Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2017 May 25;189(2):197–210. doi: 10.1111/cei.12978

Regulatory T cells: tolerance induction in solid organ transplantation

T Vaikunthanathan 1,, N Safinia 1, D Boardman 1, R I Lechler 1, G Lombardi 1,
PMCID: PMC5508315  PMID: 28422316

Summary

The concept of regulatory T cell (Treg) therapy in transplantation is now a reality. Significant advances in science and technology have enabled us to isolate human Tregs, expand them to clinically relevant numbers and infuse them into human transplant recipients. With several Phase I/II trials under way investigating Treg safety and efficacy it is now more crucial than ever to understand their complex biology. However, our journey is by no means complete; results from these trials will undoubtedly provoke both further knowledge and enquiry which, alongside evolving science, will continue to drive the optimization of Treg therapy in the pursuit of transplantation tolerance. In this review we will summarize current knowledge of Treg biology, explore novel technologies in the setting of Treg immunotherapy and address key prerequisites surrounding the clinical application of Tregs in transplantation.

Keywords: cell therapy, clinical trials, immunotherapy, regulatory T cells, tolerance, transplantation

OTHER ARTICLES PUBLISHED IN THIS REVIEW SERIES

Immune tolerance in transplantation. Clinical and Experimental Immunology 2017, 189: 133–4.

Transplantation tolerance: the big picture. Where do we stand, where should we go? Clinical and Experimental Immunology 2017, 189: 135–7.

Operational tolerance in kidney transplantation and associated biomarkers. Clinical and Experimental Immunology 2017, 189: 138–57.

Immune monitoring as prerequisite for transplantation tolerance trials. Clinical and Experimental Immunology 2017, 189: 158–70.

Transplantation tolerance: don't forget about the B cells. Clinical and Experimental Immunology 2017, 189: 171–80.

Murine models of transplantation tolerance through mixed chimerism: advances and roadblocks. Clinical and Experimental Immunology 2017, 189: 181–9.

Chimerism‐based tolerance in organ transplantation: preclinical and clinical studies. Clinical and Experimental Immunology 2017, 189: 190–6.

Introduction

Innovations in surgery and therapeutics have led to remarkable advancements in solid organ transplantation. Despite this, allotransplantation is threatened by the prospect of graft rejection. While there have been improvements in averting acute rejection and supporting short‐term graft survival, there has been an accelerated rise in morbidity and mortality associated with the relative toxicity of immunosuppression regimens and chronic rejection 1, 2, 3, 4, 5. With a growing transplant waiting‐list, means by which we can ensure the prosperity of a transplanted organ are highly sought after.

A proposed panacea to this issue has been the induction of immunological tolerance: a concept centred on the manipulation of immune cells so as to design an immunological environment favouring regulation, supporting graft survival, without interfering with crucial immune surveillance mechanisms and, concurrently, abolishing immunosuppressant‐related toxicity. Indeed, there have been reports in the literature of a state of spontaneous operational tolerance in liver and renal transplant recipients 6, 7, 8. In such cases individuals forgo the need for therapeutic immunosuppressants, while synchronously maintaining normal graft function. However, this phenomenon is rare, and in the setting of liver transplantation has been shown to occur late after transplantation 9. Furthermore, ongoing immunosuppressant therapy has been thought to hinder the development of operational tolerance through its indiscriminate clearance of immune cells thought necessary for tolerance induction. Importantly, however, research has suggested that operational tolerance can be predicted before the cessation of immunosuppression through the detection of distinct immune signatures and clinical parameters, which will prove invaluable when devising future tolerance‐inducing protocols 6, 8, 10.

In the ongoing pursuit for the Holy Grail of transplant immunology, considerable effort has been directed on the search for regulatory immune cells. During the past few decades there have been countless discoveries of immunoregulatory cells spanning all lineages of immunity; arguably, however, one particular family that has received an abundance of attention has been regulatory T cells (Tregs).

Tregs are known for their importance in mediating immune tolerance to self‐antigens with their impairment linked to the development of organ‐specific autoimmune diseases, including type 1 diabetes (T1D) mellitus, autoimmune hepatitis and various haematological disorders 11, 12.

In the setting of transplantation, these cells have undoubtedly proved their worth. Animal models conducted from the point of their discovery have demonstrated their abilities to promote transplantation tolerance 11, 13, 14, 15. Furthermore, studies investigating the phenomenon of operational tolerance in liver transplantation have revealed that operationally tolerant individuals exhibited a significantly higher number of circulating and intrahepatic Tregs compared to non‐tolerant or healthy individuals 16. In addition, in both kidney and lung transplantation recent studies have shown how stable graft function is associated with increased circulating and intra‐organ Tregs 17, 18.

Having alluded to their merit in the field of tolerance induction, the biology of Tregs has come under intense investigation, with the aim of translating their apparent tolerogenic facets through to the clinic. In this review we will summarize current knowledge on these cells, prerequisites surrounding their clinical application in solid organ transplantation and recent advancements in the field.

Regulatory T cells

Phenotype

Tregs exist as 5–10% of circulating CD4+ cells and are characterized by their high and stable expression of the interleukin (IL)‐2 receptor α chain, CD25, and the master transcription factor forkhead box protein 3 (FoxP3), crucial to their development and maintenance of function. In humans FoxP3 exists in two isoforms: the full‐length protein, consisting of 11 exons and a shorter isoform which lacks exon 2, both of which are expressed at equivalent levels 19, 20. Mutations in the FoxP3 gene have been linked with the development of immune dysregulation, polyendocrinopathy, enteropathy and X‐linked (IPEX) syndrome, a multi‐system autoimmune disease affecting the intestines, endocrine gland and the skin 21, 22. While FoxP3 expression in mouse Tregs is confined purely to the Treg compartment, unfortunately in humans its expression is less faithful. It has been reported that CD4+CD25 T effector (Teff) cells can express the transcription factor transiently without displaying signs of regulation 23. Furthermore, its intracellular location makes the identification of Tregs through this marker unfeasible, requiring cell permeabilization to confirm its expression. As a result, until recently Tregs were grossly identified as CD4+CD25hi cells.

More recent work has identified an inverse relationship between the surface antigen CD127, the α chain of the IL‐7 receptor, and FoxP3 expression 24, 25, with data demonstrating refined suppressor potency in CD4+CD25+CD127low Tregs compared to Tregs classified by CD4+CD25+ expression alone 24, 26, 27. As such, demarcation of a bona fide population of human Tregs is now based on the markers CD4+CD25+CD127low.

Miyara et al. compartmentalized Tregs further into three main subsets based on their expression of CD45RA and FoxP3. CD45RA is deemed to demarcate naive suppressive FoxP3+ cells (CD45RA+) present in the cord blood and in adult blood. Tregs were classified into: CD45RA+FoxP3high resting Tregs (rTregs), CD45RAFoxP3high activated Tregs (aTregs) and CD45RAFoxP3low non‐suppressive Tregs. With regard to the latter population of Tregs described by Miyara et al., their initial ‘non‐suppressive’ identity has come under scrutiny. While data have corroborated their potential to differentiate into IL‐17‐producing cells 28 in association with CD161 expression, findings from our laboratory and others have suggested that this population do indeed possess suppressor capabilities in vitro 29, 30.

The phenotypic and functional diversity of human Tregs has been highlighted further by Duhen et al., who identified distinct populations of Tregs cells in human blood based on the expression of chemokine receptors. Additional analysis revealed that while each population was functionally suppressive they displayed differing expressions of lineage specific transcription factors and an array of cytokine production 31.

Tregs have also been classified in accordance with their origin: naive thymus‐derived Tregs (tTregs), peripheral antigenically stimulated Tregs (pTregs) and in‐vitro‐driven iTregs. While in mice the expression of neuropilin‐1 on tTregs can differentiate between pTregs, in human counterparts this marker has not yet been identified 32, 33. More recently, the expression of the Ikaros transcription factor, Helios, on tTregs has now potentially allowed for the phenotypical distinction between the two populations; however, its validity remains contentious 34, 35

To date, the only reliable way to distinguish between the subsets is through the analysis of epigenetic DNA methylation levels on the non‐coding conserved region of the FoxP3 gene termed the Treg‐specific demethylation region (TSDR) 36. In conventional Teff cells this region is fully methylated. tTregs, however, exhibit a fully demethylated TSDR, whereas pTregs only a partially demethylated region 36, 37, 38.

Indeed, these identified Treg populations are by no means comprehensive 39. With the progression of technology we have discovered that Tregs are, in fact, far more heterogeneous than we imagined initially. The dawn of mass cytometry has brought with it the potential to characterize Treg populations based on more than 40 different markers 40, 41. Here, by using cytometry time‐of‐flight (CyTOF) technology, where each cell is analysed through a multi‐dimensional deep phenotype, the complexity of the Treg subsets has been laid bare. Our recent work has highlighted the intricacy of the Treg compartment in healthy individuals, revealing 22 distinct subpopulations 42. Furthermore, recent work has proposed that results from this intense phenotypical interrogation can predict novel biomarkers for determining treatment success. Following CyTOF immune phenotyping of Tregs, Kordasti et al. were able to identify an immune signature in individuals with aplastic anaemia, which predicted the response to immunosuppressive therapy 43.

Mechanism of action

Tregs have been reported to exert their suppressive function through several different means (Fig. 1); indeed, the most reliable way of classifying Tregs would be through their function. Unfortunately, however, the expression of various phenotypical markers does not always correspond with the method of suppression utilized by a specific Treg. With regard to the specific mode of suppression, Tregs have been described to possess several cell‐contact‐dependent and ‐independent mechanisms, ranging from cytokine release, receptor endocytosis and purinergic signalling to cell cytotoxic mechanisms (Fig. 1).

Figure 1.

Figure 1

Mechanism of regulatory T cell (Treg) suppression. Contact‐independent: (a) anti‐inflammatory cytokine production. The secretion of anti‐inflammatory cytokines such as interleukin (IL)‐10, IL‐35 and transforming growth factor (TGF)‐ β has been linked with inhibition of T cell activation in vivo. (b) Exosomes: transfer of miRNA through extracellular vesicles of endosomal membranes can silence specific genes in T cells preventing cytokine production/proliferation. Contact‐dependent: modulation of antigen‐presenting cell (APC) maturation and function. The interaction of cytotoxic T lymphocyte antigen (CTLA)‐4 on Tregs with its ligand CD80/86 on APCs, delivers a negative signal for T cell activation. CTLA‐4's mechanism of action is varied, including: the capture of its APC expressed ligands and subsequent trans‐endocytosis and also the up‐regulation of indoleamine 2, 3‐dioxygenase (IDO) and the generation of kynurenines. Induction of apoptosis. Tregs have the capacity to induce apoptosis directly via granzyme A/B and perforin, inducible cyclic adenosine monophosphate (cAMP) early repressor (TRAIL), the Fas/Fas‐ligand pathway, the galectin‐9/T cell immunoglobulin and mucin domain‐3 (TIM‐3) pathway. Disruption of metabolic pathways. The ectoenzyme CD39, expressed on Tregs, result in the metabolism of adenosine triphosphate (ATP) to adenosine monophosphate (AMP), in turn producing the immunoregulatory purine, adenosine. Tregs have also been found to express high levels of intracellular cAMP. This is transferred to T effector cells through gap junctions, which leads to the up‐regulation of inducible cAMP early repressor (ICER) and in turn the inhibition nuclear factor of activated T cells (NFAT) and IL‐2 transcription leading to apoptosis by IL‐2 deprivation. High expression of IL‐2 receptor alpha chain, CD25, depletes surrounding IL‐2. DC = dendritic cells.

More recently, the role of exosomes in immunoregulation has been described. Exosomes are extracellular vesicles formed from endosomal membrane that carry various proteins, cytokines and microRNA 44, 45. Our laboratory first described their release from murine Tregs following activation 46. Furthermore, the ectonucleotidase CD73 was shown to play a significant role in mediating exosome‐related suppression through the purinergic generation of adenosine 46. Additionally, the transfer of miRNA in mechanisms of Treg suppression has been described. Here, Treg‐derived miRNA, in particular Let‐7d, transported in exosomes, have been shown to prevent T effector cell proliferation and cytokine secretion through specific gene silencing 47.

While no single predominant mechanism of suppression has been identified as of yet, it could be probable that Treg suppression is, in fact, reliant upon several mechanisms operating at once.

Treg clinical use: manufacture and considerations

Treg isolation

The majority of clinical trials to date have looked to use adult peripheral blood as their source of Tregs. However, there have been studies investigating Treg isolation from umbilical cord blood (UBC). It has been reported that approximately 5 − 7·5 ×106 Tregs can be isolated from one unit of UBC 48. Furthermore, CD25high Tregs are purified more readily from this source, and in addition are virtually devoid of CD25+ antigen‐experienced memory T cells 49, 50, 51.

More recently, Treg isolation from discarded paediatric thymuses, resected routinely following cardiac surgery, has revealed a novel source for Treg isolation 52. Tregs isolated from this source are in utter abundance. Here, 1 g of thymus yields Tregs in the order of 500 : 1 in comparison to 1 ml of peripheral blood. These isolated Tregs demonstrate stable properties and long telomeres, correlated with improved replicative capabilities, functional potency and in‐vivo survival. This source may prove invaluable in future trials of Treg therapy, in the paediatric population in particular, given trials faced with isolated Tregs from blood/UBC 52.

In order for the translation of Treg therapy through to the clinic, protocols outlining the manufacture of Tregs need to be in place that comply with good manufacturing practice (GMP). Because of the wealth of markers defining different populations of Tregs, much debate has been centred upon the chosen markers for Treg isolation. Until only recently Treg isolation for cell therapy has been limited to using the CliniMACs (Miltenyi Biotec, Bisley, UK) system, based on the selection of Tregs through a two‐step magnetic bead isolation. Methods have involved initial depletion of CD8+/CD19+ cells, followed subsequently by CD25 positive selection 53. However, this technique does not allow for Treg selection based on multiple parameters, limiting its use for selection of Tregs with specific characteristics. Furthermore, this method is indiscriminate when it comes to selecting markers with broad expression patterns, and with the advent of the CyTOF system 42, 43 it may well be that disease‐specific optimal Tregs will be identified, with the potential for cell therapy application.

The concept of fluorescence‐activated cell sorting (FACS) has been acknowledged widely for many decades. However, it is only recently that this method of cell isolation has been deemed GMP compliant in the United Kingdom. FACS allows for cell sorting whereby each cell is interrogated on an individual level following fluorescent labelling. This method permits cell isolation based on several parameters. Because of its recent GMP accreditation it now opens up the possibility of Treg isolation based on the highly researched markers of suppression, stability and specificity 54. While the concept of FACS isolation is shared, GMP‐certified machines used for this process of Treg isolation differ around the world. Both the United States and Poland use the BD FACSAria™, Germany uses the BD Influx™ and the United Kingdom plans to use the MACSQuant® Tyto, which is currently under validation.

One concern with isolating Tregs based on more stringent markers is the risk of obtaining poorer yields. Indeed, it has been hypothesized that sorting Tregs based on the high expression of CD25 will be too restrictive when considering the yield of cells required for ex‐vivo expansion. Putnam et al., however, devised a protocol for the FACS‐based isolation of CD4+CD25+CD127low Tregs using the BD FACSAria II 55. Sorting based on these selected markers allowed for a greater overall yield through encompassing the spectrum of CD25‐expressing Tregs, and ensured the isolation of a pure Treg population, > 95% FoxP3, through the inclusion of the marker CD127. Additionally, studies have highlighted the inclusion of isolating cells on CD45RA expression, demonstrating the superior purity and suppressive capabilities of CD45RA+ Tregs following isolation and culture 55, 56. However, one concern regarding this population is its waning with age 28. A recent study by our laboratory, however, demonstrated the feasibility of isolating and expanding this CD4+CD25+CD127lowCD45RA+Treg population in patients with inflammatory bowel disease, reiterating their superior function and stability 57. This has led subsequently to the planning of a clinical trial of Treg immunotherapy in the setting of Crohn's disease at King's College London (TRIBUTE).

Treg expansion

Tregs exist in relative paucity in the peripheral blood. The rationale behind tolerance induction stands to tip the balance in favour of regulation by the in‐vivo increase in Treg numbers over Teffs. Extrapolated data from mouse models, where Tregs have been co‐infused with Teff to determine efficacious ratios for tolerance, have suggested anywhere between 1 : 2–5 : 1, Tregs : Teff 58, 59, 60. Therefore, where Tregs currently exist at 5–10% of circulating CD4+ T cells it has been suggested that this Treg pool needs to be increased by a minimum of 33% to prevent transplant rejection 61. This requires the substantial expansion of the Treg pool for clinical efficacy; as such, the feasibility of adoptive cell therapy is reliant upon protocols for the ex‐vivo expansion of Tregs to numbers needed for their clinical application.

Tregs can be expanded in vitro using polyclonal stimulation with bead‐bound or soluble anti‐CD3 and anti‐CD28 monoclonal antibodies concomitantly with high‐dose IL‐2 55, 62. To date, the GMP‐compatible protocols have been reliant upon the CliniMACS‐based isolation of the Tregs, the aforementioned of which can often be contaminated with Teff cells. In these culture conditions Teffs will thrive in competition, leading to contamination of the final product. FACS‐sorting the starting product would circumvent this concern. However, there have been reports that even when starting with a highly pure population of Tregs repeated stimulation results in the loss of FoxP3 expression 63, 64, yet simply reducing the rounds of stimulation can often lead to insufficient overall Treg yields 65. We and others have developed Treg expansion protocols which ensure the purity of the final product, reaching clinically applicable numbers 62, 66, 67

Optimization of culture conditions has included the addition of the mammalian target of rapamycin (mTOR) inhibitor, rapamycin 68. This immunosuppressant has been shown to preferentially inhibit the proliferation and function of Teffs, as a result favouring Treg expansion and stability, permitting the growth of Tregs even from a mixed starting population 62, 69, 70. In our recent publications we set to isolate and expand Tregs from patients with end‐stage renal and liver disease, with the aim of using these cells for adoptive immunotherapy post‐transplantation. Tregs were isolated by adopting the CliniMACS separation technique and the expansion of the cells for 36 days, in the presence of rapamycin, in the GMP Clinical Research Facility at Guy's Hospital. At final harvest we reported Treg purities of more than 95%, reaching clinically applicable numbers for use in adoptive cell therapy and, importantly, showed the stability of the final Treg product 62, 67. The clinical translation of this to date has seen 12 and five patients, respectively, having received the final product post‐renal/liver transplantation as part of the two‐dose escalation studies of Treg adoptive transfer, ONE study (NCT02129881) and ThRIL (NCT02166177).

Stability

There is now growing evidence that Tregs are plastic with a potential to convert into proinflammatory cells 71. In agreement, the presence of a population of FoxP3+ T cells capable of secreting the proinflammatory cytokine, IL‐17, has been observed in human peripheral blood, thus questioning the stability of these cells. In this regard, the importance of assessing Treg stability has been proposed, especially in view of the clinical application of these cells.

Despite this strict government of FoxP3 expression, emerging data suggest that Tregs can down‐regulate FoxP3 in the presence of inflammatory cytokines. In agreement, Yang et al. have shown that exposure of Tregs to IL‐6 and IL‐1 in vitro results in the expression of IL‐17 72. To determine the origin of the unstable Tregs, the extrinsic factors resulting in their instability and the fate of these ‘exTregs’ is of particular importance.

In this regard, Hori et al. proposed a ‘heterogeneity model’, suggesting that FoxP3 expression does not necessarily determine Treg lineage commitment. They showed that uncommitted FoxP3‐positive cells can lose FoxP3 expression, acquiring transient activation‐induced FoxP3 expression, converting to effector‐like ex‐FoxP3 cells under inflammatory conditions or committing to a Treg fate (upon demethylation of TSDR) 73.

Of note, however, it is important to make the distinction between Treg instability and functional specialization. Studies have also shown that while under inflammatory conditions Tregs can acquire the ability to produce effector cytokines while still maintaining high FoxP3 expression and suppressive activity. In support of this, it has been demonstrated that IL‐17 is produced by a subset of highly suppressive human Tregs that express CCR6, the chemokine receptor used by Th17 cells for their recruitment to sites of inflammation 74.

Moreover, our group and others have shown the expression of CD161, the killer cell lectin‐like receptor subfamily B, to be expressed on a subpopulation of human Tregs, that produce IL‐17 upon in‐vitro activation in the presence of IL‐1β, but not IL‐6. In addition, evidence has supported the suppressive capacity of these cells 30, 75.

These studies highlight that Treg expression of effector cytokines alone cannot be viewed simply as a marker of plasticity or lack of stability, but may be a hallmark of their functional specialization. In support of this, Campbell et al. have shown that Treg expression of transcription factors and cytokines specific for Th1, Th2 and Th17 enables them to control inflammation mediated by Th1, Th2 and Th17 cells by responding to the same environmental cues 76.

As the function of Tregs is highly dependent upon the constitutively high expression of FoxP3 77, many groups have sought to find ways to stabilize its expression. As discussed above, epigenetic modification of the FOXP3 locus has a major role in controlling FOXP3 transcription, with demethylation of key regions correlated with suppressive function and lineage stability 78. In this regard, in‐vitro treatment with demethylating agents such as azacytidine have shown to promote the stability of FoxP3 expression in Tregs, resulting in the potent ability of these treated cells to protect from graft‐versus‐host disease (GVHD) 79. In addition, a recent Phase I trial has shown that patients with acute myeloid leukaemia treated with azacytidine immediately after allogeneic stem cell transplantation had a higher proportion of Tregs compared to time‐matched controls 80.

FoxP3 levels are regulated not only through transcriptional control, but also through post‐translational modifications. In the context of transplantation, most work has focused upon acetylation of lysine residues, which is known to stabilize the FoxP3 protein 81, 82. It has been shown that inhibiting deacetylation with histone deacetylase (HDAC) inhibitors or genetically removing Sirtuin‐1, a histone and protein deacetylase, leads to an improvement in Treg function and stability, leading ultimately to improved allograft survival 83. Thus, future directions of adoptive Treg cell therapy will necessitate further understanding of factors that cause Tregs to lose FoxP3 expression and ways to stabilize its expression.

Dosing

It is important to note that despite protocols designed for the expansion of Tregs, there are limited data on the optimal Treg dose for use in clinical trials of Treg adoptive cell therapy. The majority of trials have focused initially upon the safety of Treg cell therapy and are infusing cells at a much lower number than is likely to be clinically relevant.

Several predisposing factors must be taken into consideration when calculating the optimal dose. When looking to tip the balance in favour of regulation, the initial starting population of effector cells must also be considered, and therefore assess the current and predicted future of immunocompetency in the recipient. In the absence of lymphodepletion it is postulated that a dose of 49–79 × 109 Tregs will be needed to increase the Treg pool to clinically efficacious numbers 61. However, in concert with immunosuppressive agents such as anti‐thymocyte globulin (ATG), it is suggested that a single dose of 3–5 × 109 of Tregs will serve to boost the Treg percentage to > 33% 61. Furthermore, in association with the targeted effect of antigen‐specific cells, it has been suggested that only 1/100th–1/10th antigen‐specific Tregs (described in detail in the next section) are needed to achieve the same efficacy as polyclonal Tregs, further reducing the estimated clinically efficacious Treg dose, although it is important to bear in mind that these extrapolations have been made from mice. At this point, the Treg dose postulated to be efficacious in humans is unknown; as current clinical trials demonstrate the safety of Treg therapy at increasing doses it is only a matter of time before we can begin to determine Treg clinical efficacy accurately.

Antigen specificity

Although recent clinical trials have demonstrated the safety of Treg therapy in a variety of contexts 51, 64, 65, 84, 85, 86, convincing data for the efficacy and therapeutic benefits of this treatment option remain to be obtained. With half of BMT recipients dying from GVHD, despite Treg treatment 84, 85, and a limited efficacy being observed in T1D patients 86, it is clear that strategies to increase the potency of these cells are required. One approach is to use Tregs with a given antigen specificity 15, 87, 88, 89, 90, 91, 92; namely, in a transplant context, using Tregs with direct or indirect allospecificity.

In this context it is important to discuss briefly the pathways used by Tregs in the recognition of alloantigens, which will inevitably inform more directed Treg therapy in this setting. Figure 2 depicts the three pathways of allorecognition; direct, the indirect and the semidirect.

Figure 2.

Figure 2

Direct, indirect and semidirect pathway of allorecognition. Direct pathway: in the direct pathway, intact major histocompatibility complex (MHC) on donor antigen‐presenting cells (APCs) is recognized directly by recipient CD4+ and CD8+ T cells. Indirect pathway: the indirect pathway is characterized by recipient APC uptake of allogeneic donor MHC that has been shed through apoptosis or necrosis. This is then processed, resulting in presentation of donor antigens in the context of recipient MHC class II to recipient CD4+ T cells. Semidirect pathway: semidirect allorecognition results from the transfer of cellular membrane components, including intact donor MHC, from donor APCs to recipient APCs. This process may occur through mechanisms such as cell–cell contact or through the transfer of donor exosomes that fuse with recipient APC cell membranes. Recipient APCs are then chimeric for MHC, and are able to stimulate both CD4+ and CD8+ recipient T cells.

One of the first studies to investigate the therapeutic potential of allospecific Treg therapy was performed by Taylor et al. in 2002 88. In this study, it was demonstrated that murine Tregs with direct allospecificity could be enriched from a polyclonal population of Tregs by stimulating the cells with allogeneic splenocytes and expanding these cells ex vivo. The enhanced protective capacity of these cells, compared to Tregs expanded using anti‐CD3, was demonstrated using a GVHD model. These findings were subsequently investigated further by our laboratory in various rodent solid‐tissue transplant models 89, 90. More recently, we highlighted the translational potential of this approach using a humanized mouse model in which human Tregs with direct allospecificity were generated by stimulating polyclonal cells with allogeneic DCs 92 or B cells 91. In both studies, Tregs with direct allospecificity were shown to protect human skin xenografts from alloimmune‐mediated injury more effectively than polyclonal Tregs. The success of these studies, combined with the recent development of GMP‐compatible protocols for expanding Tregs with direct allospecificity ex vivo 93, 94, 95, has facilitated the approval of clinical trials which aim to assess the safety and efficacy of these cells in kidney (NCT02711826 and NCT02244801) and liver (NCT01624077, NCT02188719 and NCT02474199) transplant recipients.

Enriching Tregs with direct allospecificity using a preferential expansion approach is due possibly to the disproportionately high precursor frequency with which these cells exist in individuals 96. Conversely, the preferential expansion of Tregs with indirect allospecificity has proved more challenging due to the low frequency of these cells naturally present in the periphery prior to a transplant procedure 97, 98. An alternative approach which may be adopted to obtain antigen‐specific Tregs is to confer specificity through genetic modification. Indeed, the concept of using α and β T cell receptor (TCR) chain gene‐modified antigen‐specific Tregs has been investigated on numerous occasions for the treatment of various autoimmune conditions 99, 100, 101, 102, 103, 104, as well as GVHD 88, 105 and solid‐tissue transplant rejection 58, 89, 90, 91, 92. In the case of the latter, we and others employed a TCR transduction approach to generate murine Tregs with dual direct and indirect allospecificity 15, 89, 90. Interestingly, our results demonstrated that in a fully mismatched heart transplant model (BALB/c → B6), multiple injections of Tregs with direct allospecificity alone were insufficient at inducing indefinite survival of an allograft. However, only Tregs with dual specificity protected the heart allografts from chronic vasculopathy, with maintenance of myocardial architecture and luminal obstruction inhibited 90. The fact that dual‐specific Tregs protected the integrity more effectively than any population of Tregs found naturally highlighted the clinical potential for gene‐modified cell therapy.

The studies summarized above describe the ability to generate antigen‐specific Tregs by delivering designated TCR α and β chain genes. However, a different approach to confer antigen specificity which has gained popularity during the past few years is to use chimeric antigen receptors (CAR) 106. CARs are clinically relevant synthetic fusion proteins which combine various protein elements to redirect the specificity of T cells towards desired antigens of interest. CARs have been developed principally as a means of generating tumour antigen‐specific T cells 107 and their clinical use 108 has seen resounding success, particularly in the treatment of various B cell malignancies 109, 110, 111, 112, 113, 114, 115, 116, 117, 118.

In addition to their use in various cancer settings, CAR technology has also been employed to generate antigen‐specific Tregs. Initial studies generated CAR Tregs to investigate their potential for treating various autoimmune conditions 119, 120, 121, 122. However, more recently, we and others have explored the potential of CAR Tregs for the treatment of xeno‐GVHD and allograft rejection 123, 124, 125. In these studies, CARs were developed to redirect human Tregs towards a designated human leucocyte antigen (HLA) class I molecule (HLA‐A2). A donor HLA class I molecule was selected as this alloantigen is expressed ubiquitously in allografts, unlike HLA class II. Together with the findings of Noyan et al., our results demonstrated that CAR Tregs inhibited alloimmune‐mediated injury of human skin xenografts more effectively than polyclonal Tregs 124, 125, thus suggesting a potential avenue for future clinical trials. This is particularly likely given the fact that the biotechnology company TxCell (Valbonne, France) recently announced the approval of a patent for the development of CAR Tregs for treating various autoimmune and inflammatory diseases (patent identification number: EP 2126054 A2).

Survival in vivo

An additional concern post‐Treg infusion is their survival in vivo. Initially, research suggested that Treg survival post‐adoptive administration is short‐lived, at approximately 14 days after monitoring Treg donor‐specific HLA markers post‐infusion 51. However, Bluestone et al. in his most recent trial of Treg cell therapy in type 1 diabetes described an elegant method for the tracking of Tregs post‐infusion 86. In this trial, during expansion Tregs were labelled with deuterium (2H2), which was incorporated into replicating Treg DNA. Following this, Tregs were analysed at various time‐points post‐administration for deuterium enrichment. Results from this trial showed that the number of Tregs reached a maximum at 7–14 days post‐transplantation, with a subsequent decline. Despite this initial fall‐off, after 14 days the percentage of Tregs soon stabilized and were detectable up to a year after infusion. As such, it was postulated that Treg survival is centred on a two‐phase decline curve, t1/2 of the first decay at 19·2 days and t1/2 of the second decay at a year or more 86. Further work in this area is warranted not only to define Treg survival in vivo, but also to investigate Treg localization post‐infusion.

Cryopreservation

Based on the suggested doses for Treg therapy, it is uncertain whether a single dose will be sufficient or whether multiple doses are required for tolerance induction. The feasibility of multiple injections and, along this line, the practicality of cryopreservation has been questioned. Cryopreservation in Treg manufacturing allows for the long‐term storage of isolated and expanded Tregs until they are needed, giving more flexibility in terms of ease of transport to patients’ location, follow‐up injections and allows for investigation into whether the final product fits the set release criteria 126; however, it can pose various technical challenges. Our recent publication concerning the manufacture of GMP‐compatible Tregs for cell therapy investigated the feasibility of cryopreservation. Here cells were suspended in 10% dimethylsulphoxide (DMSO) and placed into a controlled‐rate freezer before being transferred to liquid nitrogen (vapour phase) for long‐term storage. After 3 months, these cells were thawed and assessed. We demonstrated that Treg viability post‐defrosting was > 85% with concurrent maintenance of function and phenotype 62.

Treg therapy in solid organ transplantation to date

There has been a wealth of clinical trials investigating Treg adoptive cell therapy in bone marrow transplantation and T1D 51, 64, 65, 84, 86. The main readouts from the trials were safety and tolerability, which were demonstrated. However, although not tested explicitly, there were hints of efficacy, evidenced in trials of BMT with the amelioration of GVHD.

Definitive trials demonstrating the relative safety and efficacy of adoptive Treg therapy in the context of solid organ transplantation are scarce. Only recently have a Japanese group published the results of Treg cell therapy in the setting of liver transplantation 127, where iTregs were generated through the culture of recipient lymphocytes with irradiated donor cells in the presence of anti‐CD80/CD86 monoclonal antibodies. These iTregs were then infused 13 days after living donor liver transplantation in 10 recipients. Immunosuppression weaning was commenced 6 months after transplantation, with complete cessation established at 18 months. Of the 10 LT recipients, seven allowed for complete cessation of immunosuppression while maintaining tolerance for more than 12 months, with four now demonstrating tolerant graft function between 29 and 33 months post‐IS withdrawal 127. The proportion of Tregs dosed amounted to an average of 3·4 × 106/kg, a value much lower than that postulated by Tang et al. to be necessary for the induction of tolerance. However, in this study the iTregs were deemed to be donor‐antigen specific, recipients were treated with cyclophosphamide prior to infusion and subjects were splenectomized, all of which may have contributed to the apparent efficacy of Treg therapy at this reduced dose.

Currently there are several ongoing/planned trials in the setting of liver and kidney transplantation (Table 1). The clinical trials ThRIL and the ONE study are the leading trials of autologous Treg immunotherapy worldwide in the setting of liver and kidney transplantation, respectively.

Table 1.

Table listing details of all ongoing trials of Treg adoptive cell therapy in solid organ transplantation recorded on clinicaltrials.gov

Clinical trial Sponsor Setting Phase Status Product Treg doses Timing of infusion
ONETreg1 NCT02129881 Guy's and St Thomas’ NHS Foundation Trust Kidney transplantation 1/2 Recruiting Polyclonal, autologous CD4+CD25+ Tregs Single infusion of 1–10 × 106/kg 5 days post‐renal transplantation
DART NCT02244801 University of California Kidney transplantation 1 Recruiting Donor‐alloantigen‐reactive autologous regulatory T cells Single infusion of 300 × 106 and 900 × 106 Timing not specified
ONEnTreg13 NCT02371434 Charite University, Berlin, Germany Kidney transplantation 1/2 Recruiting: invitation only Polyclonal, autologous CD4+CD25+ Tregs Single infusion of 0.5 × 106, 1 ×  106 and 3 × 106 cells/kg Timing not specified
NCT02091232 Massachusetts General Hospital Kidney transplantation 1 Ongoing: no longer recruiting Belatacept anergized donor‐alloantigen‐reactive Tregs Infusion dose not specified 3 days post‐renal transplantation
TRACT NCT02145325 Northwestern University Kidney transplantation 1 Ongoing: no longer recruiting Polyclonal, autologous CD4+CD25+ Tregs Infusion dose not specified 2 months post‐renal transplantation
TASK NCT02088931 University of California Kidney transplantation 1 Ongoing: no longer recruiting Polyclonal, autologous CD4+CD25+CD127 Tregs Single infusion of 320 × 106 On their 6‐month surveillance biopsy post‐renal transplantation
TASK NCT02711826 National Institute of Allergy and Infectious Diseases Subclinical inflammation in kidney transplantation 1/2 Recruiting Polyclonal and donor‐alloantigen‐reactive autologous regulatory T cells Single infusion 400 × 106 On their 6‐month surveillance biopsy post‐renal transplantation
NCT01446484 Pirogov Russian National Research Medical University Kidney transplantation 1/2 Unknown Polyclonal, autologous CD4+CD25+ CD127lowFoxP3+ Tregs Two injections at 200 × 106 30 days and 6 months post‐renal transplantation
ThRIL NCT02166177 Guy's and St Thomas’ NHS Foundation Trust Liver transplantation 1 Ongoing: no longer recruiting Polyclonal autologous CD4+CD25+ Tregs Single infusion of 1 × 106 cells/kg and 4.5 × 106 cells/kg 12 weeks post‐liver transplantation
NCT01624077 Nanjing Medical University Liver transplantation 1 Ongoing: no longer recruiting Polyclonal and donor‐alloantigen‐reactive regulatory autologous T cells CD4+CD25+CD127 Tregs Multiple doses of 1 ×  106 cells/kg at several intervals Timing not specified
deLTa NCT02188719 National Institute of Allergy and Infectious Diseases Liver transplantation 1 Recruiting Donor‐alloantigen‐reactive autologous Tregs Single infusion 50 × 106, 200 × 106 and 800 × 106 Timing not specified
ARTEMIS NCT02474199 National Institute of Allergy and Infectious Diseases Liver transplantation 1/2 Recruiting Donor‐alloantigen‐reactive, autologous Tregs Single infusion of 400 × 106 Timing not specified

The ONE Study (NCT02129881) is a multi‐centre Phase I/II study funded by the European Union FP7 programme. This trial investigates the safety of and potential efficacy of infusing ex‐vivo‐expanded Tregs, among other regulatory cells, in the setting of renal transplantation.

ThRIL (NCT02166177) is a Phase I clinical trial of Treg immunotherapy the setting of liver transplantation, and has been initiated at King's College London, in which the safety, tolerability and efficacy of polyclonally expanded Tregs in combination with depletion therapy and short‐term immunosuppression will be assessed. At present, three patients have been dosed, with encouraging safety data to date.

Future

The understanding and use of Treg therapy is an important and emerging area of science/medicine. Following completion of the ongoing clinical trials, we will have safety and tolerability data as well as initial efficacy data in the transplantation population. This will serve as proof‐of‐principle for use of these cells in solid organ transplantation and provide the basis for progression to larger Phase II/III studies. Concurrent immune monitoring integrated as part of the Treg therapy trials will help to gain further mechanistic insight on Treg function in patients, further informing future trials. However, as highlighted in this review, with the advent of new technology such as the CyTOF we now appreciate the phenotypical complexity of Treg compartment and can characterize comprehensively rare and complex disease‐specific populations. With the GMP certification of the FACS cell sorter, as discussed before, and the information gleaned from CyTOF analysis, we will be able to isolate the optimal cell populations for expansion and cell therapeutic application.

Parallel to this work, we will also see improvements in the clinical protocol and the use of adjunct therapy to ensure Treg survival in vivo. Currently, ongoing trials have looked at optimizing Treg therapy through modulating immunosuppressive regimens which favour Treg expansion and stability (NTC02166177) and pretreating Tregs with anergistics such as belatacept (NCT02091232). As Tregs depend upon exogenous IL‐2 for survival, recent strategies to ensure the longevity of the injected cells have explored the use of low‐dose IL‐2, which lacks the toxicity and immunostimulatory effects of the higher IL‐2 doses used to treat cancer patients 128, 129, 130.

This approach has been shown recently to increase the number of endogenous Tregs in patients with chronic GVHD 131, providing supporting evidence that this therapeutic strategy may be an ideal adjuvant to adoptive Treg cell therapy by promoting Treg expansion in an otherwise inflammatory setting. However, further studies on the isolation and expansion of patient‐derived Tregs are warranted, in view of publications highlighting the differential ability of ‘healthy’ and patient‐derived Tregs to respond to IL‐2. In line with these reports, the ability of Tregs to expand and respond to treatments such as low‐dose IL‐2 might be compromised in certain disease settings due to defective Treg responses to IL‐2 and IL‐2 signalling 132, 133.

It is also pertinent to note that at present the cost of manufacturing a single ‘personalized’ injection of Tregs in our GMP facility is £25–30 000. We anticipate that the future optimization of the manufacturing process for larger‐scale trials and commercialization would reduce the cost, enabling the broader application of this therapy 134.

In conclusion, these advances have highlighted further the relative depth and individuality of Treg biology in disease, and as such efforts will be focused upon tailoring Treg therapy with the goal of developing an optimized, personalized treatment in the striving for tolerance in solid organ transplantation.

Disclosure

The authors declare no financial or commercial conflicts of interest.

Acknowledgements

The authors thank all the research subjects and patients who are currently taking part in the ONE study and ThRIL. A special thank you to the GMP scientists, head of GMP production and head of GMP quality at Guy's Hospital. This research is supported by the King's Health Partners Research and Development Challenge Fund, Guy's and St Thomas’ Charity (grant no. R1405170), BHF, ONE Study, the Medical Research Council (MRC), within the MRC Centre for Transplantation, King's College London, UK – MRC grant no. MR/J006742/1 and the National Institute for Health Research (NIHR) Biomedical Research Centre at Guy's and St Thomas’ NHS Foundation Trust and King's College London. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health.

Contributor Information

T. Vaikunthanathan, Email: trishan_vaik@yahoo.com

G. Lombardi, Email: giovanna.lombardi@kcl.ac.uk

References

  • 1. Feng S. Long‐term management of immunosuppression after pediatric liver transplantation: is minimization or withdrawal desirable or possible or both? Curr Opin Organ Transplant 2008; 13:506–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ojo AO, Held PJ, Port FK et al Chronic renal failure after transplantation of a nonrenal organ. N Engl J Med 2003; 349:931–40. [DOI] [PubMed] [Google Scholar]
  • 3. Textor SC, Taler SJ, Canzanello VJ, Schwartz L, Augustine JE. Posttransplantation hypertension related to calcineurin inhibitors. Liver Transpl 2000; 6:521–30. [DOI] [PubMed] [Google Scholar]
  • 4. Penn I. Cancers complicating organ transplantation. N Engl J Med 1990; 323:1767–9. [DOI] [PubMed] [Google Scholar]
  • 5. Katabathina V, Menias CO, Pickhardt P, Lubner M, Prasad SR. Complications of immunosuppressive therapy in solid organ transplantation. Radiol Clin North Am 2016; 54:303–19. [DOI] [PubMed] [Google Scholar]
  • 6. Benitez C, Londono MC, Miquel R et al Prospective multicenter clinical trial of immunosuppressive drug withdrawal in stable adult liver transplant recipients. Hepatology 2013; 58:1824–35. [DOI] [PubMed] [Google Scholar]
  • 7. Liu XQ, Hu ZQ, Pei YF, Tao R. Clinical operational tolerance in liver transplantation: state‐of‐the‐art perspective and future prospects. Hepatobiliary Pancreat Dis Int 2013; 12:12–33. [DOI] [PubMed] [Google Scholar]
  • 8. Sagoo P, Perucha E, Sawitzki B et al Development of a cross‐platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest 2010; 120:1848–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sanchez‐Fueyo A. Hot‐topic debate on tolerance: immunosuppression withdrawal. Liver Transpl 2011; 17:S69–73. [DOI] [PubMed] [Google Scholar]
  • 10. Sanchez‐Fueyo A. Tolerance profiles and immunosuppression. Liver Transpl 2013; 19:S44–8. [DOI] [PubMed] [Google Scholar]
  • 11. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self‐tolerance maintained by activated T cells expressing IL‐2 receptor alpha‐chains (CD25). Breakdown of a single mechanism of self‐tolerance causes various autoimmune diseases. J Immunol 1995; 155:1151–64. [PubMed] [Google Scholar]
  • 12. Longhi MS, Ma Y, Bogdanos DP, Cheeseman P, Mieli‐Vergani G, Vergani D. Impairment of CD4(+)CD25(+) regulatory T‐cells in autoimmune liver disease. J Hepatol 2004; 41:31–7. [DOI] [PubMed] [Google Scholar]
  • 13. Hanash AM, Levy RB. Donor CD4+CD25+ T cells promote engraftment and tolerance following MHC‐mismatched hematopoietic cell transplantation. Blood 2005; 105:1828–36. [DOI] [PubMed] [Google Scholar]
  • 14. Joffre O, Gorsse N, Romagnoli P, Hudrisier D, van Meerwijk JP. Induction of antigen‐specific tolerance to bone marrow allografts with CD4+CD25+ T lymphocytes. Blood 2004; 103:4216–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Joffre O, Santolaria T, Calise D et al Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ regulatory T lymphocytes. Nat Med 2008; 14:88–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Li Y, Koshiba T, Yoshizawa A et al Analyses of peripheral blood mononuclear cells in operational tolerance after pediatric living donor liver transplantation. Am J Transplant 2004; 4:2118–25. [DOI] [PubMed] [Google Scholar]
  • 17. Meloni F, Morosini M, Solari N et al Peripheral CD4+ CD25+ Treg cell expansion in lung transplant recipients is not affected by calcineurin inhibitors. Int Immunopharmacol 2006; 6:2002–10. [DOI] [PubMed] [Google Scholar]
  • 18. Bestard O, Cruzado JM, Mestre M et al Achieving donor‐specific hyporesponsiveness is associated with FOXP3+ regulatory T cell recruitment in human renal allograft infiltrates. J Immunol 2007; 179:4901–9. [DOI] [PubMed] [Google Scholar]
  • 19. Allan SE, Passerini L, Bacchetta R et al The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest 2005; 115:3276–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Bacchetta R, Gambineri E, Roncarolo MG. Role of regulatory T cells and FOXP3 in human diseases. J Allergy Clin Immunol 2007; 120:227–35; quiz 36–7. [DOI] [PubMed] [Google Scholar]
  • 21. Bacchetta R, Barzaghi F, Roncarolo MG. From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation. Ann NY Acad Sci 2016; doi: 10.1111/nyas.13011. [DOI] [PubMed] [Google Scholar]
  • 22. Torgerson TR, Ochs HD. Immune dysregulation, polyendocrinopathy, enteropathy, X‐linked: forkhead box protein 3 mutations and lack of regulatory T cells. J Allergy Clin Immunol 2007; 120:744–50; quiz 51–2. [DOI] [PubMed] [Google Scholar]
  • 23. Ziegler SF. FOXP3: not just for regulatory T cells anymore. Eur J Immunol 2007; 37:21–3. [DOI] [PubMed] [Google Scholar]
  • 24. Liu W, Putnam AL, Xu‐Yu Z et al CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ Treg cells. J Exp Med 2006; 203:1701–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Seddiki N, Santner‐Nanan B, Martinson J et al Expression of interleukin (IL)‐2 and IL‐7 receptors discriminates between human regulatory and activated T cells. J Exp Med 2006; 203:1693–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Nadig SN, Wieckiewicz J, Wu DC et al In vivo prevention of transplant arteriosclerosis by ex vivo‐expanded human regulatory T cells. Nat Med 2010; 16:809–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Issa F, Hester J, Goto R, Nadig SN, Goodacre TE, Wood K. Ex vivo‐expanded human regulatory T cells prevent the rejection of skin allografts in a humanized mouse model. Transplantation 2010; 90:1321–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Miyara M, Yoshioka Y, Kitoh A et al Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009; 30:899–911. [DOI] [PubMed] [Google Scholar]
  • 29. Booth NJ, McQuaid AJ, Sobande T et al Different proliferative potential and migratory characteristics of human CD4+ regulatory T cells that express either CD45RA or CD45RO. J Immunol 2010; 184:4317–26. [DOI] [PubMed] [Google Scholar]
  • 30. Afzali B, Mitchell PJ, Edozie FC et al CD161 expression characterizes a subpopulation of human regulatory T cells that produces IL‐17 in a STAT3‐dependent manner. Eur J Immunol 2013; 43:2043–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Duhen T, Duhen R, Lanzavecchia A, Sallusto F, Campbell DJ. Functionally distinct subsets of human FOXP3+ Treg cells that phenotypically mirror effector Th cells. Blood 2012; 119:4430–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Weiss JM, Bilate AM, Gobert M et al Neuropilin 1 is expressed on thymus‐derived natural regulatory T cells, but not mucosa‐generated induced Foxp3+ Treg cells. J Exp Med 2012; 209:1723–42. S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Yadav M, Louvet C, Davini D et al Neuropilin‐1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo . J Exp Med 2012; 209:1713–22, S1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Getnet D, Grosso JF, Goldberg MV et al A role for the transcription factor Helios in human CD4(+)CD25(+) regulatory T cells. Mol Immunol 2010; 47:1595–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Szurek E, Cebula A, Wojciech L et al Differences in expression level of Helios and neuropilin‐1 do not distinguish thymus‐derived from extrathymically‐induced CD4+Foxp3+ regulatory T cells. PLOS ONE 2015; 10:e0141161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Polansky JK, Kretschmer K, Freyer J et al DNA methylation controls Foxp3 gene expression. Eur J Immunol 2008; 38:1654–63. [DOI] [PubMed] [Google Scholar]
  • 37. Huehn J, Polansky JK, Hamann A. Epigenetic control of FOXP3 expression: the key to a stable regulatory T‐cell lineage? Nat Rev Immunol 2009; 9:83–9. [DOI] [PubMed] [Google Scholar]
  • 38. Polansky JK, Schreiber L, Thelemann C et al Methylation matters: binding of Ets‐1 to the demethylated Foxp3 gene contributes to the stabilization of Foxp3 expression in regulatory T cells. J Mol Med 2010; 88:1029–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Povoleri GA, Scotta C, Nova‐Lamperti EA, John S, Lombardi G, Afzali B. Thymic versus induced regulatory T cells – who regulates the regulators? Front Immunol 2013; 4:169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Diggins KE, Ferrell PB Jr, Irish JM. Methods for discovery and characterization of cell subsets in high dimensional mass cytometry data. Methods 2015; 82:55–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Irish JM. Beyond the age of cellular discovery. Nat Immunol 2014; 15:1095–7. [DOI] [PubMed] [Google Scholar]
  • 42. Mason GM, Lowe K, Melchiotti R et al Phenotypic complexity of the human regulatory T cell compartment revealed by mass cytometry. J Immunol 2015; 195:2030–7. [DOI] [PubMed] [Google Scholar]
  • 43. Kordasti S, Costantini B, Seidl T et al Deep phenotyping of Tregs identifies an immune signature for idiopathic aplastic anemia and predicts response to treatment. Blood 2016; 128:1193–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Bobrie A, Colombo M, Raposo G, Thery C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic 2011; 12:1659–68. [DOI] [PubMed] [Google Scholar]
  • 45. Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol 2002; 2:569–79. [DOI] [PubMed] [Google Scholar]
  • 46. Smyth LA, Ratnasothy K, Tsang JY et al CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur J Immunol 2013; 43:2430–40. [DOI] [PubMed] [Google Scholar]
  • 47. Okoye IS, Coomes SM, Pelly VS et al MicroRNA‐containing T‐regulatory‐cell‐derived exosomes suppress pathogenic T helper 1 cells. Immunity 2014; 41:89–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Riley JL, June CH, Blazar BR. Human T regulatory cell therapy: take a billion or so and call me in the morning. Immunity 2009; 30:656–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Godfrey WR, Ge YG, Spoden DJ et al In vitro‐expanded human CD4(+)CD25(+) T‐regulatory cells can markedly inhibit allogeneic dendritic cell‐stimulated MLR cultures. Blood 2004; 104:453–61. [DOI] [PubMed] [Google Scholar]
  • 50. Godfrey WR, Spoden DJ, Ge YG et al Cord blood CD4(+)CD25(+)‐derived T regulatory cell lines express FoxP3 protein and manifest potent suppressor function. Blood 2005; 105:750–8. [DOI] [PubMed] [Google Scholar]
  • 51. Brunstein CG, Miller JS, Cao Q et al Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2011; 117:1061–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Dijke IE, Hoeppli RE, Ellis T et al Discarded human thymus is a novel source of stable and long‐lived therapeutic regulatory T cells. Am J Transplant 2016; 16:58–71. [DOI] [PubMed] [Google Scholar]
  • 53. Hoffmann P, Boeld TJ, Eder R et al Isolation of CD4+CD25+ regulatory T cells for clinical trials. Biol Blood Marrow Transplant 2006; 12:267–74. [DOI] [PubMed] [Google Scholar]
  • 54. Scheffold A. How can the latest technologies advance cell therapy manufacturing? Curr Opin Organ Transplant 2014; 19:621–6. [DOI] [PubMed] [Google Scholar]
  • 55. Putnam AL, Brusko TM, Lee MR et al Expansion of human regulatory T‐cells from patients with type 1 diabetes. Diabetes 2009; 58:652–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Hoffmann P, Eder R, Boeld TJ et al Only the CD45RA+ subpopulation of CD4+CD25high T cells gives rise to homogeneous regulatory T‐cell lines upon in vitro expansion. Blood 2006; 108:4260–7. [DOI] [PubMed] [Google Scholar]
  • 57. Canavan JB, Scotta C, Vossenkamper A et al Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn's disease. Gut 2016; 65:584–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Golshayan D, Jiang S, Tsang J, Garin MI, Mottet C, Lechler RI. In vitro‐expanded donor alloantigen‐specific CD4+CD25+ regulatory T cells promote experimental transplantation tolerance. Blood 2007; 109:827–35. [DOI] [PubMed] [Google Scholar]
  • 59. Nishimura E, Sakihama T, Setoguchi R, Tanaka K, Sakaguchi S. Induction of antigen‐specific immunologic tolerance by in vivo and in vitro antigen‐specific expansion of naturally arising Foxp3+CD25+CD4+ regulatory T cells. Int Immunol 2004; 16:1189–201. [DOI] [PubMed] [Google Scholar]
  • 60. Graca L, Thompson S, Lin CY, Adams E, Cobbold SP, Waldmann H. Both CD4(+)CD25(+) and CD4(+)CD25(–) regulatory cells mediate dominant transplantation tolerance. J Immunol 2002; 168:5558–65. [DOI] [PubMed] [Google Scholar]
  • 61. Tang Q, Lee K. Regulatory T‐cell therapy for transplantation: how many cells do we need? Curr Opin Organ Transplant 2012; 17:349–54. [DOI] [PubMed] [Google Scholar]
  • 62. Safinia N, Vaikunthanathan T, Fraser H et al Successful expansion of functional and stable regulatory T cells for immunotherapy in liver transplantation. Oncotarget 2016; 7:7563–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hoffmann P, Boeld TJ, Eder R et al Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur J Immunol 2009; 39:1088–97. [DOI] [PubMed] [Google Scholar]
  • 64. Trzonkowski P, Bieniaszewska M, Juscinska J et al First‐in‐man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127‐ T regulatory cells. Clin Immunol 2009; 133:22–6. [DOI] [PubMed] [Google Scholar]
  • 65. Marek‐Trzonkowska N, Mysliwiec M, Dobyszuk A et al Therapy of type 1 diabetes with CD4(+)CD25(high)CD127‐regulatory T cells prolongs survival of pancreatic islets – results of one‐year follow‐up. Clin Immunol 2014; 153:23–30. [DOI] [PubMed] [Google Scholar]
  • 66. Battaglia M, Stabilini A, Migliavacca B, Horejs‐Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol 2006; 177:8338–47. [DOI] [PubMed] [Google Scholar]
  • 67. Afzali B, Edozie FC, Fazekasova H et al Comparison of regulatory T cells in hemodialysis patients and healthy controls: implications for cell therapy in transplantation. Clin J Am Soc Nephrol 2013; 8:1396–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Battaglia M, Stabilini A, Roncarolo MG. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood 2005; 105:4743–8. [DOI] [PubMed] [Google Scholar]
  • 69. Scotta C, Esposito M, Fazekasova H et al Differential effects of rapamycin and retinoic acid on expansion, stability and suppressive qualities of human CD4(+)CD25(+)FOXP3(+) T regulatory cell subpopulations. Haematologica 2013; 98:1291–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Zeiser R, Leveson‐Gower DB, Zambricki EA et al Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells. Blood 2008; 111:453–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Zhou X, Bailey‐Bucktrout S, Jeker LT, Bluestone JA. Plasticity of CD4(+) FoxP3(+) T cells. Curr Opin Immunol 2009; 21:281–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Yang XO, Nurieva R, Martinez GJ et al Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 2008; 29:44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Hori S. Regulatory T cell plasticity: beyond the controversies. Trends Immunol 2011; 32:295–300. [DOI] [PubMed] [Google Scholar]
  • 74. Voo KS, Wang YH, Santori FR et al Identification of IL‐17‐producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci USA 2009; 106:4793–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Pesenacker AM, Bending D, Ursu S, Wu Q, Nistala K, Wedderburn LR. CD161 defines the subset of FoxP3+ T cells capable of producing proinflammatory cytokines. Blood 2013; 121:2647–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol 2011; 11:119–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Allan SE, Crome SQ, Crellin NK et al Activation‐induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol 2007; 19:345–54. [DOI] [PubMed] [Google Scholar]
  • 78. Floess S, Freyer J, Siewert C et al Epigenetic control of the foxp3 locus in regulatory T cells. PLOS Biol 2007; 5:e38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Sanchez‐Abarca LI, Gutierrez‐Cosio S, Santamaria C et al Immunomodulatory effect of 5‐azacytidine (5‐azaC): potential role in the transplantation setting. Blood 2010; 115:107–21. [DOI] [PubMed] [Google Scholar]
  • 80. Goodyear OC, Dennis M, Jilani NY et al Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood 2012; 119:3361–9. [DOI] [PubMed] [Google Scholar]
  • 81. Kwon HS, Lim HW, Wu J, Schnolzer M, Verdin E, Ott M. Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. J Immunol 2012; 188:2712–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Liu Y, Wang L, Han R, Beier UH, Hancock WW. Two lysines in the forkhead domain of foxp3 are key to T regulatory cell function. PLoS One 2012; 7:e29035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Beier UH, Wang L, Bhatti TR et al Sirtuin‐1 targeting promotes Foxp3+ T‐regulatory cell function and prolongs allograft survival. Mol Cell Biol 2011; 31:1022–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Di Ianni M, Falzetti F, Carotti A et al Tregs prevent GVHD and promote immune reconstitution in HLA‐haploidentical transplantation. Blood 2011; 117:3921–8. [DOI] [PubMed] [Google Scholar]
  • 85. Theil A, Tuve S, Oelschlagel U et al Adoptive transfer of allogeneic regulatory T cells into patients with chronic graft‐versus‐host disease. Cytotherapy 2015; 17:473–86. [DOI] [PubMed] [Google Scholar]
  • 86. Bluestone JA, Buckner JH, Fitch M et al Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015; 7:315ra189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Tang Q, Henriksen KJ, Bi M et al In vitro‐expanded antigen‐specific regulatory T cells suppress autoimmune diabetes. J Exp Med 2004; 199:1455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft‐versus‐host disease lethality. Blood 2002; 99:3493–9. [DOI] [PubMed] [Google Scholar]
  • 89. Tsang JY, Tanriver Y, Jiang S et al Indefinite mouse heart allograft survival in recipient treated with CD4(+)CD25(+) regulatory T cells with indirect allospecificity and short term immunosuppression. Transpl Immunol 2009; 21:203–9. [DOI] [PubMed] [Google Scholar]
  • 90. Tsang JY, Tanriver Y, Jiang S et al Conferring indirect allospecificity on CD4+CD25+ Tregs by TCR gene transfer favors transplantation tolerance in mice. J Clin Invest 2008; 118:3619–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Putnam AL, Safinia N, Medvec A et al Clinical grade manufacturing of human alloantigen‐reactive regulatory T cells for use in transplantation. Am J Transplant 2013; 13:3010–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Sagoo P, Ali N, Garg G, Nestle FO, Lechler RI, Lombardi G. Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci Transl Med 2011; 3:83ra42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Peters JH, Hilbrands LB, Koenen HJ, Joosten I. Ex vivo generation of human alloantigen‐specific regulatory T cells from CD4(pos)CD25(high) T cells for immunotherapy. PLOS ONE 2008; 3:e2233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Noyan F, Lee YS, Hardtke‐Wolenski M et al Donor‐specific regulatory T cells generated on donor B cells are superior to CD4+CD25high cells in controlling alloimmune responses in humanized mice. Transplant Proc 2013; 45:1832–7. [DOI] [PubMed] [Google Scholar]
  • 95. Chen LC, Delgado JC, Jensen PE, Chen X. Direct expansion of human allospecific FoxP3+CD4+ regulatory T cells with allogeneic B cells for therapeutic application. J Immunol 2009; 183:4094–102. [DOI] [PubMed] [Google Scholar]
  • 96. Boardman DA, Jacob J, Smyth LA, Lombardi G, Lechler RI. What is direct allorecognition? Curr Transplant Rep 2016; 3:275–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Veerapathran A, Pidala J, Beato F, Yu XZ, Anasetti C. Ex vivo expansion of human Tregs specific for alloantigens presented directly or indirectly. Blood 2011; 118:5671–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Jiang S, Tsang J, Game DS, Stevenson S, Lombardi G, Lechler RI. Generation and expansion of human CD4+ CD25+ regulatory T cells with indirect allospecificity: potential reagents to promote donor‐specific transplantation tolerance. Transplantation 2006; 82:1738–43. [DOI] [PubMed] [Google Scholar]
  • 99. Zhou P, Borojevic R, Streutker C, Snider D, Liang H, Croitoru K. Expression of dual TCR on DO11.10 T cells allows for ovalbumin‐induced oral tolerance to prevent T cell‐mediated colitis directed against unrelated enteric bacterial antigens. J Immunol 2004; 172:1515–23. [DOI] [PubMed] [Google Scholar]
  • 100. Stephens LA, Malpass KH, Anderton SM. Curing CNS autoimmune disease with myelin‐reactive Foxp3+ Treg. Eur J Immunol 2009; 39:1108–17. [DOI] [PubMed] [Google Scholar]
  • 101. DiPaolo RJ, Brinster C, Davidson TS, Andersson J, Glass D, Shevach EM. Autoantigen‐specific TGFbeta‐induced Foxp3+ regulatory T cells prevent autoimmunity by inhibiting dendritic cells from activating autoreactive T cells. J Immunol 2007; 179:4685–93. [DOI] [PubMed] [Google Scholar]
  • 102. Fujio K, Okamoto A, Araki Y et al Gene therapy of arthritis with TCR isolated from the inflamed paw. J Immunol 2006; 177:8140–7. [DOI] [PubMed] [Google Scholar]
  • 103. Wright GP, Notley CA, Xue SA et al Adoptive therapy with redirected primary regulatory T cells results in antigen‐specific suppression of arthritis. Proc Natl Acad Sci USA 2009; 106:19078–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Tarbell KV, Petit L, Zuo X et al Dendritic cell‐expanded, islet‐specific CD4+ CD25+ CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice. J Exp Med 2007; 204:191–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Trenado A, Sudres M, Tang Q et al Ex vivo‐expanded CD4+CD25+ immunoregulatory T cells prevent graft‐versus‐host‐disease by inhibiting activation/differentiation of pathogenic T cells. J Immunol 2006; 176:1266–73. [DOI] [PubMed] [Google Scholar]
  • 106. Boardman D, Maher J, Lechler R, Smyth L, Lombardi G. Antigen‐specificity using chimeric antigen receptors: the future of regulatory T‐cell therapy? Biochem Soc Trans 2016; 44:342–8. [DOI] [PubMed] [Google Scholar]
  • 107. Sadelain M, Brentjens R, Riviere I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol 2009; 21:215–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Dai H, Wang Y, Lu X, Han W. Chimeric antigen receptors modified T‐cells for cancer therapy. J Natl Cancer Inst 2016; 108:djv439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor‐modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365:725–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Kalos M, Levine BL, Porter DL et al T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 2011; 3:95ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Grupp SA, Kalos M, Barrett D et al Chimeric antigen receptor‐modified T cells for acute lymphoid leukemia. N Engl J Med 2013; 368:1509–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Brentjens RJ, Davila ML, Riviere I et al CD19‐targeted T cells rapidly induce molecular remissions in adults with chemotherapy‐refractory acute lymphoblastic leukemia. Sci Transl Med 2013; 5:177ra38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Porter DL, Hwang WT, Frey NV et al Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015; 7:303ra139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Maude SL, Frey N, Shaw PA et al Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371:1507–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Davila ML, Riviere I, Wang X et al Efficacy and toxicity management of 19‐28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014; 6:224ra25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Till BG, Jensen MC, Wang J et al CD20‐specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4‐1BB domains: pilot clinical trial results. Blood 2012; 119:3940–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Kochenderfer JN, Wilson WH, Janik JE et al Eradication of B‐lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood 2010; 116:4099–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kohn DB, Dotti G, Brentjens R et al CARs on track in the clinic. Mol Ther 2011; 19:432–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Elinav E, Waks T, Eshhar Z. Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice. Gastroenterology 2008; 134:2014–24. [DOI] [PubMed] [Google Scholar]
  • 120. Elinav E, Adam N, Waks T, Eshhar Z. Amelioration of colitis by genetically engineered murine regulatory T cells redirected by antigen‐specific chimeric receptor. Gastroenterology 2009; 136:1721–31. [DOI] [PubMed] [Google Scholar]
  • 121. Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z. Suppression of murine colitis and its associated cancer by carcinoembryonic antigen‐specific regulatory T cells. Mol Ther 2014; 22:1018–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Fransson M, Piras E, Burman J et al CAR/FoxP3‐engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery. J Neuroinflammation 2012; 9:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. MacDonald KG, Hoeppli RE, Huang Q et al Alloantigen‐specific regulatory T cells generated with a chimeric antigen receptor. J Clin Invest 2016; 126:1413–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Boardman DA, Philippeos C, Fruhwirth GO et al Expression of a chimeric antigen receptor specific for donor HLA class I enhances the potency of human regulatory T cells in preventing human skin transplant rejection. Am J Transplant 2017; 17:931–43. [DOI] [PubMed] [Google Scholar]
  • 125. Noyan F, Zimmermann K, Hardtke‐Wolenski M et al Prevention of allograft rejection by use of regulatory T cells with a MHC‐specific chimeric antigen receptor. Am J Transplant 2017; 17:917–30. [DOI] [PubMed] [Google Scholar]
  • 126. Safinia N, Scotta C, Vaikunthanathan T, Lechler RI, Lombardi G. Regulatory T cells: serious contenders in the promise for immunological tolerance in transplantation. Front Immunol 2015; 6:438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Todo S, Yamashita K, Goto R et al A pilot study of operational tolerance with a regulatory T‐cell‐based cell therapy in living donor liver transplantation. Hepatology 2016; 64:632–43. [DOI] [PubMed] [Google Scholar]
  • 128. Moon BI, Kim TH, Seoh JY. Functional modulation of regulatory T cells by IL‐2. PLOS ONE 2015; 10:e0141864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Whitehouse GGE, Mastoridis S, Kodela E, Sanchez‐Fueyo A, Martinez‐Llordella M. IL‐2 therapy restores the impaired function of regulatory T cells induced by calcineurin inhibitors in liver transplantation. Am J Transplant 2016; 16 (Suppl 3). [Google Scholar]
  • 130. Boyman O, Sprent J. The role of interleukin‐2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12:180–90. [DOI] [PubMed] [Google Scholar]
  • 131. Koreth J, Matsuoka K, Kim HT et al Interleukin‐2 and regulatory T cells in graft‐versus‐host disease. N Engl J Med 2011; 365:2055–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Long SA, Cerosaletti K, Bollyky PL et al Defects in IL‐2R signaling contribute to diminished maintenance of FOXP3 expression in CD4(+)CD25(+) regulatory T‐cells of type 1 diabetic subjects. Diabetes 2010; 59:407–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Cerosaletti K, Schneider A, Schwedhelm K et al Multiple autoimmune‐associated variants confer decreased IL‐2R signaling in CD4+ CD25(hi) T cells of type 1 diabetic and multiple sclerosis patients. PLoS One 2013; 8:e83811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Leslie M. Immunology. Regulatory T cells get their chance to shine. Science 2011; 332:1020–1. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES