few people exhibiting CD56bright natural killer cell expansion119. A phase II study to treat SLE (NCT04433585) showed some symptom improvement120 and there is reported success in treating patients with eczema (NCT04081350) or psoriasis (NCT04119557)120.

A third approach, which is still in preclinical development, is to couple IL-2 with another moiety for increased functionality. For example, an anti-human IL-2 antibody (F5111) was converted into a single-chain antibody and complexed with human IL-2, creating a so-called ‘immunocytokine’ (licensed by Cartesian Therapeutics). This immunocytokine preferentially activates and expands Treg cells, showing efficacy in mouse models of colitis and immune-checkpoint inhibitor-induced diabetes mellitus121. In another approach, IL-2 has been fused to other beneficial cytokines. For example, IL233 is a hybrid cytokine linking IL-2 and IL-33 (licensed by Slate Bio) so that IL-2 can induce IL-33R expression on Treg cells, making them IL-33 responsive. The combined effects of IL-2 and IL-33 expand Treg cells in vivo, protect against autoimmunity and reduce inflammation122,123.

Overall, boosting Treg cells with IL-2, using either the native cytokine or various modified forms, has not resulted in the major success originally hoped for based on data from preclinical models. Apart from the continuing challenge that Treg cells are not the only cells expressing CD25, a fundamental question with this approach is whether transiently increasing Treg cells in an antigen non-specific way is sufficient to induce tolerance. The answer to this question likely depends on the disease context and patient-specific factors. More work is needed to understand how IL-2 therapy might enhance disease-relevant Treg cells and how it could be combined with other approved therapies for synergistic effects.

Nanomedicine-based therapy to boost Treg cells

Thanks to the success of the COVID-19 mRNA vaccines, there has been an explosion of interest in using biodegradable nanomedicines to target Treg cells, with most approaches seeking antigen-specific Treg cell induction or expansion124 (Table 2). In mice, nanoparticles loaded with autoantigen-encoding mRNA125 or complexed with autoimmune peptides126 prevent progression of experimental autoimmune encephalomyelitis and T1DM, respectively, leading to Treg cell expansion. Nanoparticles can also be conjugated with antibodies to target them to certain locations. For example, antigen-complexed nanoparticles targeting scavenger and mannose receptors on liver sinusoidal endothelial cells promote Treg cell expansion and subsequent suppression of airway inflammation in mice127. Moreover, nanoparticles can be loaded with Treg-promoting substances such as rapamycin128, TGFβ129 or F5111 immunocytokine130.

Table 2 Clinical-stage nanomedicines to boost Treg cells

This preclinical success has led to multiple companies entering clinical-stage testing of such nanomedicines to treat allergy and autoimmunity (Table 2). Each approach uses nanoparticles with immunomodulatory properties expected to increase Treg cell activity. For example, both Moderna and Cartesian Therapeutics have developed nanoparticles that deliver IL-2 muteins. Furthermore, rapamycin nanoparticles (ImmTOR) that reduced the development of anti-drug antibodies specific for uricase (used to treat gout)131 were shown to induce significant Treg cell expansion when combined with an IL-2 mutein132. TOPAS Therapeutics and COUR (in partnership with Takeda) are developing nanoparticles loaded with tolerogenic autoantigens that induce Treg cells to treat a variety of autoimmune disorders and allergies. Given the broad interest in nanoparticle-based therapies as well as in modulating Treg cell activity, there are likely to be increased numbers of clinical trials in this area in the future.

Treg cellular therapy

The strategies discussed above to expand Treg cells in vivo are relatively low cost and feasible but the effects could vary significantly depending on the immunological history of a patient and the environmental context. An alternate approach to bolster Treg cells is to administer them as a cell-based therapy (Fig. 3). Numerous approaches are being developed for obtaining, expanding and engineering Treg cells for optimal therapeutic application.

Fig. 3: Treg cell therapy steps and considerations.
figure 3

a, Regulatory T (Treg) cells are commonly sorted from blood, umbilical cord blood or thymus based on their expression of CD4+CD25highCD127 and, commonly, CD45RA+. Alternatively, conventional CD4+ T cells can be genetically engineered to express FOXP3. b, Treg cells can be expanded ex vivo either using a polyclonal general T cell stimulus or by antigen-presenting cells (APCs) presenting disease-relevant antigens. The latter approach also enriches Treg cells for antigen specificity. c, Treg cell antigen specificity can also be modified by genetically engineering cells to express a T cell receptor (TCR) or chimaeric antigen receptor (CAR). d, Treg cell therapies are often administered to patients in combination with existing immunosuppressive regimens. In the future, gene editing could be used to incorporate orthogonal receptors and/or remove deleterious proteins. e, Bespoke Treg cell products with optimal function, stability and persistence hold promise to treat many diseases.

Sources of natural Treg cells for therapy

Early clinical trials investigating the safety and feasibility of Treg cell therapy were focused on autologous Treg cells isolated from peripheral blood. Treg cells were enriched from peripheral blood mononuclear cells using magnetic beads to deplete CD8+ T cells and enrich CD25+ cells. However, because of the relatively low purity of Treg cells achieved with this approach, these cells must be expanded in the presence of rapamycin to limit growth of contaminating conventional T cells and yield a potent, immunosuppressive Treg cell product133. An alternate approach is to use flow cytometry-based cell sorting to obtain a purer starting population of Treg cells, with sorting typically based on a combination of CD4, CD25 and CD127. Sorting strategies can also further select naive Treg cells based on CD45RA134,135, CD226 (ref. 136) or GPA33 (ref. 137) to enrich for cells co-expressing FOXP3 and HELIOS.

In an effort to decrease the cost and complexity of Treg cell therapy, several groups are exploring the possibility of manufacturing Treg cells from allogeneic donors, with a focus on two tissues that contain a large proportion of naive Treg cells: umbilical cord blood (UCB) and thymus. Advantages of UCB Treg cells include an increased TCR repertoire diversity and lineage stability relative to blood-derived cells138,139. UCB Treg cell therapy also decreases the risk of acute GVHD140,141. Furthermore, fucosylation (the addition of a Siayl-Lewis X moiety onto P-selectin) has been used to alter the homing potential of UCB-derived Treg cells, with promising preliminary data and an ongoing phase II clinical study142. A key question is the requirement for HLA matching with these allogeneic cells. In the studies by Brunstein et al.140,141, the administered Treg cells were partially HLA matched (4–6 of 6 HLA antigens), whereas a recent study of UCB Treg cells for COVID-19-associated acute respiratory distress syndrome did not perform intentional HLA matching and did not observe an increase in anti-HLA antibodies compared to the placebo group143. UCB-derived polyclonal Treg cells developed by Cellenkos are being used to treat refractory bone marrow failure syndrome (NCT03773393). Preclinical studies are also testing UCB Treg cells in a variety of non-traditional contexts such as traumatic brain injury144 and lung inflammation145.

Thymuses are routinely removed from infants during cardiac surgery procedures and contain large numbers (0.3–3 billion) of Treg cells. Thymus-derived Treg cells are more suppressive than adult blood-derived cells146, leading to the development of protocols for their isolation and expansion that are compatible with good manufacturing practices147,148,149. Recently, the first testing of thymus-derived Treg cells was reported in a single patient, with the delivery of 20 × 106 cells/kg autologous cells 9 days after a heart transplantation. Notably, despite ongoing treatment with calcineurin inhibitors as immunosuppressants, the patient maintained Treg cells at higher levels than before the transplantation throughout the 2-year follow-up period150, suggesting this approach stably increased the Treg cell pool.

Generating therapeutic Treg cells from conventional T cells

Given the difficulties in isolating pure populations of Treg cells, another approach is to re-programme the more numerous conventional T cells into Treg cells. To increase FOXP3 expression, conventional CD4+ T cells can be cultured in the presence of rapamycin, retinoic acid and/or TGFβ to generate induced Treg (iTreg) cells, which express FOXP3 for a period of time. Therapeutic administration of iTreg cells (CD4+CD25 T cells cultured in rapamycin and TGFβ1) was shown to be safe in patients at risk of GVHD151. However, there is some doubt about the stability of iTreg cells because their levels of TSDR demethylation never approach those of ex vivo Treg cells and they do not express HELIOS152,153.

Recently, two groups generated iTreg cells via differentiation of induced pluripotent stem (iPS) cells. CD4+ T cells were first derived from iPS cells, and then FOXP3+, TSDR-demethylated Treg cells were generated by expanding the CD4+ T cells in a cocktail of CDK8 and CDK19 inhibitor, rapamycin, TGFβ, and an agonistic TNFR2 antibody154 or, as reported in a preprint, by expanding them in TGFβ plus all-trans retinoic acid155. Given that iPS cells can theoretically give rise to unlimited numbers of well-characterized cells, these methods will undoubtedly be of significant interest.

Treg cells can also be obtained by overexpressing FOXP3 in conventional T cells. To achieve the desired effect, FOXP3 must be controlled by a strong, constitutive promoter so that expression does not significantly diminish as cells enter a resting state7,156. CRISPR-based methods are also being attempted, whereby a strong promoter is inserted immediately upstream of the endogenous FOXP3 locus to drive constitutive expression157. FOXP3 manipulation can also promote Treg cell differentiation from haematopoietic stem and progenitor cells (HSPCs). Interestingly, constitutive overexpression of FOXP3 in UCB-derived CD34+ HSPCs promotes stem cell quiescence and impairs T cell differentiation158, but this can be overcome by delivering FOXP3 with its regulatory elements to recapitulate physiological, non-constitutive expression159. Further, genome engineering to co-express FOXP3 and HELIOS can generate more functional Treg cells than engineering to express FOXP3 alone160.

An outstanding question is whether overexpression of FOXP3 will fully recapitulate all the functions of natural Treg cells. So far, the functional properties of human, polyclonal conventional T cells overexpressing FOXP3 have only been tested using in vitro suppression assays or in vivo xenogeneic models of GVHD and IPEX161. T cells that constitutively express FOXP3 following lentivirus transduction are being investigated to treat IPEX (NCT05241444), and data from this study will be key for understanding the therapeutic potential of FOXP3-expressing conventional T cells as a replacement for Treg cells.

Expanding polyclonal or antigen-specific Treg cells

To acquire a clinically relevant number of cells, autologous Treg cells are usually stimulated and expanded ex vivo. Most clinical trials have used polyclonally expanded cells (Fig. 4), which have been tested in many different diseases (see Bluestone et al. for a recent comprehensive review162). Of particular interest is the ONE study, an international, multi-centre study that tested several cell therapies in living donor kidney transplantation with the goal of minimizing the requirement for immunosuppressive drugs. Patients who received Treg cell therapy did not undergo increased levels of rejection despite not being given the complete standard-of-care immunosuppressive therapy and, moreover, they had lower rates of viral infections163. As part of the ONE study, Roemhild et al. showed that most (8 out of 11) Treg-treated patients achieved stable immunosuppression with a single drug, whereas a reference group remained on standard dual or triple immunosuppressive drugs164. Interestingly, Treg cells in the blood shifted to a less diverse TCR repertoire, suggesting there could be an alloantigen-driven selection process for these cells. The ongoing TWO study (ISRCTN: 11038572) aims to confirm the benefit of polyclonal Treg cell therapy in transplantation by testing whether immunosuppressive drug treatment can be tapered to a low-dose of tacrolimus alone following polyclonal Treg cell infusion in kidney transplant recipients165. A subset of patients from this trial showed successful transition to tacrolimus monotherapy without transplant rejection166.

Fig. 4: Clinical trials of Treg cell therapy.
figure 4

Time course of the number of clinical trials investigating regulatory T (Treg) cell therapy, based on data available from ClinicalTrials.gov and EudraCT. Ongoing (striped bars) refers to the number of clinical trials that were registered as recruiting in each given year. Complete (solid bars) refers to the number of clinical trials that were recorded as complete or terminated in each given year. Trials are separated into disease areas. GVHD, graft-versus-host disease.

Clinical trials exploring the use of polyclonal Treg cells to inhibit GVHD following HSPC transplantation have also yielded promising results, with most treated patients showing clinical improvement and decreased GHVD activity167,168,169,170,171. An interesting new approach is the manipulation of the graft itself by infusing a polyclonal Treg cell-enriched product, a strategy being investigated by Orca Bio. In several studies, patients received a CD34+-selected HSPC graft supplemented with Treg cells (sorted from the graft), followed by infusion of defined numbers of donor-derived conventional T cells 2 days later167,170. Patients treated with this product had early myeloid engraftment, and low rates of GVHD and relapse compared to standard of care167,170,171. A phase III study known as Precision-T (NCT05316701), has completed enrolment with results expected in 2025. These intriguing results suggest that more research is needed to understand how Treg cells can benefit HSPC engraftment. Indeed, the bone marrow is known to be a rich source of Treg cells, yet little is known about their biology and function in this tissue.

In T1DM, there have been mixed reports about the clinical efficacy of autologous polyclonal Treg cells to preserve β-cell function. Some studies showed preservation of insulin production and reduced dependence on exogenous insulin172,173, particularly when combined with a B cell-depleting antibody (rituximab)174, whereas others failed to find clinical impacts62,175,176. These outcome variations could be due to differences in Treg doses, number of infusions or the timing of treatment with respect to disease onset.

Polyclonal Treg cells have also been sporadically tested in single individuals for the treatment of SLE177 and ulcerative colitis178, further confirming the wide applicability and excellent safety profile of this strategy. These studies noted an enrichment of Treg cells in the skin and intestine, respectively, but the low number of patients treated limits the ability to draw meaningful conclusions about therapeutic efficacy. An ongoing phase II clinical trial in amyotrophic lateral sclerosis (NCT05695521) will reveal potential utility beyond traditional immune-mediated diseases (Box 1).

There have been attempts to expand antigen-specific Treg cells using APCs ex vivo. For the majority of autoimmune conditions and inflammatory disorders, the feasibility of this expansion approach is poor as only a small number of disease-relevant Treg cells naturally reside in the periphery. However, in a transplant context, approximately 1–10% of peripheral Treg cells can be activated by donor antigens179. Consequently, ex vivo stimulation with graft donor B cells expands autologous Treg cells that predominantly recognize donor-derived antigen–MHC complexes, termed donor-alloantigen reactive Treg (darTreg) cells180. This approach has been successfully tested in three kidney transplant recipients181. However, a separate study in liver transplantation emphasized the difficulties of manufacturing clinically relevant doses using this strategy, with 44% and 33% of recruited participants yielding insufficient or only partial doses of Treg cells, respectively182. Furthermore, despite a promising safety profile from these clinical studies, data from a recent non-human primate heart transplantation model suggested that darTreg cells might not be stable as they lose signature Treg cell markers following infusion183.

Genetic engineering of Treg cells

The expansion phase of the Treg cell manufacturing protocol provides an opportunity for genetic manipulation and enhancement. Engineering can improve Treg cell function, stability, trafficking and/or persistence in vivo following adoptive transfer. Most research has focused on improving Treg cell potency by conferring antigen specificity with TCRs or chimaeric antigen receptors (CARs). Key considerations for this approach are whether overexpression of a TCR or CAR is more suitable (Table 3) and selection of the target antigen (Box 2).

Table 3 Comparison of key features of engineered TCRs versus CARs

TCR engineering

Antigen engagement and subsequent TCR signalling by Treg cells drive retention of the cells in the relevant tissue and enhanced suppression; thus, a logical approach to redirect Treg cell specificity is to introduce new TCR α-chain and β-chain genes. This idea has been explored in a variety of contexts, most notably in mouse models of T1DM184. Initial studies in human cells focused on modifying blood-derived Treg cells185, and TCR-engineered blood-derived Treg cells are now being pursued for the treatment of multiple sclerosis by Abata Therapeutics (ABA-101). More recently, TCR delivery has been combined with FOXP3 editing157 to generate islet-specific Treg cells from conventional T cells186. Mouse TCR-transgenic Foxp3-edited Treg cells were shown to home to the pancreas, stably persist, and prevent disease development in an adoptive transfer model of T1DM. This multi-editing approach is now being pursued commercially for the treatment of T1DM by GentiBio (GNTI-122), using conventional T cells engineered to express FOXP3, an islet-specific TCR and a rapamycin-activated, chemically induced IL-2 signalling complex (allowing rapamycin to drive IL-2 signalling in these cells)187.

CAR engineering

CARs are synthetic fusion proteins that typically bypass the requirement for antigen–MHC interactions through the use of an antibody-derived antigen-targeting moiety attached to TCR signalling domains188. Early studies investigating the potential of CARs to confer specificity to human Treg cells were performed in the context of transplantation, where HLA-A2 was targeted as a clinically relevant human MHC molecule that is commonly mismatched. In vivo studies of mouse or human HLA-A2-specific CAR (A2-CAR) Treg cells showed that CAR expression controlled Treg cell homing to transplanted HLA-A2+ skin or islet grafts, in turn delaying transplant rejection189,190,191.

Multiple groups also refined CAR design for optimal Treg cell function by testing various intracellular signalling domain configurations. These studies uniformly showed that CARs encoding CD28 co-stimulatory signalling domains but not the CD137 co-stimulatory domain are optimal for Treg cells82,192,193. Interestingly, in a skin transplant model, a first-generation CAR with no co-stimulation domain was also effective, revealing that CAR Treg cells respond to endogenous co-stimulation193. Overall, the potent effects of A2-CAR Treg cells led to the rapid development of good manufacturing practice-compatible protocols for CAR Treg generation39, and two clinical trials are testing the safety and efficacy of A2-CAR Treg cells in kidney (NCT04817774) and liver (NCT05234190) transplant recipients.

In the context of GVHD, the B cell antigen CD19 has been pursued as a CAR Treg target194,195 with the rationale that systemic CD19+ B cells will enable widespread CAR Treg cell stimulation. Remarkably, CD19-specific CAR Treg cells not only reduced GVHD severity but also controlled the growth of CD19+ tumour cells. Importantly, this was achieved in the absence of cytokine release syndrome, a common adverse event associated with CAR T cell therapy but not with CAR Treg cell therapy, given the latter’s relative lack of inflammatory cytokine production196. CD19-CAR Treg cells might also be suitable for the treatment of autoimmune diseases in which B cells and/or antibodies play pathological roles. Recently, FOXP3-overexpressing, CD19-CAR Treg cells reduced autoantibody generation and delayed lymphopenia in a humanized mouse model of SLE197.

A fundamental advantage of using CARs to redirect T cell specificity is the ability to engage target antigens in an MHC-independent manner (Table 3). However, in some cases, ideal antigens might be intracellular or predominantly soluble, making them difficult CAR targets. This limitation was recently overcome by generating so-called ‘TCR-like’ CARs specific for an insulin peptide presented by MHC class II. In two parallel studies, Treg cells expressing insulin-MHC-specific CARs suppressed pathogenic T cell proliferation in vitro and significantly delayed or prevented T1DM in vivo198,199.

Depending on the structure and environmental context of an antigen, it is possible to use CARs to redirect Treg cell specificity to non-membrane-bound proteins. For example, CAR Treg cells targeting the blood coagulation factor VIII are efficacious in mouse and humanized mouse models of haemophilia A200,201. More recently, CAR Treg cells targeting flagellin, the protein component of bacterial flagella, were explored as a treatment option for inflammatory bowel disease202. Flagellin was selected as it is naturally oligomeric and primarily accessible to immune cells during periods of inflammation and gastrointestinal damage. Flagellin-specific CAR expression promoted intestinal trafficking, and CAR Treg cells were significantly more suppressive and could promote intestinal epithelial cell integrity in the presence of their target antigen.

Ensuring stability of antigen receptor-engineered Treg cells

A general concern with Treg cells, particularly those modified to express antigen-specific receptors, is lineage stability. Injection of human CAR Treg cells into mice systemically expressing HLA-A2 did not reveal significant loss of FOXP3 (refs. 38,39,190). Similarly, in a non-human primate model of islet transplantation, infusion of autologous Bw6-specific CAR Treg cells into Bw6+ animals did not result in any overt toxicity despite ubiquitous target antigen expression in vivo40. Nevertheless, CAR-induced lineage changes are possible as evidenced by the loss of HELIOS (but not FOXP3) expression in mouse and human Treg cells expressing CARs encoding TNFR family co-receptors82,193.

Strategies to safeguard against instability and promote stable FOXP3 expression include expanding cells in the presence of FOXP3-promoting molecules (such as rapamycin or TGFβ) or genetic manipulation to overexpress FOXP3. For example, McGovern et al. developed a method to transduce an enriched population of Treg cells with a vector encoding FOXP3 and a myelin basic protein-specific TCR under the same promoter, restricting transgenic TCR expression to FOXP3+ cells203.

Another factor that could influence Treg cell stability is dysfunction induced by overstimulation. In studies of tonic-signalling CARs in Treg cells, chronic stimulation caused an exhausted phenotype and loss of function204 and, in another study, was associated with loss of FOXP3 and HELIOS expression (although the cells did not acquire an effector phenotype)205. This tonic signalling can be mitigated by careful selection of CAR co-stimulatory domains and, possibly, by also using CRISPR technology to insert CARs into the TCRα constant (TRAC) locus, resulting in TCR-like transcriptional control and a more physiological level of CAR expression190.

Combining engineered Treg cells with immunosuppression

A consideration yet to be explored is how to condition patients so they are ideally suited to receive engineered Treg cells. Ideally, Treg cell therapy should be integrated with existing regimens and opportunities identified to combine it with new types of less toxic immunosuppression. As examples, CAR Treg cells can work in synergy with rapamycin191, and TCR-transgenic Treg cells can be combined with low-dose IL-2 (ref. 206) or anti-CD3 (ref. 207). Ongoing work is also defining the optimal strategy to combine Treg cell therapy with anti-thymocyte globulin208 or CD28 blockade53. Development of combination therapies is a particularly important consideration in transplantation, where patients receive drug-based immunosuppression that could reduce the function of Treg cell therapy209. For example, gene editing to knock out FK506-binding protein 12 or CD52 could result in Treg cells that are resistant to tacrolimus210 or alemtuzumab (an anti-CD52 antibody)211, respectively. One can also envision bespoke CAR engineering combined with specific types of immunosuppression, for example, to deliver signals blocked by antibodies or recombinant proteins.

Other gene-editing strategies

Building on the success of TCR and CAR engineering, additional gene-editing strategies are being explored to further enhance the efficacy, survival and/or stability of adoptively transferred Treg cells. As previously discussed, IL-2 therapy is an effective strategy to bolster Treg cells. Limitations related to lack of Treg specificity can be overcome by using engineered, so-called ‘orthogonal’, systems in which synthetic versions of IL-2 and the IL-2 receptor interact with each other but not the endogenous versions212,213. Engineered expression of an orthogonal IL-2 receptor in adoptively transferred Treg cells allows selective stimulation in vivo by administration of orthogonal IL-2. The benefits of this approach have been demonstrated in mouse models of GVHD214 and heart transplantation215.

A similar approach is the use of chimaeric cytokine receptors, or switch receptors, which detect one extracellular cytokine and deliver the stimulatory signal of another. Although this approach has not yet been reported in Treg cells, a similar concept was developed whereby extracellular detection of pro-inflammatory cytokines triggered CD3ζ and CD28 signalling in Treg cells216. Treg cells expressing these so-called artificial immune receptors were significantly more effective at alleviating GVHD in a mouse model.

CAR Treg cell therapy can delay but not prevent tissue rejection191,217, suggesting the need to further enhance Treg cell-suppressive mechanisms. In an early test of this concept, A2-CAR Treg cells were genetically modified to constitutively express IL-10 (ref. 218), which is normally expressed at relatively low levels in Treg cells compared to IL-10-producing type 1 Treg cells219. A similar strategy could be used to introduce other beneficial molecules such as TGFβ188.

Removal of deleterious genes from therapeutic Treg cell products can also be achieved with gene editing. In oncology, PD1 ablation improves the efficacy of CAR T cells220,221 and, given the negative impact of PD1 signalling on Treg cells82, adapting this approach could be beneficial. This strategy could also be used to enhance the function of allogeneic Treg cells to minimize their immunogenicity. For example, in work reported in a preprint, McCallion et al.222 used a humanized mouse model of skin transplantation to show that CD8+ T cell-mediated killing of allogeneic Treg cells could be overcome by CRISPR–Cas9-mediated silencing of MHC. Moreover, expression of a non-polymorphic HLA-E–β2-microglobulin fusion protein further protected Treg cells lacking MHC expression from natural killer cell-mediated killing. Other strategies to evade natural killer cells, such as expression of HLA-E223,224,225, Siglec ligands226 or CD47 (ref. 227), are also likely to have applications in Treg cells.

Overall, it is clear that, to fully maximize the potential of Treg cell therapies, gene editing should be employed. Fortunately, the extensive work on gene editing of conventional T cells can be leveraged for Treg cells, albeit for a different purpose.

Depleting Treg cells to treat cancer

Cancer evades immunity by creating an immunosuppressive environment that promotes Treg cells. The prognostic value of Treg cell abundance in tumours varies depending on the type of cancer but, generally, a higher frequency of intratumoural Treg cells is associated with decreased overall survival (reviewed in ref. 228). Therefore, many cancer therapies aim to deplete Treg cells in order to invigorate the anticancer functions of other immune cells.

Targeted Treg cell depletion

As discussed earlier, there have been many attempts to selectively expand Treg cells based on their high expression of CD25. Targeting CD25 has also been used to deplete Treg cells in cancer. Denileukin diftitox (ONTAK) is a human IL-2 protein fused to diphtheria toxin that was tested for many years; however, Treg cell depletion was transient and, in some cases, expansion of antigen-specific effector T cells was halted229. E7777 is a new, purer IL-2–diptheria toxin fusion protein under investigation. A recent clinical trial treating patients with relapsed or refractory lymphoma showed an objective response in one-third of participants; however, half of patients experienced a serious adverse event230. A different approach tested RG6292, a non-blocking, CD25-depleting antibody that binds CD25 to trigger antibody-dependent cellular cytotoxicity of Treg cells but leaves the substrate-binding domain of CD25 open to receive IL-2 signalling on remaining effector T cells231. It exhibited preclinical efficacy without immune-related toxicities and a monotherapy clinical trial in patients with advanced solid tumours was recently concluded (NCT04158583).

Because all therapies targeting CD25 have a risk of off-target effects, several new approaches have been developed to more selectively target Treg cells. For example, AstraZeneca developed an antisense oligonucleotide targeting FOXP3 (AZD8701) that is in clinical testing (NCT04504669). Preclinical modelling showed effective knockdown of FOXP3 throughout the body, including the tumour, as well as reduced Treg cell-suppressive capacity and a parallel boost in CD8+ T cell antitumour activity232. Preclinical models have also demonstrated the efficacy of reducing intratumoural Treg cells by targeting CCR8 (refs. 233,234), a strategy now in clinical testing by Shionogi as a combination therapy with the PD1 inhibitor pembrolizumab (NCT05101070). Interestingly, the controversial relevance of NRP1 in human Treg biology has recently been re-examined, with Chuckran et al.235 finding its expression is prevalent in tumour-resident Treg cells, supporting the rationale for anti-NRP1 therapy (NCT03565445).

Combating the suppressive microenvironment

Immunosuppression is a hallmark of cancer; reducing immunosuppressive cells and/or signals can re-invigorate anticancer immunity. Cyclophosphamide, a cytotoxic alkylating agent used to treat malignancies, also selectively depletes Treg cells236. Mechanistically, Treg cells have less ATP than effector T cells and thus produce less glutathione, which detoxifies cyclophosphamide, making them more susceptible to this drug. Several clinical trials are testing CD39 and CD73 inhibitors in attempts to increase extracellular, pro-inflammatory ATP levels. One preclinical study showed that antibodies blocking CD39 and CD73 were effective in activating effector T cells and slowing tumour progression; however, there was no apparent impact on Treg cells58. Administration of oleclumab, an anti-CD73 antibody, together with an anti-PDL1 antibody (durvalumab) also increased overall response rate and progression-free survival relative to those treated with durvalumab alone237.

Tryptophan and kynurenine are metabolites that influence the balance of effector cells to Treg cells. Their relative abundance is regulated by the intracellular enzyme indoleamine 2,3-dioxygenase (IDO1), which converts tryptophan into kynurenine. The latter metabolite decreases effector T cell proliferation and survival and promotes pTreg cell differentiation. IDO1 is expressed in APCs and many tumour cells and has become an anticancer drug target. However, although there were high hopes for the IDO1 inhibitor epacadostat, it ultimately failed to have any effect in a phase III clinical trial238. Despite this disappointing result, which was possibly related to suboptimal trial design and lack of mechanistic understanding239, there are multiple new IDO1-targeting candidates in clinical trials (reviewed in ref. 240).

Galunisertib is a TGFβ type I receptor kinase small-molecule inhibitor that can reverse Treg cell suppression in vitro241. In preclinical models, it lowered Treg cell abundance and increased lymphoma-related survival242. In a phase II trial for patients with rectal cancer, galunisertib in combination with neoadjuvant chemoradiotherapy improved the complete response rate243. Interestingly, although the frequency of blood Treg cells increased with treatment, intratumoural Treg density and TGFβ signalling decreased in tumour biopsies over time243. Overall, many anticancer therapies are hypothesized to work, at least in part, via suppressing Treg cell function. Continued investigation of the impact of these drugs on Treg cells will provide mechanistic insights that could be leveraged for both pro-Treg and anti-Treg cell therapies.

ICIs and Treg cells

Immune-checkpoint inhibitors (ICIs) are now a mainstay of cancer therapy, working to prevent effector T cell exhaustion and re-invigorate anticancer immunity. For example, inhibitors of the checkpoint proteins PD1 and CTLA4 block effector T cells from receiving inhibitory signals that would otherwise prevent them from killing cancer cells. The effects of these ICIs on Treg cells are just starting to be elucidated. As with effector T cells, PD1 signalling inhibits Treg cells, reducing their proliferation and suppressive effects244,245,246. Thus, an unintended consequence of PD1 blockade can be enhanced Treg cell function. A small fraction of patients treated with a PD1 ICI develop hyper-progressive disease associated with an increased frequency of proliferating Treg cells in the blood245. Furthermore, patients who expressed PD1 on over half of their Treg cells had poor survival following ICI treatment246. Of note, the ratio of tumour-resident PD1+ CD8+ T cells to PD1+ Treg cells is predictive of clinical response to PD1 ICIs, with a higher ratio predicting favourable outcome246. Thus, in the future, identification of tumours with high PD1+ Treg cells prior to treatment might enable personalized approaches to first deplete Treg cells before administering PD1 ICIs.

CTLA4 inhibitors are another major class of ICI in clinical use. As discussed above, Treg cells express constitutively high levels of CTLA4 so it is not surprising that anti-CTLA4 antibodies inhibit them247,248. In mice, the depletion of intratumoural Treg cells by CTLA4 ICIs required the presence of tumour macrophages expressing the IgG Fc receptor FcγRIV247. The human homologue of FcγRIV is FcγRIIIA and, in vitro, FcγRIIIA+ monocytes mediate the antibody-dependent cell cytotoxicity of CTLA4 ICI-treated Treg cells248. Interestingly, the abundance of these monocytes is higher in tumours of CTLA4 ICI responders than in non-responders, suggesting that Treg cell depletion contributes to clinical benefit. However, results are overall inconsistent, leading to the conclusion that the effects of CTLA4 ICIs on Treg cells vary due to tumour type and time of sampling relative to ICI treatment248,249,250.

Many patients treated with ICIs experience immune-related adverse events (IRAEs). These events vary in severity and are essentially due to localized and, in some cases, antigen-specific inflammation251. Whether Treg cells are implicated in IRAEs is unclear. In a study of ICI-induced colitis in patients with melanoma, single-cell sequencing analysis of the colon paradoxically found increased frequencies of Treg cells in the patients with colitis compared with ICI-treated patients without colitis252. However, these Treg cells were skewed towards an inflammatory and potentially unstable phenotype, perhaps, as the authors suggest, in response to local inflammation252. Interestingly, the risk of IRAEs is increased by a history of autoimmune disease or use of a CTLA4 ICI253, suggesting that Treg cell depletion or destabilization (which would be stronger with a CTLA4 ICI) could tip the balance of tolerance in healthy tissues to become permissive of an inflammatory response. In accordance, IRAE incidence positively correlates with response rate and survival254, suggesting that a brief loss of tolerance might be required to allow anticancer immunity to function.

Conclusions and future directions

Treg cells are undoubtedly a cornerstone of a healthy immune system; however, tracking their contribution to disease and harnessing their full therapeutic potential has remained somewhat elusive. Major limitations in tracking Treg cells as biomarkers have included the lack of FOXP3 specificity and challenges in quantifying antigen-specific cells. Tools to more accurately identify Treg cells, such as combining FOXP3 and HELIOS staining, measuring TSDR methylation, and gene signatures, can now be routinely applied to better discriminate between Treg and conventional T cells. In terms of specificity, AIM assays and a growing repertoire of multimers provide new ways to follow antigen-specific cells (both in blood and other tissues) so that changes in disease-relevant cells can be more accurately quantified. Early use of such tools in allergy and autoimmunity clearly shows that changes in antigen-specific Treg cells are relevant for human disease74,77 and malleable in response to therapy (Table 1).

In terms of therapeutic targeting to increase Treg cell activity, the past two decades have focused on in vivo modulation using relatively unspecific tools or non-antigen-specific adoptive cell therapy. These studies have hinted that it is possible to enhance tolerance and reduce reliance on non-specific immunosuppression. The field is now evolving to address challenges and to create more specific and effective approaches (Fig. 5). For adoptive Treg cell therapy, although few phase II trials powered to measure efficacy have yet to be completed, it is a significant advance that polyclonal and darTreg cells can be used to replace or reduce immunosuppressive drugs in the context of organ transplantation163,181. Even if Treg cell therapy does not induce long-term tolerance, the ability to reduce toxic immunosuppression would have a major impact on quality of life and morbidity. The potential to expand the application of polyclonal Treg cell therapy to a variety of inflammatory diseases is also a very exciting new direction (Box 1).

Fig. 5: Evolution of Treg therapies.
figure 5

Over the last 10 years, regulatory T (Treg) cell-directed therapies have primarily focused on in vivo (red) or polyclonal adoptive cell therapy-based ex vivo approaches (blue) to enhance disease-relevant function. These first-generation approaches are consistently safe but there are significant current challenges such as a lack of antigen specificity and poor control over the ultimate effects. The field is rapidly moving to next-generation approaches focused on more precise and specific Treg cell targeting using protein engineering and nanomedicines for in vivo targeting and gene engineering for optimal adoptive cell therapy approaches. CAR, chimaeric antigen receptor; TCR, T cell receptor.

However, the most exciting potential lies in the evolution of methods to induce antigen-specific Treg cells either directly in vivo or through adoptive cell transfer. Nanomedicine-based approaches represent a precision approach to tolerance induction; one could envision targeting nanomedicines specifically to Treg cells, for example, through antibody-mediated targeting255 or mRNA-mediated or microRNA-mediated control of Treg cell-specific expression256. These approaches have the potential to be significantly more potent than approaches such as AIT, yet still rely on the patient’s own immune system. Delivery of engineered Treg cells by adoptive transfer offers an even more precise approach and we can foresee that, similarly to T cell therapy for cancer, cells that have been progressively more engineered will be tested over the next decade.

Tolerance is undoubtedly a balance and there is a significant opportunity to apply lessons learned from cancer to autoimmunity and transplantation and vice versa. Understanding the cellular mechanisms underlying the side effects of various ICIs has the potential to reveal significant insight into the biology of human Treg cells and hence pathways that can be harnessed for tolerance.