Abstract
CD4+FoxP3+ regulatory T cells (Tregs) are a non-redundant population critical for the maintenance of self-tolerance. Over the past decade, the use of these cells for therapeutic purposes in transplantation and autoimmune disease has emerged based on their capacity to inhibit immune activation. Basic science discoveries have led to identifying key receptors on Tregs that can regulate their proliferation and function. Notably, the understanding that IL-2 signaling is crucial for Treg homeostasis promoted the hypothesis that in vivo IL-2 treatment could provide a strategy to control the compartment. The use of low-dose IL-2 in vivo was shown to selectively expand Tregs versus other immune cells. Interestingly, a number of other Treg cell surface proteins, including CD28, CD45, IL-33R and TNFRSF members, have been identified which can also induce activation and proliferation of this population. Pre-clinical studies have exploited these observations to prevent and treat mice developing autoimmune diseases and graft-versus-host disease post-allogeneic hematopoietic stem cell transplantation. These findings support the development of translational strategies to expand Tregs in patients. Excitingly, the use of low-dose IL-2 for patients suffering from graft-versus-host disease and autoimmune disease has demonstrated increased Treg levels together with beneficial outcomes. To date, promising pre-clinical and clinical studies have directly targeted Tregs and clearly established the ability to increase their levels and augment their function in vivo. Here we review the evolving field of in vivo Treg manipulation and its application to allogeneic hematopoietic stem cell transplantation.Introduction
The identification of CD4FoxP3 regulatory T cells (Tregs) as a non-redundant cell population essential for the maintenance of peripheral self-tolerance has stimulated strong interest in their potential therapeutic application to promote allograft acceptance and ameliorate autoimmune diseases.51 The finding that Tregs are often present at tumor sites has also raised the prospect of augmenting antitumor immunity by diminishing their numbers or function.1161 Accordingly, the fields of transplantation, autoimmunity and oncology have converged on a common objective to selectively manipulate the Treg compartment to inhibit or promote conventional T-cell (Tconv) antigen-specific adaptive immune responses.
Clinical procedures developed to harvest Tregs for study and therapeutic application have been primarily based on cell surface expression of CD4, CD25 and CD127.1412 Employing magnetic bead or flow cytometric isolation methodology, viable and enriched preparations of Tregs have been generated for subsequent ex vivo expansion and translational use in patients.1815 Inherent in such in vitro manipulations is the absence of the precise in situ microenvironment wherein individual cell populations differentiate, undergo expansion and mediate effector function. Several established strategies have incorporated the use of micro-bead and antigen-presenting cell (APC)-based technologies to expand Tregs incorporating anti-CD3, CD28, and anti-TNFR family mAbs together with cytokines (e.g. IL-2, TGFβ, and retinoic acid).19 Successful expansion ranging from approximately 100-1300× was reported from starting populations of peripheral blood (CD4CD127) and umbilical cord (CD25) cells.2015 Notably, employing these Tregs in phase I studies reported no apparent toxicities or adverse effects.2115
Although Tregs can be induced to expand in vitro, size and purity of the initiating culture, the maintenance of FoxP3 expression and functional activity during culture vary and depend both on starting cell populations and culture conditions. Consistency of reagents from batch to batch also poses challenges for ultimate clinical application. Generation of a GMP product containing high cell numbers for adoptive therapy requires infrastructure and significant economic investment.2422 Accordingly, routinely generating adequate numbers of functional Tregs in vitro as a readily available adoptive therapy remains translationally challenging.25 Several excellent articles which include discussion of in vitro expansion methods have recently been published and we refer readers to these thorough reviews.3026 Strategies to manipulate Tregs in vivo have and continue to be examined to circumvent the practical and economic considerations that limit the feasibility of in vitro approaches. The provocative finding that low-dose IL-2 more efficiently stimulates Tregs versus Tconv populations has fostered optimism that selective manipulation of the FoxP3 compartment in situ can be exploited for clinical benefit. Because the production and expansion of effector versus Tregs is associated with the development of chronic graft-versus-host disease (cGvHD), correction of a Treg:Tconv cell imbalance would have therapeutic benefit.31 This review presents a historical overview and survey of experience with in vivo Treg expansion and associated changes in their functional capacity. Pre-clinical and clinical studies designed to augment Treg levels and function in vivo examining therapeutic benefit in the setting of GvHD prevention and therapy will be discussed.
Targeting cell surface receptors for in vivo Treg expansion, function and therapeutic application
Experimentally, a number of molecules expressed on Tregs have been shown to expand natural Tregs and/or augment their functional activity in vivo, including IL-2, several members of the TNFR family (TNFR2/TNF; TNFRSF25/TL1A; OX-40/OX-40L; 4-1BB/4-1BB-L) as well as CD28 and IL-33 to suppress autoimmune responses, allograft rejection and GvHD (Table 1 and Figure 1). While other molecules have been found to modulate Tregs in vivo (e.g. CD45, GITR/GITRL), these are not discussed here because they have not been assessed in GvHD.3432
IL-2 / CD25
Over the past decade, IL-2 treatment has been the approach most extensively utilized in pre-clinical and clinical settings to expand Tregs in vivo to ameliorate GvHD. Therapeutic strategies have varied the reagents, timing of administration and targeting donor/recipient populations (Table 1).
IL-2 is a pleiotropic cytokine which plays a dual role in maintaining tolerance and contributing to immunity in vivo.35 Tregs but not Tconv cells constitutively express high CD25 levels, although Tconv up-regulate this receptor after activation. Importantly, downstream signaling in Tregs versus Tconv is more sensitive to IL-2 stimulation.36 Accordingly, high doses (HD) of IL-2 can target CD4 effector cells and stimulate immunity whereas low-dose (LD; 100-fold lower) IL-2 selectively activates Tregs, promoting tolerance.36 Human recombinant IL-2 was first approved by the US Food and Drug Administration (FDA) in 1998 for use at HD to stimulate immunity toward metastatic cancers (renal cell carcinoma and melanoma).37 LD IL-2 has minimal side effects38 and, together with its effects on in vivo Treg expansion, is of interest for tolerance induction.
Multiple studies demonstrated that free LD IL-2 treatment results in Treg expansion leading to efficient reversion of autoimmune type 1 diabetes (T1D),39 amelioration of experimental autoimmune encephalomyelitis (EAE)40 and improved long-term allograft survival in a corneal transplant model.41 These findings led to combination therapy with synergistic effects on Treg expansion using free LD IL-2 with sirolimus in transplant models, i.e. cornea42 and skin.43 Similar results in combination with dexamethasone (Dex) were observed in EAE.44
To increase circulating IL-2 half-life and decrease the required dose, antibody/cytokine (a-IL-2/IL-2) complexes (IL-2C) are under development. Notably, in vivo utilization of different IL-2C resulted in targeting specific cell subsets, i.e. mAbJES-1A12/IL-2 and S4B6/IL-2 preferentially expanded Tregs and Tconv, respectively.45 The IL-2C (mAbJES-1A12+IL-2) caused selective Treg expansion and suppression of allergic airway inflammation,46 contact hypersensitivity,47 and experimental myasthenia gravis.48 Furthermore, in renal and cardiac ischemia reperfusion models, IL-2C expanded Tregs attenuated acute renal damage improving renal and myocardial recovery.5049 Using a combination therapy of IL-2C, sirolimus and islet Ag peptide, protection against spontaneous and induced T1D in NOD mice occurred following increased Treg levels and function.51 Recombinant adeno-associated viral vector that continuously releases IL-2 achieved persistent and sufficient levels of LD IL-2 while avoiding toxic effects.5352 Importantly, this viral vector controlled diabetes after sustained Treg expansion without impairing immune responses to infection, vaccination and cancer. Recently, an IL-2 modification creating a fusion protein between murine IL-2 and CD2554 achieved markedly extended half-life [16 hours vs. free IL-2 (<15 minutes)] and selective Treg expansion controlling T1D. Other examples of long-lived IL-2 fusion protein (FP) include: IgG-IL2 FP55 and IL-2+IL-33 (IL-233),56 discussed below.
Adoptive transfer of Tregs is a promising therapy to diminish GvHD. In humans, development of cGvHD is associated with poor Treg reconstitution post HSCT.5857 We and others have been examining the application of donor Tregs as a prophylactic strategy to prevent the development of GvHD in experimental models.6559 Experiments demonstrated that donor Tregs inhibit lethal acute GvHD (aGvHD) only at high ratios, i.e. 1:1 (Treg:Tconv).59 Because circulating Tregs account for only approximately 5-10% of CD4 T cells, a practical obstacle is collecting sufficient numbers of Tregs from donors or recipients for use in allogeneic hematopoietic stem cell transplantation (aHSCT) to suppress activation of anti-host reactive Tconv in T-cell-replete grafts, thus in vivo expansion is attractive.
In the early 1990s, using a fully MHC-mismatched bone marrow transplant (BMT) model, studies demonstrated that short-term human recombinant IL-2 administration (50,000 U twice daily) starting the day of the BMT significantly reduced GvHD mortality.66 Moreover, IL-2 treatment did not prevent allogeneic engraftment and preserved graft-versus-leukemia (GvL) effect. The IL-2 effect on GvHD but not on GvL was explained by selective inhibition of CD4-mediated activity.66 Clinical and immunological effects of IL-2 administration in patients following TCD allogeneic and autologous BMT were evaluated. Strikingly, patients who received TCD-BM and LD IL-2 (2-6×10 U/m daily for 90 days) to enhance GvL exhibited high circulating NK cells.6867 Since CD25 Tregs had not yet been identified, the success of these studies was not immediately attributed to Treg expansion. Later, discovery of this suppressive population69 and the recognition that their infusion inhibited immune responses opened a new era of GvHD prophylaxis and treatment.65646259 Indeed, the addition of IL-2 (6×10 IU/m daily) with donor CD4 T cells resulted in expansion of Tregs in patients post transplant.70 A seminal phase I dose-escalation study demonstrated that therapy of LD IL-2 daily for eight weeks in patients with steroid refractory active cGvHD was well tolerated and induced significant Treg expansion (Online Supplementary Table S1).71 This treatment diminished cGvHD manifestations, including decreased cutaneous sclerosis in a considerable number of patients.71 Notably, this IL-2 regimen induced selective activation of pStat5 in Tregs versus Tconv, which was associated with increased thymopoiesis and production of Tregs.72 These observations demonstrated that more nTreg homeostasis appeared to be restored following IL-2 therapy in patients with cGvHD.73
Ultra LD IL-2 (0.1-0.2×10 IU/m thrice weekly) was administered as aGvHD prophylaxis in pediatric patients after aHSCT starting <day 30 and continuing 6-12 weeks. Treg levels increased in recipients of matched related donor and the Treg functional suppressor activity was restored. IL-2 treated recipients had diminished humoral but not virus-specific cellular immune responses. Importantly, treated patients showed GvHD inhibition with no increase in leukemia/lymphoma relapse rates versus controls, suggesting that ultra LD IL-2 increased Tregs without impairing GvL.74 In contrast, when IL-2 (1×10 IU/m daily) was administered in a patient seven years post-GvHD onset, therapeutic effects were decreased.75 The patient experienced partial improvement of GvHD symptoms, Treg levels were increased after one week but declined despite continual IL-2 administration for approximately two months. A clinical trial randomized 90 subjects to determine whether LD IL-2 administration post transplant could reduce the incidence of both leukemia relapse and cGvHD. Patients were treated with LD IL-2 post-HSCT (day 60) for two weeks (followed by a 2-week hiatus) and compared with untreated controls. The treated group had a lower incidence of cGvHD, accompanied by a significant increase in GvHD-free and GvHD progression-free survival at three years, but administration did not decrease the incidence of leukemia relapse. Correlative studies demonstrated that circulating Treg and natural killer (NK) cells were increased in the IL-2 cohort during the treatment periods.76
In pre-clinical studies (Figure 2), treatment with LD IL-2C in MHC-matched HSCT recipients transplanted following reduced-intensity conditioning (RIC) regiments, administered either prior or following aHSCT, promoted expansion and activation of host Tregs within the first 7-10 days post transplant. Notably, host-versus-graft responses (HvG) were inhibited, resulting in enhanced donor chimerism and long-term hematopoietic cell engraftment.77 This work indicated for the first time that in vivo administration of IL-2C following BMT was a viable approach to expand host Tregs and consequently regulate alloimmunity following transplantation.78 In contrast, using an MHC-haploidentical aHSCT model, IL-2 administration post transplant for ten days did not prevent GvHD nor improve survival; comparable results were observed in a xenogeneic GvHD model.79 In a murine major-MHC mismatched aHSCT, no effect occurred when treating recipients with LD IL-2 (twice daily) alone. However, the concomitant administration of the mTOR inhibitor sirolimus (0.5 kg/mg daily) in the first week post transplant improved survival and diminished aGvHD. Importantly, in vivo treatment with IL-2 plus sirolimus resulted in higher donor Treg expansion versus either IL-2 or sirolimus alone. These donor-derived expanded Tregs comprised both expanded natural Tregs and increased conversion of induced Tregs.80 Initially in a human trial, superior Treg reconstitution and aGvHD prevention was observed in patients treated with the combination of sirolimus and tacrolimus versus peri-transplant methotrexate and tacrolimus.81 In order to improve Treg recovery and based on previous reports, LD IL-2 administration (3 times/wk for 90 days) post transplant was added to the sirolimus/tacrolimus treatment regimen in 20 patients (clinicaltrials.gov identifier: 01927120) (Online Supplementary Table S1). This phase II trial demonstrated augmented peripheral blood Treg reconstitution in the first month post HSCT in the LD IL-2 (200,000 IU/m) plus sirolimus/tacrolimus treated group compared to sirolimus/tacrolimus alone. However, this early Treg expansion was not maintained and did not ameliorate acute or cGvHD.82 National Institutes of Health clinical trials involving IL-2 and aHSCT are summarized (Online Supplementary Table S1).
Using an MHC-haploidentical aHSCT model, LD IL-2 treatment in donor mice was inadequate as a GvHD prevention strategy. Although IL-2 administration over five days elevated Treg levels in the graft, no improvement in recipient weight loss or survival was observed.79 These results were attributed to a <1:1 ratio of Tregs:Tconv using a LD IL-2 only Treg expansion strategy. Notably, when donor mice were treated with Dex (5 mg/kg daily) and LD IL-2 for three days, higher Treg levels were observed in comparison to treatment with either reagent alone. In a fully-MHC mismatched aHSCT, mice receiving Dex+IL-2 pretreated donor cells exhibited improvement in survival and diminution of aGvHD83 (Figure 2). We are currently not aware of any studies reporting the use of T cells from IL-2 treated HSCT human donors. A recent report noted that treatment of healthy individuals with ultra LD rhIL-2 (50,000-200,000 U/m/day) for five days elevated peripheral Tregs with augmented suppressive activity without detection of acute or long-term on/off-target effects.84
TNFR super family
The TNF family currently comprises 29 receptors and 19 ligands. In this section, we discuss those members reported to be capable of regulating the Treg compartment in vivo including TNFR2, OX-40, 4-1BB and TNFRSF25.85
TNFRSF1B (TNFR2)/TNF-α
TNFR2 is highly and constitutively expressed on murine Tregs and other immune cells and further up-regulated after activation. It is also found on some endothelial and cells of the nervous system whereas TNFR1 is ubiquitously expressed. TNF-α, the natural ligand, is a pleiotropic cytokine with dual function (pro- and anti-inflammatory) which exists in both membrane-bound and soluble forms. Membrane bound TNF-α preferentially interacts with TNFR2 and results in a suppressive function due to the lack of a cytoplasmic death domain. TNF-α/TNFR1 interactions lead to pro-inflammatory responses.8786 TNF-α/TNFR2 interactions reportedly promoted activation and expansion of murine Tregs in vivo.88 This interaction correlates with suppressive function and phenotypic stability of those Tregs.89 Furthermore, TNF-α expanded Tregs were protective against T1D90 as well as in the setting of infections91 and septic shock.88 Additionally, TNFR2-deficient Tregs lost their capacity to control colitis92 and EAE.93 Using a selective agonist to increase binding specificity and target only mouse TNFR2 (TNC-scTNF80) Treg expansion, reduced inflammation in a model of chronic inflammation94 and established arthritis were reported.95
Serum TNF-α levels are elevated in individuals with GvHD. Due to its pleiotropic nature, this cytokine and the balance between its corresponding receptors modulate both GvHD and GvL. TNF-α in vivo selectively enhanced proliferation and activation of Tregs versus Tconv and in vitro increased Treg CTLA-4 and TGF-β levels.96 Notably, “unfavorable” (low) numbers of donor TNF-α-primed Tregs (1:10 Treg:Tconv), decreased aGvHD and augmented survival in recipients of a fully MHC-mismatched aHSCT.96 This diminution of aGvHD promoted by TNF-α primed Tregs was further explored by other investigators demonstrating that this cytokine is produced by Tconv and the effect is dependent on TNFR2 expressed by Tregs.97 In vivo treatment of recipient mice before aHSCT with STAR2, a selective mouse TNFR2 agonist, resulted in expansion of radiation-resistant host Tregs concomitant with a reduction of aGvHD and increased survival. Importantly, STAR2 treatment did not interfere with the transplanted T-cell-mediated GvL or the immune response against infectious opportunistic (cytomegalocirus) pathogens.98
TNFRSF4 (OX-40)/TNFSF4 (OX-40-L)
OX-40 (CD134) is constitutively and highly expressed on murine Tregs and is up-regulated upon activation on CD4/CD8 Tconv cells and to a lesser extent on NK, NKT cells and neutrophils. OX40 ligand, (OX-40-L, CD252), is expressed on a number of different cell types including activated professional APCs, activated T cells, NK cells, mast cells and endothelial cells.99 A role for OX-40 in the development, homeostasis and suppressive activity of Tregs has been implicated from studies using young knockout (KO) OX-40 mice.101100
The role of OX-40 on Treg expansion and function remains controversial. The use of agonistic anti-OX-40 mAb (OX-86) or APCs over-expressing OX-40L resulted in clear but weak proliferation and expansion of mouse Tregs in vivo and enhanced suppressive capacity in models of colitis and EAE.104100 Addition of IL-2 together with anti-OX-40 mAb further amplified Treg expansion as well as suppressive activity in a heart transplant model.105 However, other studies where clone OX-86 was administered failed to induce Treg expansion and found inhibition of Treg function in models of skin grafts and cancer.107106
Two decades ago, inhibition of the OX-40/OX-40-L axis was found to diminish murine lethal aGvHD in recipients post aHSCT.108 Indeed, infusion of pre-treated Tregs with OX-86 or intraperitoneal (i.p.) injection of OX-86 resulted in the inhibition of Treg suppressive activity on GvHD development.109 Additionally, when an OX-40L blocking antibody (KY1005) was combined with sirolimus in a nonhuman primate GvHD model, synergistic inhibition of T-cell activation while preserving post-HSCT Treg reconstitution was observed.110 Importantly, KY1005/sirolimus GvHD prophylaxis resulted in long-term GvHD-free survival and significant control of aGvHD.110 Interfering with the OX-40L/OX-40 pathway in the setting of aHSCT did not reduce Treg reconstitution although impairment of this pathway resulted in impaired Treg development in young mice.101100
TNFRSF9 (4-1BB)/TNFSF9 (4-1BB-L)
Analogous to OX-40, 4-1BB (CD137) is constitutively expressed on Tregs and is up-regulated upon activation on CD4 and CD8, B, NK and myeloid cells. CD137-L is the only known intercellular ligand for CD137 and is expressed on APC after activation, although the extracellular domain of 4-1BB binds to fibronectin and galectin-9. 4-1BB co-stimulatory activity is well appreciated to promote proliferation and survival of CD8 T cells.112111 However, this pathway can also induce Treg proliferation in vivo, as shown by experiments wherein Tregs were coated with 4-1BBL-Fc prior to infusion.113 Accordingly, agonistic anti-4-1BB mAb was shown to enhance the numbers of Tregs and ameliorate or inhibit disease in several experimental models of autoimmunity including, T1D,114 colitis,115 EAE,116 and psoriasis.117
Several laboratories have investigated the implication of 4-1BB/4-1BBL interactions in GvHD and GvL. 4-1BB/4-1BB-L interactions increased GvHD-induced lethality and allogeneic bone marrow rejection mediated by either CD4 or CD8 donor T cells. Moreover, treatment with agonistic anti-4-1BB mAb augmented GvL effects of delayed lymphocyte infusion in an acute myeloid leukemia (AML) model by stimulating an allogeneic anti-tumor response.118 Blockade of 4-1BB-L in F1 mice ameliorated aGvHD, but aggravated cGvHD with high levels of IgE and anti-dsDNA IgG autoantibody.119 Furthermore, stimulation of 4-1BB using an agonistic mAb prevented cGvHD by inhibition of autoantibody production through activation induced donor CD4 T-cell death accompanied by diminished host B-cell activation and decreased autoantibody production.120 As noted above, it has been demonstrated that stimulation of 4-1BB promotes in vivo Treg proliferation and suppressive activity.113 Following anti-4-1BB mAb (3H3) in vivo treatment, IL-2 production by memory T cells was increased resulting in the induction of Treg expansion in an Ag-independent manner.121 Notably, in this haploidentical (parent-into-F1) aGvHD model, host Tregs survived long term. Furthermore, pre-conditioning with anti-4-1BB mAb ameliorated GvHD by increasing Treg suppressive activity against alloreactive donor T cells. When anti-4-1BB mAb-primed host Tregs were adoptively transferred into BDF1 mice, less severe disease was observed.121
TNFRSF25 (DR3)/TNFSF15 (TL1A)
TNFRSF25 is constitutively expressed on Tregs and up-regulated upon activation on CD4 and CD8 T cells. Low levels are also present on NK, NKT cells and ILC2/3 subsets. Its natural ligand, TL1A, is primarily expressed on endothelial cells and upon activation on APC and T cells, including Tregs.123122 TNFRSF25 co-stimulation promotes proliferation, effector function and survival, as well as apoptosis depending on signal strength.125124
Recent studies, using either agonistic mAb (clone 4C12) or a TL1A-Ig fusion protein, have shown that triggering the TNFRSF25 pathway in the absence of antigen leads to a significant expansion of Tregs in vivo which is dependent on the TCR and IL-2.127126 Notably, TNFRSF25 stimulation expands Tregs to a greater degree than CD25 stimulation (via IL-2) alone. Moreover, TNFRSF25 stimulation alone has the strongest in vivo effect on Treg expansion compared to other TNFRSF members including GITR, OX-40 or 4-1BB.126 Stimulating multiple receptors on Tregs can have synergistic effects, though the levels of expansion achieved depend on the molecular targets. Thus, targeting two TNF family members, TNFRSF25 (4C12) and OX-40 (OX-86)129128 in the context of co-stimulation or vaccination led to increased frequency of Tregs versus either alone. However, when both TNFRSF25 and CD25 are stimulated,130 there is a marked elevation of the compartment, much greater than administration of TL1A-Ig or IL-2 alone or 4C12 together with OX-86. It is likely that the former is observed because sufficient IL-2 is provided to maintain the elevated Treg levels.
TNFRSF25-induced expansion, either alone or in combination with IL-2, leads to upregulation of activation markers on Tregs and enhanced suppressive activity.132131 Our own studies have demonstrated that ICOS-1, Nrp-1, PD-1 and other molecules are more highly expressed after combined TNFRSF25 and CD25 in vivo stimulation compared with targeting the individual receptors.132 Tregs expanded in vivo via the TNFRSF25 are protective against allergic lung inflammation and EAE.133127126 Furthermore, they also prolonged graft survival in a mouse model of heterotopic allogeneic heart transplantation.134
As noted above, TNFRSF25 stimulation using 4C12 or TL1A-Ig selectively and dramatically promotes in vivo Treg expansion. Single administration of 4C12 resulted in a significant increase in splenic and lymph node Treg numbers with enhanced suppressive function.135 In a major MHC-mismatched GvHD model, recipients of 4C12-treated donor T cells showed a significant increase in overall survival and diminished GvHD.135130 Importantly, T cells from 4C12-treated donors exhibited preserved graft-versus-tumor (GvT) activity.135 Expansion of Tregs with 4C12 treatment induced phenotypic changes in Tregs, including higher expression of activation and maturation molecules.131 Mice treated with purified 4C12-Tregs (CD4CD25) showed diminished GvHD with improved survival, demonstrating that these Tregs possessed higher in vivo suppressive activity.131 Notably, administration of 4C12 into recipients with ongoing GvHD augmented mortality and worsened the disease because upon alloantigen exposure TNFRSF25 stimulation induced effector T-cell proliferation and activation.131 However, prophylaxis of recipients with 4C12 induced host-derived Treg expansion with a reduction in GvHD severity.131 Critically, the success of this approach to prevent GvHD is dependent on the timing of TNFRSF25 stimulation and the status of donor T-cell activation.
Because of the success of IL-2 in pre-clinical and clinical GvHD studies and the expression of TNFRSF25 in the Treg population, we developed a strategy to transiently manipulate Tregs in vivo combining LD IL-2C with TL1A-Ig. This “two-pathway” approach markedly expands (5-7-fold) and selectively activates Tregs. Transplantation of TL1A-Ig/IL-2C Treg expanded donor spleen into recipients resulted in amelioration of GvHD severity in both fully MHC-mismatched and MHC-matched aHSCT settings132130 (Figure 2). In vivo Treg expansion was superior with TL1A-Ig/IL-2C treatment compared to 4C12 mAb administration and GvHD was more completely ameliorated after transplant of two-pathway expanded donor Tregs. Furthermore, recipients of transplants using spleen cells from TL1A-Ig/IL-2 expanded donors demonstrated preserved GvL while GvHD was effectively diminished.130 In fact, because TL1A-Ig/IL-2 expanded Tregs expressed higher activation and functional molecules, very low numbers of these expanded cells (corresponding to a ratio of 1:6 expanded Tregs/Tconv) very effectively suppress GvHD post aHSCT.132 Furthermore, TL1A-Ig/IL-2 expanded Treg therapy was shown to be as effective as post-transplant cyclophosphamide for GvHD prophylaxis while more rapid thymic reconstitution providing earlier recovery of recipient immune function.136 GvHD is promoted by alloreactive donor T cells and inflammation; therefore, we proposed to regulate both pathways. Donor Treg expansion was combined with EP11313 (a bromodomain and extra-terminal bromodomain inhibitor, BETi) using short-term treatment in the recipient in a fully MHC-mismatched aHSCT. This strategy was found to significantly reduce early GvHD clinical scores including decreased ocular and skin involvement. Importantly, utilizing highly purified TL1A-Ig/IL-2 expanded donor Tregs in vivo, this second assessment of the combinatorial strategy further supported the notion that selective BETi can be used for GvHD treatment in combination with Treg adoptive therapy.137
CD28/CD80/86 (B7.1/2)
CD28, a key co-stimulatory molecule, is expressed on all T cells and provides signals for activation and survival. Its ligands, CD80 and CD86, are expressed on APC. CD28 super-agonists (CD28SA) are mAbs which induce poly-clonal T-cell proliferation in the absence of TCR ligation.139138 In rodents, in vivo application of CD28SA at low doses efficiently expands Tregs and partially inhibits expansion of autoreactive T cells. In addition, CD28SA expanded Tregs showed enhanced suppressive activity (increased IL-10 production) and migrated to inflamed tissues without causing cytokine release syndrome.142139 Treg expansion by CD28SA is dependent on paracrine IL-2 from CD28SA-stimulated Tconv cells.142 Accordingly, CD28SA expanded Tregs are highly effective in the treatment/prevention of disease in rodent models of autoimmunity, like EAE,141 experimental autoimmune neuritis143 and T1D,144 as well as inflammation, e.g. arthritis,146145 glomerulonephritis147 and trypanosomiasis-associated inflammation.148 Furthermore, CD28SA expanded Tregs prolong graft survival in experimental models of solid organ (renal/heart) transplantation.150149
A single dose of CD28SA (clone JJ316) administration in vivo to Lewis rats induced a selective 4-fold expansion of Tregs over Tconv. Adoptive transfer of anti-CD28 mAb treated splenocytes in a rat transplantation model reduced lethality and suppressed GvHD. Importantly, this therapeutic effect was mediated by expanded Tregs detected in high levels after transfer of anti-CD28 mAb-treated lymphocytes.151 Moreover, this mAb was utilized to treat F1 rat recipients at different time-points pre- or post aHSCT. GvHD mortality was absent or suppressed when recipients were treated with anti-CD28, three days before T-cell transfer or on day 0, respectively. In contrast, no protective effect was observed in recipients treated on day 7 or 10. Importantly, GvHD lethality reduction was mediated by anti-CD28SA selective Treg expansion in an antigen-specific manner.152 In a murine major-MHC mismatched aHSCT model, adoptive transfer of unfractionated polyclonal activated donor Tregs, with CD28SA (D665) significantly reduced GvHD-associated clinical signs and histopathological changes. Notably, this mAb administered to recipients or in vitro pre-stimulated donor T cells conserved a potent anti-lymphoma (GvL) response.153 The CD28SA in vitro approach may have clinical potential for patients because an anti-human CD28 mAb (TGN1412) induced a cytokine storm during the first in human clinical study.154
IL-33/ST2
IL-33, an IL-1 family member, is constitutively expressed in epithelial and endothelial cells at barrier sites where it functions as an endogenous danger signal in response to tissue damage. IL-33 is also a pleiotropic cytokine found in fibroblastic reticular cells of secondary lymphoid organs. It binds to the ubiquitously expressed IL-1R accessory protein (IL-1RAcP) and the more selectively expressed receptor ST2. The ST2 receptor is constitutively expressed on innate (mast cells, ILC2s, eosinophils, basophils, NK cells) and adaptive immune cells (Tregs, Th2, NKT) and up-regulated upon activation on Th1 and cytotoxic T cells. A soluble form of ST2 can be produced by alternative splicing and serves as a decoy receptor to limit IL-33 signaling.156155 Notably, the quantitative differences in ST2 expression among different T-cell subsets potentially could lead to competition for IL-33. Tissue Tregs, which express constitutively high levels of ST2, could therefore sequester IL-33 from inflammatory cells which would give them an advantage over effector T cells in situations of limited IL-33. Soluble ST2 released from intestinal tissue has recently been identified as an important biomarker of GvHD that is highly predictive for early post-HSCT mortality.157
Several reports have shown that administration of IL-33 leads to a ST2 dependent expansion of Tregs in vivo.159158 ST2 Tregs represent an activated subset of Tregs and are preferentially expressed in non-lymphoid tissues, like lung (20-30% of Treg), gastrointestinal tract (GI) (approx. 20%), and liver (50-60%).162160 ST2 Tregs exhibit superior suppressive function compared to ST2 Tregs.161 In a model of allogeneic heart transplantation, IL-33 expanded ST2 Tregs migrated to the graft and prolonged survival.164163 Furthermore, in a model of collagen induced rheumatoid arthritis, IL-33 expanded ST2 Tregs suppressed clinical and histological signs of arthritis.165 In addition, IL-33 signaling is apparently crucial for liver Treg expansion, accumulation and suppression of infection after murine CMV infection.166 A combination of IL-33 together with IL-2 administration has a synergistic effect with regard to Treg expansion.56 This group also generated a hybrid fusion protein between those 2 cytokines, IL-233, which bears the activities of both. IL-233 treatment significantly increased the number of Tregs in blood, spleen and renal compartments and prevented ischemia reperfusion injury more efficiently than a mixture of IL-2 and IL-33.
In aHSCT, the IL-33/ST2 axis has recently emerged as a novel therapeutic target for GvHD. In particular, high levels of IL-33 and ST2 were detected in recipients post conditioning, contributing to lethal aGvHD via donor TH1 alloimmune responses.167 Therefore, transplants involving either IL-33 recipients, ST2 donor T cells or IL-33 antagonist (ST2-Fc fusion protein) administration resulted in increased recipient survival.167 Conversely, in vivo administration of peri-aHSCT IL-33 (day -10 to day 4) to recipients induced a diminution of clinical GvHD scores and prolonged survival. This protection against GvHD was promoted by IL-33-expanded recipient ST2 Tregs (persisted after total body irradiation) which controlled M1 macrophage activation and reduced effector T-cell levels. The underlying mechanism of IL-33/ST2-induced Treg proliferation was mediated by activation of p38 MAPK.168 Subsequent studies found that transfer of ST2 donor Tregs affect diminished protection of GvHD after an MHC-mismatched aHSCT.167 We are unaware of any ongoing clinical trials regulating the IL-33/ST2 pathway in vivo to manipulate Tregs.
“Unintentional” effects on the Treg compartment: potential impact in hematopoietic stem cell transplantation
While direct targeting of molecules on the Treg cell surface can induce their expansion, unintentional but clinically useful procedures in HSCT including extracorporeal photopheresis (ECP),169 donor stem cell mobilization,172170 PTCy,174173 azacytidine,175 JAK1/2 inhibitors177176 and ROCK1/2 inhibitors179178 also affect the Treg compartment although to a lesser extent than intentional interventions. Although Tregs have demonstrated promising results in regulating GvHD, combining their regulatory activity with other strategies being used in the clinic may further improve transplant outcomes for patients. Therefore, if strategies partially diminish or do not interfere with Tregs, augmenting the compartment should be useful. For example, in addition to alloreactive Tconv deletion, Tregs are reportedly needed for optimal PTCy (day 3-4) GvHD prophylaxis.181180 Treg expansion subsequent to PTCy may, therefore, augment the effectiveness of this reagent. In contrast, if strategies elevate Tregs this should provide additional benefit and further enable synergistic expansion. For example, azacytidine (AzaC), like PTCy, preferentially inhibits Teff versus Treg proliferation, but additionally converts Teffs to Tregs via hypomethylation of the Foxp3 promoter.175 Pre-clinical and clinical studies have demonstrated that post ECP, Treg levels in mouse and humans are elevated, suggesting that these effects on the Tregs might be a primary mechanism underlying the relative success of this clinical approach.169 Regarding stem cell mobilization, studies in primates and healthy HSCT donors showed an increase in Treg frequency using plerix-afor.172171 Development of balanced JAK1/JAK2 targeted inhibitors has led to promising clinical results in aHSCT.176 Notably, baricitinib is significantly more potent than ruxolitinib in preventing GvHD and demonstrates high Treg expansion by preserving the JAK3-STAT5 IL-2R pathway.177 Additionally, the Rho kinase inhibitors which down-regulate ROCK1/ROCK2 inhibiting inflammation have also shown promise in suppressing GvHD.179178 ROCK2 inhibition leads to pSTAT5 upregulation inducing an increase in Tregs.
Cellular “cross-talk” with Tregs can also result in an expansion of this population. For example, host iNKT cells were shown to induce in vivo donor Treg proliferation via IL-4 while preventing lethal aGvHD in mice.182 Subsequently, reports showed that donor iNKT cells diminished aGvHD and cGvHD through the expansion of donor Tregs.184183 These approaches are currently being examined in clinical trials of a-galactosylceramide, which has been shown to increase numbers of Tregs in model systems by expanding iNKT cells.185
Future perspectives
Approaches manipulating Tregs in vivo continue to advance. Systemic administration of IL-2 and other compounds can elevate the peripheral compartment for extended time-periods without apparent alteration of global immune function. Notably, not all reagents have equivalent efficacy in inducing and maintaining Treg expansion, which is a result of the specific receptor targeted and reagent persistence. For example, administration of modified IL-2 compared with free IL-2 results in greater Treg frequency in large measure due to prolonging the half-life of the cytokine.18618045 This has fostered increasing numbers of novel IL-2 constructs including fusokines, muteins and IL-2/receptor fusion proteins.18818718154 A recent study reported human IL-2 complexed to a human anti-IL2 mAb with in vivo enhancing activity.189 The development of highly CD25 specific signaling reagents with increased persistence suggests in vivo delivery to selectively target Tregs versus Tconv may lead to more effective therapeutic application during disorders where effector cells are present.
To date, approaches targeting Tregs have focused primarily on systemic (vs. local) homeostasis. Local manipulation of Tregs might provide an effective strategy for the treatment of widespread (systemic lupus erythematosus, etc.) as well as regional (e.g. GI diseases, encephalitis) inflammation. Treg expansion in the conjunctiva/ocular adnexa can be induced targeting CD25,41 or more potently through CD25 and TNFRSF25 (Copsel et al., unpublished data, 2019). Such strategies may be useful to treat ocular GvHD and uveitis. In the context of HSCT, conditioning using targeted total lymphoid irradiation promotes selective survival of Tregs locally, for example the GI. Subsequent administration of reagents may therefore “selectively” manipulate local intestinal Tregs which could be particularly effective in inhibiting GvHD.191190
Allogeneic hematopoietic stem cell transplantation is primarily administered to patients with hematologic malignancies. There has been increasing usage of checkpoint inhibitors (CI) including PD-1 for the treatment of these cancers and their effect on Tregs remains controversial. Studies have suggested that the PD-L1/PD-1 pathway drives Treg stability/function and expansion.1931926 while others have reported Treg inhibition.195194 Clinical improvement of experimental GvHD was associated with increased PD-1 levels on Tregs consistent with the hypothesis that this pathway promotes Treg-mediated tolerance.196 Stimulation of PD-1 on Tregs in hematologic tumors have been reported to promote the inhibition of effector T cells, reducing anti-tumor immune responses.193 Although early reports noted that severe GvHD may result from administering pembrolizumab post HSCT in patients with hematologic malignancies,197 evaluation using CI after PTCy deletion of alloreactive T cells should be considered. In addition to promoting CD8 anti-tumor specific T cells, the success of this therapeutic approach may also depend on direct effects of PD-1 inhibitors on Tregs, since increasing or decreasing their numbers/function could positively or negatively modulate anti-tumor immunity. Taken together, since no single prophylactic treatment including adoptive Treg transfer is likely to abolish GvHD, we suggest combining in vivo Treg expansion strategies with promising reagents being translated to clinical aHSCT is feasible and could provide a significant advance in the field.
Conclusions
The Treg compartment can now be manipulated in vivo as a consequence of intentionally targeting identified receptors. Pre-clinical and clinical studies contributed by a large number of laboratories have directly targeted these cells and reported the ability to efficiently augment their numbers and function in vivo. While there is an increasing number of molecules which effectively expand Tregs, for the moment IL-2 is the only FDA-approved compound in use in GvHD clinical trials. It is not surprising that efforts to improve in vivo IL-2 efficacy through the generation of modified IL-2 molecules are currently underway. Other strategies stimulating different signaling pathways promoting Tregs have shown promise in preclinical models. Targeting TNF receptors for example, TNFRSF25 alone or together with LD IL-2 has demonstrated extremely potent Treg expansion and improved function. Combination approaches should, therefore, be investigated for potential clinical application.
Acknowledgments
SC: support from a Sylvester Comprehensive Cancer Center research grant for post-doctoral trainees; KVK support from: Kalish Family Foundation, Applebaum Foundation and the Sylvester Comprehensive Cancer Center; RBL support from: the Sylvester Comprehensive Cancer Center, NIH (RO1 EY024484-01) and an SRA from Heat Biologics, Inc. and Pelican Therapeutics. Due to space limitations, the authors sincerely regret not being able to cite all of our colleagues’ publications contributing to this field.
Footnotes
- ↵* SC and DW contributed equally to this work.
- Check the online version for the most updated information on this article, online supplements, and information on authorship & disclosures: www.haematologica.org/content/104/7/1309
- Received February 4, 2019.
- Accepted May 7, 2019.
References
- Sakaguchi S, Sakaguchi N, Shimizu J. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev. 2001; 182:18-32. PubMedhttps://doi.org/10.1034/j.1600-065X.2001.1820102.xGoogle Scholar
- Malek TR, Yu A, Vincek V, Scibelli P, Kong L. CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2. Immunity. 2002; 17(2):167-178. PubMedhttps://doi.org/10.1016/S1074-7613(02)00367-9Google Scholar
- Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007; 8(2):191-197. PubMedhttps://doi.org/10.1038/ni1428Google Scholar
- Piccirillo CA, Shevach EM. Naturally-occurring CD4+CD25+ immunoregulatory T cells: central players in the arena of peripheral tolerance. Semin Immunol. 2004; 16(2):81-88. PubMedhttps://doi.org/10.1016/j.smim.2003.12.003Google Scholar
- Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on IL-2. Nat Rev Immunol. 2004; 4(9):665-674. PubMedhttps://doi.org/10.1038/nri1435Google Scholar
- Wang HY, Lee DA, Peng G. Tumor-specific human CD4+ regulatory T cells and their ligands: implications for immunotherapy. Immunity. 2004; 20(1):107-118. PubMedhttps://doi.org/10.1016/S1074-7613(03)00359-5Google Scholar
- Sato E, Olson SH, Ahn J. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005; 102(51):18538-18543. PubMedhttps://doi.org/10.1073/pnas.0509182102Google Scholar
- Gao Q, Qiu SJ, Fan J. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J Clin Oncol. 2007; 25(18):2586-2593. PubMedhttps://doi.org/10.1200/JCO.2006.09.4565Google Scholar
- Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012; 12(4):298-306. PubMedhttps://doi.org/10.1038/nrc3245Google Scholar
- Galon J, Mlecnik B, Bindea G. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. J Pathol. 2014; 232(2):199-209. PubMedhttps://doi.org/10.1002/path.4287Google Scholar
- Mlecnik B, Bindea G, Angell HK. Integrative Analyses of Colorectal Cancer Show Immunoscore Is a Stronger Predictor of Patient Survival Than Microsatellite Instability. Immunity. 2016; 44(3):698-711. PubMedhttps://doi.org/10.1016/j.immuni.2016.02.025Google Scholar
- Liu W, Putnam AL, Xu-Yu Z. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006; 203(7):1701-1711. PubMedhttps://doi.org/10.1084/jem.20060772Google Scholar
- Seddiki N, Santner-Nanan B, Martinson J. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med. 2006; 203(7):1693-1700. PubMedhttps://doi.org/10.1084/jem.20060468Google Scholar
- Di Ianni M, Falzetti F, Carotti A. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood. 2011; 117(14):3921-3928. PubMedhttps://doi.org/10.1182/blood-2010-10-311894Google Scholar
- Brunstein CG, Miller JS, McKenna DH. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect. Blood. 2016; 127(8):1044-1051. PubMedhttps://doi.org/10.1182/blood-2015-06-653667Google Scholar
- Tang Q, Henriksen KJ, Bi M. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med. 2004; 199(11):1455-1465. PubMedhttps://doi.org/10.1084/jem.20040139Google Scholar
- 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(20):5671-5680. PubMedhttps://doi.org/10.1182/blood-2011-02-337097Google Scholar
- Hippen KL, Merkel SC, Schirm DK. Massive ex vivo expansion of human natural regulatory T cells (T(regs)) with minimal loss of in vivo functional activity. Sci Transl Med. 2011; 3(83):83ra41. PubMedhttps://doi.org/10.1126/scitranslmed.3001809Google Scholar
- Hippen KL, Harker-Murray P, Porter SB. Umbilical cord blood regulatory T-cell expansion and functional effects of tumor necrosis factor receptor family members OX40 and 4-1BB expressed on artificial antigen-presenting cells. Blood. 2008; 112(7):2847-2857. PubMedhttps://doi.org/10.1182/blood-2008-01-132951Google Scholar
- Putnam AL, Brusko TM, Lee MR. Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes. 2009; 58(3):652-662. PubMedhttps://doi.org/10.2337/db08-1168Google Scholar
- Bluestone JA, Buckner JH, Fitch M. Type 1 diabetes immunotherapy using poly-clonal regulatory T cells. Sci Transl Med. 2015; 7(315):315ra189. PubMedhttps://doi.org/10.1126/scitranslmed.aad4134Google Scholar
- Wang X, Riviere I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. 2016; 3:16015. Google Scholar
- Campbell A, Brieva T, Raviv L. Concise Review: Process Development Considerations for Cell Therapy. Stem Cells Transl Med. 2015; 4(10):1155-1163. PubMedhttps://doi.org/10.5966/sctm.2014-0294Google Scholar
- Gee AP. Manufacturing genetically modified T cells for clinical trials. Cancer Gene Ther. 2015; 22(2):67-71. Google Scholar
- Levings MK, Sangregorio R, Roncarolo MG. Human cd25(+)cd4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function. J Exp Med. 2001; 193(11):1295-1302. PubMedhttps://doi.org/10.1084/jem.193.11.1295Google Scholar
- Esensten JH, Muller YD, Bluestone JA, Tang Q. Regulatory T cell therapy for autoimmune and autoinflammatory diseases: the next frontier. J Allergy Clin Immunol. 2018; 142(6):1710-1718. Google Scholar
- Putnam AL, Safinia N, Medvec A. Clinical grade manufacturing of human alloantigen-reactive regulatory T cells for use in transplantation. Am J Transplant. 2013; 13(11):3010-3020. PubMedhttps://doi.org/10.1111/ajt.12433Google Scholar
- Seay HR, Putnam AL, Cserny J. Expansion of Human Tregs from Cryopreserved Umbilical Cord Blood for GMP-Compliant Autologous Adoptive Cell Transfer Therapy. Mol Ther Methods Clin Dev. 2017; 4:178-191. PubMedhttps://doi.org/10.1016/j.omtm.2016.12.003Google Scholar
- McKenna DH, Sumstad D, Kadidlo DM. Optimization of cGMP purification and expansion of umbilical cord blood-derived T-regulatory cells in support of first-in-human clinical trials. Cytotherapy. 2017; 19(2):250-262. Google Scholar
- Heinrichs J, Bastian D, Veerapathran A. Regulatory T-Cell Therapy for Graft-versus-host Disease. J Immunol Res Ther. 2016; 1(1):1-14. Google Scholar
- Alho AC, Kim HT, Chammas MJ. Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. Blood. 2016; 127(5):646-657. PubMedhttps://doi.org/10.1182/blood-2015-10-672345Google Scholar
- Gagliani N, Gregori S, Jofra T. Rapamycin combined with anti-CD45RB mAb and IL-10 or with G-CSF induces tolerance in a stringent mouse model of islet transplantation. PLoS One. 2011; 6(12):e28434. PubMedhttps://doi.org/10.1371/journal.pone.0028434Google Scholar
- Camirand G, Wang Y, Lu Y. CD45 ligation expands Tregs by promoting interactions with DCs. J Clin Invest. 2014; 124(10):4603-4613. Google Scholar
- Ronchetti S, Ricci E, Petrillo MG. Glucocorticoid-induced tumour necrosis factor receptor-related protein: a key marker of functional regulatory T cells. J Immunol Res. 2015; 2015:171520. Google Scholar
- Malek TR. The biology of interleukin-2. Annu Rev Immunol. 2008; 26:453-479. PubMedhttps://doi.org/10.1146/annurev.immunol.26.021607.090357Google Scholar
- Yu A, Snowhite I, Vendrame F. Selective IL-2 responsiveness of regulatory T cells through multiple intrinsic mechanisms supports the use of low-dose IL-2 therapy in type 1 diabetes. Diabetes. 2015; 64(6):2172-2183. PubMedhttps://doi.org/10.2337/db14-1322Google Scholar
- Lentsch AB, Miller FN, Edwards MJ. Mechanisms of leukocyte-mediated tissue injury induced by interleukin-2. Cancer Immunol Immunother. 1999; 47(5):243-248. PubMedhttps://doi.org/10.1007/s002620050527Google Scholar
- Yang JC, Sherry RM, Steinberg SM. Randomized study of high-dose and low-dose interleukin-2 in patients with metastatic renal cancer. J Clin Oncol. 2003; 21(16):3127-3132. PubMedhttps://doi.org/10.1200/JCO.2003.02.122Google Scholar
- Grinberg-Bleyer Y, Baeyens A, You S. IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J Exp Med. 2010; 207(9):1871-1878. PubMedhttps://doi.org/10.1084/jem.20100209Google Scholar
- Rouse M, Nagarkatti M, Nagarkatti PS. The role of IL-2 in the activation and expansion of regulatory T-cells and the development of experimental autoimmune encephalomyelitis. Immunobiology. 2013; 218(4):674-682. PubMedhttps://doi.org/10.1016/j.imbio.2012.08.269Google Scholar
- Tahvildari M, Omoto M, Chen Y. In Vivo Expansion of Regulatory T Cells by Low-Dose Interleukin-2 Treatment Increases Allograft Survival in Corneal Transplantation. Transplantation. 2016; 100(3):525-532. PubMedhttps://doi.org/10.1097/TP.0000000000001044Google Scholar
- Wang X, Wang W, Xu J, Le Q. Effect of rapamycin and interleukin-2 on regulatory CD4+CD25+Foxp3+ T cells in mice after allogenic corneal transplantation. Transplant Proc. 2013; 45(2):528-537. PubMedhttps://doi.org/10.1016/j.transproceed.2012.06.064Google Scholar
- Pilon CB, Petillon S, Naserian S. Administration of low doses of IL-2 combined to rapamycin promotes allogeneic skin graft survival in mice. Am J Transplant. 2014; 14(12):2874-2882. PubMedhttps://doi.org/10.1111/ajt.12944Google Scholar
- Chen X, Oppenheim JJ, Winkler-Pickett RT, Ortaldo JR, Howard OM. Glucocorticoid amplifies IL-2-dependent expansion of functional FoxP3(+)CD4(+)CD25(+) T regulatory cells in vivo and enhances their capacity to suppress EAE. Eur J Immunol. 2006; 36(8):2139-2149. PubMedhttps://doi.org/10.1002/eji.200635873Google Scholar
- Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science. 2006; 311(5769):1924-1927. PubMedhttps://doi.org/10.1126/science.1122927Google Scholar
- Wilson MS, Pesce JT, Ramalingam TR. Suppression of murine allergic airway disease by IL-2:anti-IL-2 monoclonal antibody-induced regulatory T cells. J Immunol. 2008; 181(10):6942-6954. PubMedhttps://doi.org/10.4049/jimmunol.181.10.6942Google Scholar
- El Beidaq A, Link CW, Hofmann K. In Vivo Expansion of Endogenous Regulatory T Cell Populations Induces Long-Term Suppression of Contact Hypersensitivity. J Immunol. 2016; 197(5):1567-1576. PubMedhttps://doi.org/10.4049/jimmunol.1600508Google Scholar
- Liu R, Zhou Q, La Cava A. Expansion of regulatory T cells via IL-2/anti-IL-2 mAb complexes suppresses experimental myasthenia. Eur J Immunol. 2010; 40(6):1577-1589. PubMedhttps://doi.org/10.1002/eji.200939792Google Scholar
- Kim MG, Koo TY, Yan JJ. IL-2/anti-IL-2 complex attenuates renal ischemia-reperfusion injury through expansion of regulatory T cells. J Am Soc Nephrol. 2013; 24(10):1529-1536. PubMedhttps://doi.org/10.1681/ASN.2012080784Google Scholar
- Xiao J, Yu K, Li M. The IL-2/Anti-IL-2 Complex Attenuates Cardiac Ischaemia-Reperfusion Injury Through Expansion of Regulatory T Cells. Cell Physiol Biochem. 2017; 44(5):1810-1827. Google Scholar
- Manirarora JN, Wei CH. J Immunol. 2015; 195(11):5203-5214. PubMedhttps://doi.org/10.4049/jimmunol.1402540Google Scholar
- Churlaud G, Jimenez V, Ruberte J. Sustained stimulation and expansion of Tregs by IL2 control autoimmunity without impairing immune responses to infection, vaccination and cancer. Clin Immunol. 2014; 151(2):114-126. PubMedhttps://doi.org/10.1016/j.clim.2014.02.003Google Scholar
- Johnson MC, Garland AL, Nicolson SC. beta-cell-specific IL-2 therapy increases islet Foxp3+Treg and suppresses type 1 diabetes in NOD mice. Diabetes. 2013; 62(11):3775-3784. PubMedhttps://doi.org/10.2337/db13-0669Google Scholar
- Ward NC, Yu A, Moro A. IL-2/CD25: A Long-Acting Fusion Protein That Promotes Immune Tolerance by Selectively Targeting the IL-2 Receptor on Regulatory T Cells. J Immunol. 2018; 201(9):2579-2592. PubMedhttps://doi.org/10.4049/jimmunol.1800907Google Scholar
- Bell CJ, Sun Y, Nowak UM. Sustained in vivo signaling by long-lived IL-2 induces prolonged increases of regulatory T cells. J Autoimmun. 2015; 56:66-80. PubMedhttps://doi.org/10.1016/j.jaut.2014.10.002Google Scholar
- Stremska ME, Jose S, Sabapathy V. IL233, A Novel IL-2 and IL-33 Hybrid Cytokine, Ameliorates Renal Injury. J Am Soc Nephrol. 2017; 28(9):2681-2693. PubMedhttps://doi.org/10.1681/ASN.2016121272Google Scholar
- Miura Y, Thoburn CJ, Bright EC. Association of Foxp3 regulatory gene expression with graft-versus-host disease. Blood. 2004; 104(7):2187-2193. PubMedhttps://doi.org/10.1182/blood-2004-03-1040Google Scholar
- Zorn E, Kim HT, Lee SJ. Reduced frequency of FOXP3+ CD4+CD25+ regulatory T cells in patients with chronic graft-versus-host disease. Blood. 2005; 106(8):2903-2911. PubMedhttps://doi.org/10.1182/blood-2005-03-1257Google Scholar
- Hoffmann P, Ermann J, Edinger M, Fathman CG, Strober S. Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med. 2002; 196(3):389-399. PubMedhttps://doi.org/10.1084/jem.20020399Google Scholar
- Cohen JL, Trenado A, Vasey D, Klatzmann D, Salomon BL. CD4(+)CD25(+) immunoregulatory T Cells: new therapeutics for graft-versus-host disease. J Exp Med. 2002; 196(3):401-406. PubMedhttps://doi.org/10.1084/jem.20020090Google Scholar
- Edinger M, Hoffmann P, Ermann J. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003; 9(9):1144-1150. PubMedhttps://doi.org/10.1038/nm915Google Scholar
- Trenado A, Charlotte F, Fisson S. Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J Clin Invest. 2003; 112(11):1688-1696. PubMedhttps://doi.org/10.1172/JCI200317702Google Scholar
- Jones SC, Murphy GF, Korngold R. Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol Blood Marrow Transplant. 2003; 9(4):243-256. PubMedhttps://doi.org/10.1053/bbmt.2003.50027Google Scholar
- 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(10):3493-3499. PubMedhttps://doi.org/10.1182/blood.V99.10.3493Google Scholar
- Hanash AM, Levy RB. Donor CD4+CD25+ T cells promote engraftment and tolerance following MHC-mismatched hematopoietic cell transplantation. Blood. 2005; 105(4):1828-1836. PubMedhttps://doi.org/10.1182/blood-2004-08-3213Google Scholar
- Sykes M, Romick ML, Hoyles KA, Sachs DH. In vivo administration of interleukin 2 plus T cell-depleted syngeneic marrow prevents graft-versus-host disease mortality and permits alloengraftment. J Exp Med. 1990; 171(3):645-658. PubMedhttps://doi.org/10.1084/jem.171.3.645Google Scholar
- Soiffer RJ, Murray C, Cochran K. Clinical and immunologic effects of prolonged infusion of low-dose recombinant interleukin-2 after autologous and T-cell-depleted allogeneic bone marrow transplantation. Blood. 1992; 79(2):517-526. PubMedGoogle Scholar
- Soiffer RJ, Murray C, Gonin R, Ritz J. Effect of low-dose interleukin-2 on disease relapse after T-cell-depleted allogeneic bone marrow transplantation. Blood. 1994; 84(3):964-971. PubMedGoogle Scholar
- 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(3):1151-1164. PubMedGoogle Scholar
- Zorn E, Mohseni M, Kim H. Combined CD4+ donor lymphocyte infusion and low-dose recombinant IL-2 expand FOXP3+ regulatory T cells following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2009; 15(3):382-388. PubMedhttps://doi.org/10.1016/j.bbmt.2008.12.494Google Scholar
- Koreth J, Matsuoka K, Kim HT. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med. 2011; 365(22):2055-2066. PubMedhttps://doi.org/10.1056/NEJMoa1108188Google Scholar
- Matsuoka K, Koreth J, Kim HT. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med. 2013; 5(179):179ra143. Google Scholar
- Matsuoka KI. Low-dose interleukin-2 as a modulator of Treg homeostasis after HSCT: current understanding and future perspectives. Int J Hematol. 2018; 107(2):130-137. Google Scholar
- Kennedy-Nasser AA, Ku S, Castillo-Caro P. Ultra low-dose IL-2 for GVHD prophylaxis after allogeneic hematopoietic stem cell transplantation mediates expansion of regulatory T cells without diminishing antiviral and antileukemic activity. Clin Cancer Res. 2014; 20(8):2215-2225. PubMedhttps://doi.org/10.1158/1078-0432.CCR-13-3205Google Scholar
- Kim N, Jeon YW, Nam YS. Therapeutic potential of low-dose IL-2 in a chronic GVHD patient by in vivo expansion of regulatory T cells. Cytokine. 2016; 78:22-26. PubMedhttps://doi.org/10.1016/j.cyto.2015.11.020Google Scholar
- Zhao XY, Zhao XS, Wang YT. Prophylactic use of low-dose interleukin-2 and the clinical outcomes of hematopoietic stem cell transplantation: A randomized study. Oncoimmunology. 2016; 5(12):e1250992. Google Scholar
- Shatry A, Levy RB. In situ activation and expansion of host tregs: a new approach to enhance donor chimerism and stable engraftment in major histocompatibility complex-matched allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2009; 15(7):785-794. PubMedGoogle Scholar
- Shatry A, Chirinos J, Gorin MA, Jones M, Levy RB. Targeting Treg cells in situ: emerging expansion strategies for (CD4(+)CD25(+)) regulatory T cells. Biol Blood Marrow Transplant. 2009; 15(10):1239-1243. PubMedhttps://doi.org/10.1016/j.bbmt.2009.06.009Google Scholar
- Perol L, Martin GH, Maury S, Cohen JL, Piaggio E. Potential limitations of IL-2 administration for the treatment of experimental acute graft-versus-host disease. Immunol Lett. 2014; 162(2 Pt B):173-184. PubMedhttps://doi.org/10.1016/j.imlet.2014.10.027Google Scholar
- Shin HJ, Baker J, Leveson-Gower DB. Rapamycin and IL-2 reduce lethal acute graft-versus-host disease associated with increased expansion of donor type CD4+CD25+Foxp3+ regulatory T cells. Blood. 2011; 118(8):2342-2350. PubMedhttps://doi.org/10.1182/blood-2010-10-313684Google Scholar
- Pidala J, Kim J, Jim H. A randomized phase II study to evaluate tacrolimus in com bination with sirolimus or methotrexate after allogeneic hematopoietic cell transplantation. Haematologica. 2012; 97(12):1882-1889. PubMedhttps://doi.org/10.3324/haematol.2012.067140Google Scholar
- Betts BC, Pidala J, Kim J. IL-2 promotes early Treg reconstitution after allogeneic hematopoietic cell transplantation. Haematologica. 2017; 102(5):948-957. PubMedhttps://doi.org/10.3324/haematol.2016.153072Google Scholar
- Xie Y, Wu M, Song R. A glucocorticoid amplifies IL-2-induced selective expansion of CD4(+)CD25(+)FOXP3(+) regulatory T cells in vivo and suppresses graft-versus-host disease after allogeneic lymphocyte transplantation. Acta Biochim Biophys Sin (Shanghai). 2009; 41(9):781-791. PubMedhttps://doi.org/10.1093/abbs/gmp067Google Scholar
- Ito S, Bollard CM, Carlsten M. Ultra-low dose interleukin-2 promotes immune-modulating function of regulatory T cells and natural killer cells in healthy volunteers. Mol Ther. 2014; 22(7):1388-1395. PubMedhttps://doi.org/10.1038/mt.2014.50Google Scholar
- Croft M, Benedict CA, Ware CF. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov. 2013; 12(2):147-168. PubMedhttps://doi.org/10.1038/nrd3930Google Scholar
- Salomon BL, Leclerc M, Tosello J. Tumor Necrosis Factor alpha and Regulatory T Cells in Oncoimmunology. Front Immunol. 2018; 9:444. Google Scholar
- Mancusi A, Piccinelli S, Velardi A, Pierini A. The Effect of TNF-αlpha on Regulatory T Cell Function in Graft-versus-Host Disease. Front Immunol. 2018; 9:356. Google Scholar
- Chen X, Baumel M, Mannel DN, Howard OM, Oppenheim JJ. Interaction of TNF with TNF receptor type 2 promotes expansion and function of mouse CD4+CD25+ T regulatory cells. J Immunol. 2007; 179(1):154-161. PubMedhttps://doi.org/10.4049/jimmunol.179.1.154Google Scholar
- Chen X, Subleski JJ, Kopf H. Cutting edge: expression of TNFR2 defines a maximally suppressive subset of mouse CD4+CD25+FoxP3+ T regulatory cells: applicability to tumor-infiltrating T regulatory cells. J Immunol. 2008; 180(10):6467-6471. PubMedhttps://doi.org/10.4049/jimmunol.180.10.6467Google Scholar
- Grinberg-Bleyer Y, Saadoun D, Baeyens A. Pathogenic T cells have a paradoxical protective effect in murine autoimmune diabetes by boosting Tregs. J Clin Invest. 2010; 120(12):4558-4568. PubMedhttps://doi.org/10.1172/JCI42945Google Scholar
- Myers L, Joedicke JJ, Carmody AB. IL-2-independent and TNF-αlpha-dependent expansion of Vbeta5+ natural regulatory T cells during retrovirus infection. J Immunol. 2013; 190(11):5485-5495. PubMedhttps://doi.org/10.4049/jimmunol.1202951Google Scholar
- Housley WJ, Adams CO, Nichols FC. Natural but not inducible regulatory T cells require TNF-αlpha signaling for in vivo function. J Immunol. 2011; 186(12):6779-6787. PubMedhttps://doi.org/10.4049/jimmunol.1003868Google Scholar
- Tsakiri N, Papadopoulos D, Denis MC, Mitsikostas DD, Kollias G. TNFR2 on non-haematopoietic cells is required for Foxp3+ Treg-cell function and disease suppression in EAE. Eur J Immunol. 2012; 42(2):403-412. PubMedhttps://doi.org/10.1002/eji.201141659Google Scholar
- Schmid T, Falter L, Weber S. Chronic Inflammation Increases the Sensitivity of Mouse Treg for TNFR2 Costimulation. Front Immunol. 2017; 8:1471. Google Scholar
- Lamontain V, Schmid T, Weber-Steffens D. Stimulation of TNF receptor type 2 expands regulatory T cells and ameliorates established collagen-induced arthritis in mice. Cell Mol Immunol. 2019; 16(1):65-74. Google Scholar
- Pierini A, Strober W, Moffett C. TNF-αlpha priming enhances CD4+FoxP3+ regulatory T-cell suppressive function in murine GVHD prevention and treatment. Blood. 2016; 128(6):866-871. PubMedhttps://doi.org/10.1182/blood-2016-04-711275Google Scholar
- Leclerc M, Naserian S, Pilon C. Control of GVHD by regulatory T cells depends on TNF produced by T cells and TNFR2 expressed by regulatory T cells. Blood. 2016; 128(12):1651-1659. PubMedhttps://doi.org/10.1182/blood-2016-02-700849Google Scholar
- Chopra M, Biehl M, Steinfatt T. Exogenous TNFR2 activation protects from acute GvHD via host T reg cell expansion. J Exp Med. 2016; 213(9):1881-1900. PubMedhttps://doi.org/10.1084/jem.20151563Google Scholar
- Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: Structure and function - What questions remain?. Mol Immunol. 2017; 83:13-22. Google Scholar
- Takeda I, Ine S, Killeen N. Distinct roles for the OX40-OX40 ligand interaction in regulatory and nonregulatory T cells. J Immunol. 2004; 172(6):3580-3589. PubMedhttps://doi.org/10.4049/jimmunol.172.6.3580Google Scholar
- Piconese S, Pittoni P, Burocchi A. A non-redundant role for OX40 in the competitive fitness of Treg in response to IL-2. Eur J Immunol. 2010; 40(10):2902-2913. PubMedhttps://doi.org/10.1002/eji.201040505Google Scholar
- Griseri T, Asquith M, Thompson C, Powrie F. OX40 is required for regulatory T cell-mediated control of colitis. J Exp Med. 2010; 207(4):699-709. PubMedhttps://doi.org/10.1084/jem.20091618Google Scholar
- Ruby CE, Yates MA, Hirschhorn-Cymerman D. Cutting Edge: OX40 agonists can drive regulatory T cell expansion if the cytokine milieu is right. J Immunol. 2009; 183(8):4853-4857. PubMedhttps://doi.org/10.4049/jimmunol.0901112Google Scholar
- Gri G, Piconese S, Frossi B. CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity. 2008; 29(5):771-781. PubMedhttps://doi.org/10.1016/j.immuni.2008.08.018Google Scholar
- Xiao X, Gong W, Demirci G. New insights on OX40 in the control of T cell immunity and immune tolerance in vivo. J Immunol. 2012; 188(2):892-901. PubMedhttps://doi.org/10.4049/jimmunol.1101373Google Scholar
- Piconese S, Valzasina B, Colombo MP. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med. 2008; 205(4):825-839. PubMedhttps://doi.org/10.1084/jem.20071341Google Scholar
- Kitamura N, Murata S, Ueki T. OX40 costimulation can abrogate Foxp3+ regulatory T cell-mediated suppression of antitumor immunity. Int J Cancer. 2009; 125(3):630-638. PubMedhttps://doi.org/10.1002/ijc.24435Google Scholar
- Tsukada N, Akiba H, Kobata T. Blockade of CD134 (OX40)-CD134L interaction ameliorates lethal acute graft-versus-host disease in a murine model of allogeneic bone marrow transplantation. Blood. 2000; 95(7):2434-2439. PubMedGoogle Scholar
- Valzasina B, Guiducci C, Dislich H. Triggering of OX40 (CD134) on CD4(+)CD25+ T cells blocks their inhibitory activity: a novel regulatory role for OX40 and its comparison with GITR. Blood. 2005; 105(7):2845-2851. PubMedhttps://doi.org/10.1182/blood-2004-07-2959Google Scholar
- Tkachev V, Furlan SN, Watkins B. Combined OX40L and mTOR blockade controls effector T cell activation while preserving Treg reconstitution after transplant. Sci Transl Med. 2017; 9(408)Google Scholar
- Sanchez-Paulete AR, Labiano S, Rodriguez-Ruiz ME. Deciphering CD137 (4-1BB) signaling in T-cell costimulation for translation into successful cancer immunotherapy. Eur J Immunol. 2016; 46(3):513-522. PubMedhttps://doi.org/10.1002/eji.201445388Google Scholar
- Myers LM, Vella AT. Interfacing T-cell effector and regulatory function through CD137 (4-1BB) co-stimulation. Trends Immunol. 2005; 26(8):440-446. PubMedhttps://doi.org/10.1016/j.it.2005.06.003Google Scholar
- Zheng G, Wang B, Chen A. The 4-1BB costimulation augments the proliferation of CD4+CD25+ regulatory T cells. J Immunol. 2004; 173(4):2428-2434. PubMedhttps://doi.org/10.4049/jimmunol.173.4.2428Google Scholar
- Irie J, Wu Y, Kachapati K, Mittler RS, Ridgway WM. Modulating protective and pathogenic CD4+ subsets via CD137 in type 1 diabetes. Diabetes. 2007; 56(1):186-196. PubMedhttps://doi.org/10.2337/db06-0793Google Scholar
- Lee J, Lee EN, Kim EY. Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of inflammatory bowel disease. Immunol Lett. 2005; 101(2):210-216. PubMedhttps://doi.org/10.1016/j.imlet.2005.06.001Google Scholar
- Kim YH, Choi BK, Shin SM. 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modu lating the balance between Th17 and regulatory T cells. J Immunol. 2011; 187(3):1120-1128. PubMedhttps://doi.org/10.4049/jimmunol.1002681Google Scholar
- Yoo JK, Choo YK, Kwak DH. Protective effects of agonistic anti-4-1BB antibody on the development of imiquimod-induced psoriasis-like dermatitis in mice. Immunol Lett. 2016; 178:131-139. Google Scholar
- Blazar BR, Kwon BS, Panoskaltsis-Mortari A. Ligation of 4-1BB (CDw137) regulates graft-versus-host disease, graft-versus-leukemia, and graft rejection in allogeneic bone marrow transplant recipients. J Immunol. 2001; 166(5):3174-3183. PubMedhttps://doi.org/10.4049/jimmunol.166.5.3174Google Scholar
- Nozawa K, Ohata J, Sakurai J. Preferential blockade of CD8(+) T cell responses by administration of anti-CD137 ligand monoclonal antibody results in differential effect on development of murine acute and chronic graft-versus-host diseases. J Immunol. 2001; 167(9):4981-4986. PubMedhttps://doi.org/10.4049/jimmunol.167.9.4981Google Scholar
- Kim J, Choi WS, La S. Stimulation with 4-1BB (CD137) inhibits chronic graft-versus-host disease by inducing activation-induced cell death of donor CD4+ T cells. Blood. 2005; 105(5):2206-2213. PubMedhttps://doi.org/10.1182/blood-2004-06-2080Google Scholar
- Kim J, Kim W, Kim HJ. Host CD25+CD4+Foxp3+ regulatory T cells primed by anti-CD137 mAbs inhibit graft-versus-host disease. Biol Blood Marrow Transplant. 2012; 18(1):44-54. PubMedhttps://doi.org/10.1016/j.bbmt.2011.09.004Google Scholar
- Richard AC, Ferdinand JR, Meylan F. The TNF-family cytokine TL1A: from lymphocyte costimulator to disease co-conspirator. J Leukoc Biol. 2015; 98(3):333-345. PubMedhttps://doi.org/10.1189/jlb.3RI0315-095RGoogle Scholar
- Bittner S, Knoll G, Ehrenschwender M. Death receptor 3 signaling enhances proliferation of human regulatory T cells. FEBS Lett. 2017; 591(8):1187-1195. Google Scholar
- Sidhu-Varma M, Shih DQ, Targan SR. Differential Levels of Tl1a Affect the Expansion and Function of Regulatory T Cells in Modulating Murine Colitis. Inflamm Bowel Dis. 2016; 22(3):548-559. Google Scholar
- Taraban VY, Slebioda TJ, Willoughby JE. Sustained TL1A expression modulates effector and regulatory T-cell responses and drives intestinal goblet cell hyperplasia. Mucosal Immunol. 2011; 4(2):186-196. PubMedhttps://doi.org/10.1038/mi.2010.70Google Scholar
- Schreiber TH, Wolf D, Tsai MS. Therapeutic Treg expansion in mice by TNFRSF 25 prevents allergic lung inflammation. J Clin Invest. 2010; 120(10):3629-3640. PubMedhttps://doi.org/10.1172/JCI42933Google Scholar
- Khan SQ, Tsai MS, Schreiber TH. Cloning, expression, and functional charac-terization of TL1A-Ig. J Immunol. 2013; 190(4):1540-1550. PubMedhttps://doi.org/10.4049/jimmunol.1201908Google Scholar
- Schreiber TH, Wolf D, Bodero M, Gonzalez L, Podack ER. T cell costimulation by TNFR superfamily (TNFRSF)4 and TNFRSF25 in the context of vaccination. J Immunol. 2012; 189(7):3311-3318. PubMedhttps://doi.org/10.4049/jimmunol.1200597Google Scholar
- Lambracht-Washington D, Rosenberg RN. Co-stimulation with TNF receptor super-family 4/25 antibodies enhances in-vivo expansion of CD4+CD25+Foxp3+ T cells (Tregs) in a mouse study for active DNA Abeta42 immunotherapy. J Neuroimmunol. 2015; 278:90-99. Google Scholar
- Wolf D, Barreras H, Bader CS. Marked in Vivo Donor Regulatory T Cell Expansion via Interleukin-2 and TL1A-Ig Stimulation Ameliorates Graft-versus-Host Disease but Preserves Graft-versus-Leukemia in Recipients after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2017; 23(5):757-766. Google Scholar
- Nishikii H, Kim BS, Yokoyama Y. DR3 signaling modulates the function of Foxp3+ regulatory T cells and the severity of acute graft-versus-host disease. Blood. 2016; 128(24):2846-2858. PubMedhttps://doi.org/10.1182/blood-2016-06-723783Google Scholar
- Copsel S, Wolf D, Kale B. Very Low Numbers of CD4(+) FoxP3(+) Tregs Expanded in Donors via TL1A-Ig and Low-Dose IL-2 Exhibit a Distinct Activation/Functional Profile and Suppress GVHD in a Preclinical Model. Biol Blood Marrow Transplant. 2018; 24(9):1788-1794. Google Scholar
- Madireddi S, Eun SY, Mehta AK. Regulatory T Cell-Mediated Suppression of Inflammation Induced by DR3 Signaling Is Dependent on Galectin-9. J Immunol. 2017; 199(8):2721-2728. PubMedhttps://doi.org/10.4049/jimmunol.1700575Google Scholar
- Wolf D, Schreiber TH, Tryphonopoulos P. Tregs expanded in vivo by TNFRSF25 agonists promote cardiac allograft survival. Transplantation. 2012; 94(6):569-574. PubMedGoogle Scholar
- Kim BS, Nishikii H, Baker J. Treatment with agonistic DR3 antibody results in expansion of donor Tregs and reduced graft-versus-host disease. Blood. 2015; 126(4):546-557. PubMedhttps://doi.org/10.1182/blood-2015-04-637587Google Scholar
- Wolf D, Bader CS, Barreras H. Superior immune reconstitution using Treg-expanded donor cells versus PTCy treatment in preclinical HSCT models. JCI Insight. 2018; 3(20)Google Scholar
- Copsel SN, Lightbourn CO, Barreras H. BET Bromodomain Inhibitors Which Permit Treg Function Enable a Combinatorial Strategy to Suppress GVHD in Pre-clinical Allogeneic HSCT. Front Immunol. 2019; 9:3104. Google Scholar
- Tacke M, Hanke G, Hanke T, Hunig T. CD28-mediated induction of proliferation in resting T cells in vitro and in vivo without engagement of the T cell receptor: evidence for functionally distinct forms of CD28. Eur J Immunol. 1997; 27(1):239-247. PubMedhttps://doi.org/10.1002/eji.1830270136Google Scholar
- Langenhorst D, Gogishvili T, Ribechini E. Sequential induction of effector function, tissue migration and cell death during poly-clonal activation of mouse regulatory T-cells. PLoS One. 2012; 7(11):e50080. PubMedhttps://doi.org/10.1371/journal.pone.0050080Google Scholar
- Lin CH, Hunig T. Efficient expansion of regulatory T cells in vitro and in vivo with a CD28 superagonist. Eur J Immunol. 2003; 33(3):626-638. PubMedhttps://doi.org/10.1002/eji.200323570Google Scholar
- Beyersdorf N, Gaupp S, Balbach K. Selective targeting of regulatory T cells with CD28 superagonists allows effective therapy of experimental autoimmune encephalomyelitis. J Exp Med. 2005; 202(3):445-455. PubMedhttps://doi.org/10.1084/jem.20051060Google Scholar
- Gogishvili T, Langenhorst D, Luhder F. Rapid regulatory T-cell response prevents cytokine storm in CD28 superagonist treated mice. PLoS One. 2009; 4(2):e4643. PubMedhttps://doi.org/10.1371/journal.pone.0004643Google Scholar
- Schmidt J, Elflein K, Stienekemeier M. Treatment and prevention of experimental autoimmune neuritis with superagonistic CD28-specific monoclonal antibodies. J Neuroimmunol. 2003; 140(1-2):143-152. PubMedhttps://doi.org/10.1016/S0165-5728(03)00182-6Google Scholar
- van den Brandt J, Fischer HJ, Walter L. Type 1 diabetes in BioBreeding rats is critically linked to an imbalance between Th17 and regulatory T cells and an altered TCR repertoire. J Immunol. 2010; 185(4):2285-2294. PubMedhttps://doi.org/10.4049/jimmunol.1000462Google Scholar
- Rodriguez-Palmero M, Franch A, Castell M. Effective treatment of adjuvant arthritis with a stimulatory CD28-specific monoclonal antibody. J Rheumatol. 2006; 33(1):110-118. PubMedGoogle Scholar
- Zaiss MM, Frey B, Hess A. Regulatory T cells protect from local and systemic bone destruction in arthritis. J Immunol. 2010; 184(12):7238-7246. PubMedhttps://doi.org/10.4049/jimmunol.0903841Google Scholar
- Miyasato K, Takabatake Y, Kaimori J. CD28 superagonist-induced regulatory T cell expansion ameliorates mesangioproliferative glomerulonephritis in rats. Clin Exp Nephrol. 2011; 15(1):50-57. PubMedhttps://doi.org/10.1007/s10157-010-0370-4Google Scholar
- Guilliams M, Bosschaerts T, Herin M. Experimental expansion of the regulatory T cell population increases resistance to African trypanosomiasis. J Infect Dis. 2008; 198(5):781-791. PubMedhttps://doi.org/10.1086/590439Google Scholar
- Azuma H, Isaka Y, Li X. Superagonistic CD28 antibody induces donor-specific tolerance in rat renal allografts. Am J Transplant. 2008; 8(10):2004-2014. PubMedhttps://doi.org/10.1111/j.1600-6143.2008.02358.xGoogle Scholar
- Kitazawa Y, Fujino M, Sakai T. Foxp3-expressing regulatory T cells expanded with CD28 superagonist antibody can prevent rat cardiac allograft rejection. J Heart Lung Transplant. 2008; 27(4):362-371. PubMedhttps://doi.org/10.1016/j.healun.2008.01.004Google Scholar
- Kitazawa Y, Fujino M, Li XK. Superagonist CD28 antibody preferentially expanded Foxp3-expressing nTreg cells and prevented graft-versus-host diseases. Cell Transplant. 2009; 18(5):627-637. PubMedGoogle Scholar
- Kitazawa Y, Li XK, Liu Z. Prevention of graft-versus-host diseases by in vivo supCD28mAb-expanded antigen-specific nTreg cells. Cell Transplant. 2010; 19(6):765-774. PubMedhttps://doi.org/10.3727/096368910X508870Google Scholar
- Beyersdorf N, Ding X, Hunig T, Kerkau T. Superagonistic CD28 stimulation of allogeneic T cells protects from acute graft-versus-host disease. Blood. 2009; 114(20):4575-4582. PubMedhttps://doi.org/10.1182/blood-2009-04-218248Google Scholar
- Suntharalingam G, Perry MR, Ward S. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. The New England journal of medicine. 2006; 355(10):1018-1028. PubMedhttps://doi.org/10.1056/NEJMoa063842Google Scholar
- Peine M, Marek RM, Lohning M. IL-33 in T Cell Differentiation, Function, and Immune Homeostasis. Trends Immunol. 2016; 37(5):321-333. Google Scholar
- Griesenauer B, Paczesny S. The ST2/IL-33 Axis in Immune Cells during Inflammatory Diseases. Front Immunol. 2017; 8:475. https://doi.org/10.3389/fimmu.2017.00475Google Scholar
- Paczesny S, Hakim FT, Pidala J. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: III. The 2014 Biomarker Working Group Report. Biol Blood Marrow Transplant. 2015; 21(5):780-792. PubMedhttps://doi.org/10.1016/j.bbmt.2015.01.003Google Scholar
- Matta BM, Lott JM, Mathews LR. IL-33 is an unconventional Alarmin that stimulates IL-2 secretion by dendritic cells to selectively expand IL-33R/ST2+ regulatory T cells. J Immunol. 2014; 193(8):4010-4020. PubMedhttps://doi.org/10.4049/jimmunol.1400481Google Scholar
- Matta BM, Turnquist HR. Expansion of Regulatory T Cells In Vitro and In Vivo by IL-33. Methods Mol Biol. 2016; 1371:29-41. Google Scholar
- Siede J, Frohlich A, Datsi A. IL-33 Receptor-Expressing Regulatory T Cells Are Highly Activated, Th2 Biased and Suppress CD4 T Cell Proliferation through IL-10 and TGFbeta Release. PLoS One. 2016; 11(8):e0161507. Google Scholar
- Schiering C, Krausgruber T, Chomka A. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature. 2014; 513(7519):564-568. PubMedhttps://doi.org/10.1038/nature13577Google Scholar
- Xu L, Li W, Wang X. The IL-33-ST2-MyD88 axis promotes regulatory T cell proliferation in the murine liver. Eur J Immunol. 2018; 48(8):1302-1307. Google Scholar
- Turnquist HR, Zhao Z, Rosborough BR. IL-33 expands suppressive CD11b+ Gr-1(int) and regulatory T cells, including ST2L+ Foxp3+ cells, and mediates regulatory T cell-dependent promotion of cardiac allograft survival. J Immunol. 2011; 187(9):4598-4610. PubMedhttps://doi.org/10.4049/jimmunol.1100519Google Scholar
- Brunner SM, Schiechl G, Falk W. Interleukin-33 prolongs allograft survival during chronic cardiac rejection. Transpl Int. 2011; 24(10):1027-1039. PubMedhttps://doi.org/10.1111/j.1432-2277.2011.01306.xGoogle Scholar
- Biton J, Khaleghparast Athari S, Thiolat A. In Vivo Expansion of Activated Foxp3+ Regulatory T Cells and Establishment of a Type 2 Immune Response upon IL-33 Treatment Protect against Experimental Arthritis. J Immunol. 2016; 197(5):1708-1719. PubMedhttps://doi.org/10.4049/jimmunol.1502124Google Scholar
- Popovic B, Golemac M, Podlech J. IL-33/ST2 pathway drives regulatory T cell dependent suppression of liver damage upon cytomegalovirus infection. PLoS Pathog. 2017; 13(4):e1006345. Google Scholar
- Reichenbach DK, Schwarze V, Matta BM. The IL-33/ST2 axis augments effector T-cell responses during acute GVHD. Blood. 2015; 125(20):3183-3192. PubMedhttps://doi.org/10.1182/blood-2014-10-606830Google Scholar
- Matta BM, Reichenbach DK, Zhang X. Peri-alloHCT IL-33 administration expands recipient T-regulatory cells that protect mice against acute GVHD. Blood. 2016; 128(3):427-439. PubMedhttps://doi.org/10.1182/blood-2015-12-684142Google Scholar
- Gatza E, Rogers CE, Clouthier SG. Extracorporeal photopheresis reverses experimental graft-versus-host disease through regulatory T cells. Blood. 2008; 112(4):1515-1521. PubMedhttps://doi.org/10.1182/blood-2007-11-125542Google Scholar
- Broxmeyer HE, Orschell CM, Clapp DW. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005; 201(8):1307-1318. PubMedhttps://doi.org/10.1084/jem.20041385Google Scholar
- Kean LS, Sen S, Onabajo O. Significant mobilization of both conventional and regulatory T cells with AMD3100. Blood. 2011; 118(25):6580-6590. PubMedhttps://doi.org/10.1182/blood-2011-06-359331Google Scholar
- Ukena SN, Velaga S, Goudeva L. Human regulatory T cells of G-CSF mobilized allogeneic stem cell donors qualify for clinical application. PLoS One. 2012; 7(12):e51644. PubMedhttps://doi.org/10.1371/journal.pone.0051644Google Scholar
- Robinson TM, O’Donnell PV, Fuchs EJ, Luznik L. Haploidentical bone marrow and stem cell transplantation: experience with post-transplantation cyclophosphamide. Semin Hematol. 2016; 53(2):90-97. Google Scholar
- Ganguly S, Ross DB, Panoskaltsis-Mortari A. Donor CD4+ Foxp3+ regulatory T cells are necessary for posttransplantation cyclophosphamide-mediated protection against GVHD in mice. Blood. 2014; 124(13):2131-2141. PubMedhttps://doi.org/10.1182/blood-2013-10-525873Google Scholar
- Cooper ML, Choi J, Karpova D. Azacitidine Mitigates Graft-versus-Host Disease via Differential Effects on the Proliferation of T Effectors and Natural Regulatory T Cells In Vivo. J Immunol. 2017; 198(9):3746-3754. PubMedhttps://doi.org/10.4049/jimmunol.1502399Google Scholar
- Ashami K, DiPersio JF, Choi J. Targeting IFNGR/IL6R or downstream JAK1/JAK2 to control GvHD. Oncotarget. 2018; 9(87):35721-35722. Google Scholar
- Choi J, Cooper ML, Staser K. Baricitinib-induced blockade of interferon gamma receptor and interleukin-6 receptor for the prevention and treatment of graft-versus-host disease. Leukemia. 2018; 32(11):2483-2494. Google Scholar
- Zanin-Zhorov A, Weiss JM, Nyuydzefe MS. Selective oral ROCK2 inhibitor down-regulates IL-21 and IL-17 secretion in human T cells via STAT3-dependent mechanism. Proc Natl Acad Sci U S A. 2014; 111(47):16814-16819. PubMedhttps://doi.org/10.1073/pnas.1414189111Google Scholar
- Zanin-Zhorov A, Flynn R, Waksal SD, Blazar BR. Isoform-specific targeting of ROCK proteins in immune cells. Small GTPases. 2016; 7(3):173-177. Google Scholar
- Spangler JB, Trotta E, Tomala J. Engineering a Single-Agent Cytokine/Antibody Fusion That Selectively Expands Regulatory T Cells for Autoimmune Disease Therapy. J Immunol. 2018; 201(7):2094-2106. PubMedhttps://doi.org/10.4049/jimmunol.1800578Google Scholar
- Leon K, Garcia-Martinez K, Carmenate T, Rojas G. Combining computational and experimental biology to develop therapeutically valuable IL2 muteins. Semin Oncol. 2018; 45(1-2):95-104. Google Scholar
- Pillai AB, George TI, Dutt S, Strober S. Host natural killer T cells induce an interleukin-4-dependent expansion of donor CD4+CD25+Foxp3+ T regulatory cells that protects against graft-versus-host disease. Blood. 2009; 113(18):4458-4467. PubMedhttps://doi.org/10.1182/blood-2008-06-165506Google Scholar
- Schneidawind D, Pierini A, Alvarez M. CD4+ invariant natural killer T cells protect from murine GVHD lethality through expansion of donor CD4+CD25+FoxP3+ regulatory T cells. Blood. 2014; 124(22):3320-3328. PubMedhttps://doi.org/10.1182/blood-2014-05-576017Google Scholar
- Du J, Paz K, Thangavelu G. Invariant natural killer T cells ameliorate murine chronic GVHD by expanding donor regulatory T cells. Blood. 2017; 129(23):3121-3125. PubMedhttps://doi.org/10.1182/blood-2016-11-752444Google Scholar
- Kuns RD, Morris ES, Macdonald KP. Invariant natural killer T cell-natural killer cell interactions dictate transplantation outcome after alpha-galactosylceramide administration. Blood. 2009; 113(23):5999-6010. PubMedhttps://doi.org/10.1182/blood-2008-10-183335Google Scholar
- Harvill ET, Fleming JM, Morrison SL. In vivo properties of an IgG3-IL-2 fusion protein. A general strategy for immune potentiation. J Immunol. 1996; 157(7):3165-3170. PubMedGoogle Scholar
- Carmenate T, Ortiz Y, Enamorado M. Blocking IL-2 Signal In Vivo with an IL-2 Antagonist Reduces Tumor Growth through the Control of Regulatory T Cells. J Immunol. 2018; 200(10):3475-3484. PubMedhttps://doi.org/10.4049/jimmunol.1700433Google Scholar
- Mitra S, Ring AM, Amarnath S. Interleukin-2 activity can be fine tuned with engineered receptor signaling clamps. Immunity. 2015; 42(5):826-838. PubMedhttps://doi.org/10.1016/j.immuni.2015.04.018Google Scholar
- Trotta E, Bessette PH, Silveria SL. A human anti-IL-2 antibody that potentiates regulatory T cells by a structure-based mechanism. Nat Med. 2018; 24(7):1005-1014. Google Scholar
- Slavin S, Fuks Z, Kaplan HS, Strober S. J Exp Med. 1978; 147(4):963-972. PubMedhttps://doi.org/10.1084/jem.147.4.963Google Scholar
- Pillai V, Maude SL. CART attack. Blood. 2017; 130(2):229. PubMedhttps://doi.org/10.1182/blood-2017-04-779009Google Scholar
- Periasamy S, Dhiman R, Barnes PF. Programmed death 1 and cytokine inducible SH2-containing protein dependent expansion of regulatory T cells upon stimulation With Mycobacterium tuberculosis. J Infect Dis. 2011; 203(9):1256-1263. PubMedhttps://doi.org/10.1093/infdis/jir011Google Scholar
- Zhou Q, Munger ME, Highfill SL. Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood. 2010; 116(14):2484-2493. PubMedhttps://doi.org/10.1182/blood-2010-03-275446Google Scholar
- Franceschini D, Paroli M, Francavilla V. PD-L1 negatively regulates CD4+CD25+Foxp3+ Tregs by limiting STAT-5 phosphorylation in patients chronically infected with HCV. J Clin Invest. 2009; 119(3):551-564. PubMedhttps://doi.org/10.1172/JCI36604Google Scholar
- Karim R, Jordanova ES, Piersma SJ. Tumor-expressed B7-H1 and B7-DC in relation to PD-1+ T-cell infiltration and survival of patients with cervical carcinoma. Clin Cancer Res. 2009; 15(20):6341-6347. PubMedhttps://doi.org/10.1158/1078-0432.CCR-09-1652Google Scholar
- Asano T, Meguri Y, Yoshioka T. PD-1 modulates regulatory T-cell homeostasis during low-dose interleukin-2 therapy. Blood. 2017; 129(15):2186-2197. PubMedhttps://doi.org/10.1182/blood-2016-09-741629Google Scholar
- Singh AK, Porrata LF, Aljitawi O. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin’s lymphoma. Bone Marrow Transplant. 2016; 51(9):1268-1270. Google Scholar