Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2022 Nov 18;211(2):149–163. doi: 10.1093/cei/uxac105

IL-2-based approaches to Treg enhancement

Ffion Harris 1, Yoana Arroyo Berdugo 2, Timothy Tree 3,4,
PMCID: PMC10019135  PMID: 36399073

Summary

Immune homeostasis is heavily dependent on the action of regulatory T cells (Tregs) which act to suppress the activation of many immune cell types including autoreactive conventional T cells. A body of evidence has shown that Tregs are intrinsically defective in many common autoimmune diseases, and gene polymorphisms which increase the susceptibility of autoimmune disease development have implicated the interleukin-2 (IL-2) signaling pathway as a key dysregulated mechanism. IL-2 is essential for Treg function and survival, and Tregs are highly sensitive to low levels of this cytokine in their environment. This review will revisit the rationale behind using low-dose IL-2 as a therapy to treat autoimmune diseases and evaluate the outcomes of trials to date. Furthermore, novel engineered IL-2 therapies with increased Treg specificity have shown promise in pre-clinical studies and human clinical trials for some agents have begun. Future studies will determine whether low-dose IL-2 or engineered IL-2 therapies can change the course of autoimmune and inflammatory diseases in patients.

Keywords: regulatory T cells, autoimmunity, immunotherapy, autoinflammatory disease, cytokines

Graphical Abstract

Graphical Abstract.

Graphical Abstract

What are Tregs and how do they suppress?

Tregs are vital for systemic immune regulation

The seminal paper by Sakaguchi and colleagues [1] was the first to demonstrate that CD4+ T-cell suspensions depleted of CD25hi cells were capable of inducing multi-organ autoimmunity when transferred into athymic nude mice. These CD25hi cells, later named regulatory T cells (Tregs), were identified in the human CD4+ T-cell compartment by several groups in 2001 [2–7]. There is now extensive evidence that Tregs play a vital role in preventing autoimmune diseases and in promoting graft tolerance in transplant patients.

Tregs constitute around 5% of CD4+ T cells, with the remaining 95% comprising of conventional T cells (Tconvs). Tregs can exert their suppressive function on a variety of immune cells including CD4+ Tconvs, CD8+ T cells, B cells, monocytes, dendritic cells, and natural killer cells. The mechanisms of Treg-mediated suppression have been extensively reviewed elsewhere [8, 9]. These include contact-dependent mechanisms such as expression of inhibitory immune checkpoint molecules, cytolytic molecules, secretion of suppressive cytokines such as IL-10, metabolic disruption, and sequestration of interleukin-2 (IL-2). The transcription factor Forkhead box protein P3 (FoxP3) is a master regulator of the Treg lineage [10]. The importance of FoxP3 in normal immune function is evident as the severe systemic autoimmune disease Immune Dysregulation Polyendocrinopathy Enteropathy X-linked (IPEX) syndrome is caused by mutations in the FoxP3 gene in humans [11] and a similar phenotype is observed Scurfy mice [12]. Treg survival and maintenance of their suppressive phenotype are heavily dependent on IL-2.

IL-2 is essential for Treg survival and function

IL-2 and its receptors

IL-2 is a 15.5kDa four-alpha-helix bundle glycoprotein comprising 133 amino acids [13] and is produced mostly by activated T cells [14] and B cells [15]. Notably, in contrast to Tconv cells which readily produce IL-2 upon T-cell receptor (TCR) engagement, Tregs cannot produce IL-2 [16].

As a member of the common gamma chain family of cytokines, IL-2 exerts its pleiotropic effects by binding its cell surface receptor complexes which are made up of 3 subunits; the IL-2 receptor α-chain (IL-2Rα; CD25), the IL-2 receptor β-chain (IL-2Rβ; CD122), and the common gamma-chain (IL-2Rγc; CD132) [17, 18]. Two IL-2 receptor complexes are capable of inducing signal transduction. Heterotrimeric association of all three subunits (IL-2Rα/β/γc) forms the high-affinity IL-2R complex (binding affinity Kd ≈ 10 pM) which is transiently expressed on Tconv cells upon activation, but constitutively expressed by Tregs. Association of the IL-2Rβ and IL-2Rγc chains forms the intermediate affinity complex (binding affinity Kd ≈ 1 nM) which is expressed by resting T cells and other immune cell types. The low-affinity IL-2 receptor consists of the IL-2Rα subunit alone but cannot initiate signal transduction (reviewed by [19]).

IL-2 signaling dysfunction impairs Tregs

Experiments pre-dating the discovery of Tregs show that knocking out either the IL2 [20], IL2RB [21], or IL2RA [22] genes in mice induces systemic inflammation and lymphoproliferation. These studies confounded previous expectations as IL-2 had historically been considered as a growth factor for T cells [23]). IL2RA deficiency in mice can induce Treg apoptosis leading to lethal autoimmunity, demonstrating the profound effect that defective IL-2 signaling has on Tregs [24]).

In humans, point mutations which disrupt the expression of the IL2RA and IL2RB genes can also cause autoimmune inflammatory disease [25, 26]. Tregs from CD25-deficient patients have a reduced frequency in peripheral blood and have a decreased in vitro suppressive capacity [27]. Moreover, Van Zeebroeck et al. [28] recently showed that deletion of IL2RA from human Tregs using CRISPR-Cas9 technology reduces their in vitro suppressive capacity.

IL-2 is indispensable for Treg development in the thymus and Treg survival in the periphery. IL2RB-knockout mice fail to produce Tregs in the thymus, however transgenic thymic expression of normal IL-2Rβ in these mice reconstitutes normal Treg development [29]). Moreover, administration of IL-2 neutralizing antibodies can deplete Tregs from the periphery in both neonatal and adult mice [30, 31]. Taken together, these studies show that functional Tregs are highly dependent on IL-2 and their dominant suppression of Tconv cells is lost upon removal of IL-2 signaling.

The IL-2 signaling pathway

Tregs have a specialized intracellular signaling response to IL-2

Signaling through the IL-2 receptor causes heterodimerization of the IL-2Rβ and γ-chain cytoplasmic domains, leading to recruitment of Janus kinase (JAK) non-receptor tyrosine kinases such as JAK1 and JAK3 (Figure 1) [32]. Importantly, the IL-2Rα chain (CD25) alone cannot initiate signal transduction. These JAK1 and JAK3 proteins in turn phosphorylate tiyrosine residues on the IL-2Rβ chain and can then propagate signal transduction through 3 pathways; the signal transducer and activator of transcription 5 (STAT5) pathway which is the dominant downstream pathway in Tregs [33], and the Ras/Raf/MAPK and the PI3K/Akt/mTOR pathways in Tconvs (reviewed in [34]). These pathways ultimately lead to expression of IL-2 target genes such as IL2RA, FoxP3, Cyclin D2, Bcl-2 [35].

Figure 1.

Figure 1.

IL-2 signaling and autoimmune disease susceptibility genes. IL-2 signals through receptor complexes composed of up to 3 subunits; the IL-2 receptor-α (CD25), IL-2 receptor-β (CD122), and the IL-2 receptor-γ (CD132; common-gamma chain). In conventional T cells, engagement with the intermediate affinity receptor or the high-affinity receptor initiates signal transduction through the STAT5, Ras/Raf/MAPK, and PI3K/Akt, mTOR signaling pathways leading to expression of IL-2 response genes. Regulatory T cells express high levels of PTEN which inhibits the PI3K/Akt/mTOR pathway, leaving Tregs relatively resistant to the effects of the immunosuppressant drug Rapamycin. Genome-wide association studies have identified single nucleotide polymorphisms in several elements of the IL-2 signaling pathway which increase the risk of autoimmune disease development (dashed boxes). Figure creating using BioRender.

IL-2 signaling though the STAT5 pathway is pivotal to Treg function and its dominance in Tregs marks a major difference in IL-2 signaling between Tregs and Tconvs. Signaling through the PI3K/Akt/mTOR pathway is inhibited in Tregs through high expression of PTEN (phosphatase and tensin homolog) and this mechanism restricts Treg proliferation in response to IL-2 [36]. Inhibition of signaling through this pathway appears to be important for Treg function as activating Akt causes a loss of suppressive ability in Tregs [37]. In contrast, Tconv cells strongly proliferate in response to IL-2-mediated PI3K/Akt/mTOR signaling, and this response can be inhibited by use of the immunosuppressive drug rapamycin which targets mTOR to bring about cell cycle arrest [38, 39]. As Tregs do not utilize IL-2-mediated PI3K/Akt/mTOR signaling, these cells are resistant to rapamycin-induced hypoproliferation. Addition of rapamycin to culture media upon TCR stimulation also induces a functionally suppressive phenotype in Tconvs [40].

IL-2 signaling in other immune cells

IL-2 receptor complexes are also expressed on other immune cells other than CD4+ T cells, making them responsive to IL-2. For example, IL-2 is required during initial priming of CD8+ T cells which are capable of expanding upon secondary antigen exposure [41]. Natural killer cells (NK) cells are known to proliferate strongly and produce cytokines in response to IL-2, including CD56bright NK cells which are thought to have an immunoregulatory role [42, 43]. Additionally, IL-2 promotes activation and proliferation of type 2 innate lymphoid cells (ILC2) as these cells express high levels of CD25 [44]. In humans and mice, IL-2 administration has been shown to induce secretion of IL-5 by these cells leading to downstream eosinophilia [45].

Tregs have a heightened sensitivity to IL-2

Tregs are highly sensitive to low levels of IL-2 in their environment, partially owing to their constitutive and high expression of the high-affinity IL-2 receptor complex (IL-2Rα/β/γc). This allows Tregs to sequester extracellular IL-2 from their environment, a process known as the IL-2 ‘sink’ suppressive mechanism, thus limiting Tconv growth while promoting a more suppressive Treg phenotype (reviewed by [46]).

However, Tregs experience IL-2-mediated STAT5-phosphorylation at around a 10-fold lower IL-2 concentration than either conventional memory T cells or T-cell blasts induced to express high levels of surface IL-2Rα, and activation of IL-2-dependent gene-responses can occur at a 100-fold lower IL-2 concentration than in Tconvs [47]. This suggests an intrinsically heightened sensitivity to IL-2 signaling which is independent of the high affinity IL-2 receptor and this may be due to IL-2 signal transduction being diverted away from the PI3K/Akt/mTOR and Ras/MAPK pathways. Together, Tregs have both a competitive advantage for IL-2 and a lower IL-2 signaling threshold. These features have formed the biological basis of low-dose IL-2 as a therapy, taking advantage of Tregs’ high responsiveness to concentrations of IL-2 which are too low to induce activation of Tconvs. Given the pivotal role that Tregs play in immune regulation, there is a strong rationale for therapeutically strengthening Tregs through IL-2 replenishment.

Evidence of Treg dysfunction in autoimmune diseases

Thymic selection has evolved to eliminate self-reactive cells from the T cell pool. However, this an imperfect process as low frequencies of autoreactive T cells are readily detectable in healthy individuals [48]. Despite this, autoreactive T cells are maintained in a functionally suppressed state in the periphery by Tregs thus preventing the onset of autoimmune disease. Depletion of Tregs at any stage in life can lead to autoimmunity in mice [49] and evidence suggests that Treg dysfunction in humans can lead to autoimmunity.

There has been a longstanding debate in the research community as to whether Tregs from individuals with autoimmune diseases are deficient in number or function, a subject reviewed in detail elsewhere [50–52]. Conflicting studies in classical autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) have shown either an increase [53, 54] or a decrease [55, 56] in total Treg frequency or number when compared to disease free controls. A substantial contribution to these discrepancies is likely to be caused by the precise combination of markers used to identify Tregs in flow cytometry experiments (including CD25, CD127, FOXP3, and Helios) and the fact that these can be variably expressed by activated non-regulatory T cells, especially in the inflammatory autoimmune environment. Other factors including the tissue used for testing, different stages of disease being investigated, and selection of the comparator population are likely to further affect reproducibility.

What is perhaps more widely accepted is that Treg suppressive function is reduced compared to age matched controls in a range of different autoimmune and inflammatory diseases including SLE [57–59], RA [60, 61], multiple sclerosis [62], ankylosing spondylitis [63], systemic sclerosis [64], and type 1 diabetes (T1D) [65–68]. Studies have suggested that reduced suppression is a factor both of increased resistance to suppression by effector T cells and intrinsic changes in Tregs themselves, both of which are likely to be compounded at sites of inflammation.

Although several different mechanisms leading to Treg dysfunction have been reported, IL-2 and its signaling components are commonly associated with defective Treg function in autoimmune diseases. For example, a study from our group and collaborators demonstrated that Tregs from T1D patients can exhibit reduced responsiveness to IL-2 leading to reduced Treg frequency, loss of FoxP3 expression, and reduced suppressive function [68]. This Treg phenotype is enriched in patients harboring single-nucleotide polymorphisms (SNPs) in protein tyrosine phosphatase N2 (PTPN2) and similar results have been shown in healthy individuals carrying PTPN2 SNPs [69]. PTPN2 is one of several genes encoding proteins involved in the IL-2 signaling pathway, including IL2 itself, which associate with T1D (Figure 1) [70, 71] RA, and Crohn’s disease [72] in genome wide association (GWAS) studies. This serves as evidence that IL-2 signaling discrepancies are causative factors in autoimmune diseases and not consequences of disease progression.

In addition to genetic factors which limit the ability of IL-2 to stabilize the Treg phenotype, environmental factors may also play a role. A state of persistent inflammation exists in most autoimmune diseases, and the inflammatory milieu driven by innate activation could reduce the functionality of Tregs. For example, several studies have demonstrated that IL-6 plays a role in inhibiting Treg function and promoting differentiation of Th17 Tconvs through altering the balance of STAT3 and STAT5 activation (reviewed by [73]). Elevated serum and tissue IL-6 have been reported in RA [74] and SLE [75]. Microbial dysbiosis is another environmental factor which is thought to influence autoimmune disease susceptibility. SLE and RA patients display characteristic patterns of altered microbiome diversity [76, 77] and probiotic treatment with protective bacterial strains such as L. salivarius induced anti-arthritic effects and increased the Treg:Th17 ratio in arthritis-prone mice [78].

Low-dose IL-2 as an autoimmune disease therapy: summary of trials to date

Low-dose IL-2 trials

Clinical trials using low doses of IL-2 aim to take advantage of Tregs’ high sensitivity to this cytokine while avoiding activation of Tconvs, thus expanding Treg numbers in vivo to promote immune tolerance. IL-2, also called Proleukin (brand name aldesleukin; Novartis), has been approved for use since 1992 and was first trialed at high doses as an anti-cancer drug [79]. However, a clinical benefit was observed only in unpredictable subgroups of patients and this therapy was associated with very high, sometimes lethal, toxicities. Another undesirable effect was the considerable expansion of Tregs in these cancer patients, therefore reduced doses were considered as a treatment for autoimmune and inflammatory diseases. Early studies in mouse models demonstrated a strong rationale for using low doses of IL-2 to treat T1D [80, 81].

Since the publication of the first study showing efficacy of low-dose subcutaneous IL-2 therapy in the treatment of hepatitis C-induced vasculitis [82] and graft-versus-host disease (GvHD) [83], this treatment has appeared in more than 30 different autoimmune and inflammatory disease studies and clinical trials. These include Alopecia areata, Amyotrophic lateral sclerosis (ALS), T1D, RA, ankylosing spondylitis (AS), SLE, psoriasis, Behcet’s disease, granulomatosis with polyangiitis (GPA), takayasu’s disease, Crohn’s disease (CD), ulcerative colitis (UC), autoimmune hepatitis (AIH), and sclerosing cholangitis. Brief details of treatments, biological and clinical outcomes are summarized in Table 1. These data highlight the vast range of dosing regimens that fall under the umbrella of ‘low dose IL-2’ making detailed comparison of individual studies difficult, even within a single clinical condition. In the sections that follow, we aim to draw general conclusions from these studies and highlight major areas of ongoing protocol development.

Table 1.

Low-dose IL-2 trials. Table summarizing the results of low-dose IL-2 trials in autoimmune and inflammatory diseases to date.

Condition/publication Clinical trial IL-2 treatment Biological response Clinical measures Clinical trial number
Alopecia areata
[84]
Single-centre, uncontrolled Phase I (n = 5) 15 MIU/d for 5d, followed by 3 MIU/d for 5d at week 3, 6 and 9 ↑ Tregs Improved NCT01840046
Alopecia areata
[85]
Multicentre, randomized placebo-controlled 15 MIU/d for 5d, followed by 3 MIU/d for 5d at week 3, 6 and 9 ↑ Total Tregs + naïve Tregs
↑ eosinophils
No changes NCT01840046
ALS
[86]
Single-centre, randomized (1:1:1) double-blind, placebo-controlled Phase IIa (n = 36) 3 cycles of placebo, 1 or 2 MIU/d for 5 days every 4 weeks ↑ Tregs
↑ NK cells, eosinophils, CD8+ T cells
↓ Monocytes
No changes NCT02059759
Autoimmune hepatitis
[87]
Pilot study (n = 2) 6 monthly cycles of 1 MIU/d for 5d ↑ Tregs
↓ NK cells
Improved in 1 patient (n=2) NA
GVHD
[83]
Single-centre, Phase I (n = 29) 03, 1 or 3 MIU/m2/d for 8 weeks ↑ Tregs
↑ NK cells
Improved NCT00529035
GVHD
[88]
Phase II (n = 16) 01-02 MIU/m2/d for 6-12 weeks ↑ Tregs Improved NCT00539695
GVHD
[89]
Phase I (n = 21; 11 children and 10 adults) Children 033 to 1 MIU/m2/d Adults 067 to 2 MIU/m2/d Dose escalations at weeks 2 and 4 ↑ Tregs
↑ NK cells and eosinophils
Improved NCT02318082
HCV-induced vasculitis
[82]
Single-centre, uncontrolled, Phase I/II (n = 10) 15 MIU/d for 5 days, followed by 3 MIU/d for 5d at weeks 3, 6 and 9 ↑ Tregs
↑ CD8+ Tregs
↑ NK cells and CD56bright
Improved NCT00574652
T1DM
[90]
Single-centre, uncontrolled Phase I (n = 9) 45 MIU/d 3×/week for 4 weeks; rapamycin 2 mg/d, followed by doses to maintain 5-10 ng/ml for 3 months ↑ Tregs
↑ NK cells and eosinophils
↓ B cells
Deteriorated NCT00525889
T1DM
[91]
Single-centre, randomized, placebo-controlled, double-blind Phase I/II (n = 24) 033, 1 or 3 MIU/d for 5 days ↑ Tregs
↑ eosinophils
↓ B cells
No evaluated NCT01353833
T1DM
[92]
Single-centre, randomized, placebo-controlled, double-blind, Phase I/II (n = 24 same patients [91]) 033, 1 or 3 MIU/d for 5 days ↑ Tregs + memory Tregs
↑ CD8+ Tregs
↑ NK cells and CD56bright
↓ B cells
Not evaluated NCT01353833
T1DM
[93]
Single-centre, uncontrolled, adaptative dose-finding Phase I/II (learning phase n=10, adaptative phase n = 30) Learning phase: single dose either 0004, 016, 060, 1 or 15 MIU/m2; adaptative phase: single dose to achieve Treg increases of 10%–20% from baseline ↑ Tregs
↑ NK cells CD56bright
↓ CD8+ T cells, NK and B cells
Not evaluated NCT01827735
T1DM
[94]
Multicentre, randomized, double-blind, placebo-controlled, dose-finding Phase I/II (n = 24 children) Placebo, 0125, 025 or 05 MIU/m2/d for 5 days followed by fortnightly for 1 year ↑ Tregs
↑ Eosinophils
No changes NCT01862120
Polymyositis/Dermatomyositis
[95]
Single-centre, controlled Phase I/II (n = 147; 116 ST, 31 ST + IL-2) 05 MIU/d for 5 days ↑ Tregs
↑ CD8+ T, Th1, Th2, Th17 Tconvs and B cells
Improved
Primary Sjögren’s syndrome
[96]
Single-centre, controlled Phase I/II (n = 190; 91 ST, 99 ST + IL-2) 1 MIU/d for 5 days ↑ Tregs
↑ Th17 Tconvs cells
No changes
Psoriatic arthritis
[97]
Single-centre, uncontrolled Phase I/II (n = 201; 106 HC, 73 ST, 22 ST + IL-2) 05 MIU/d for 5 days ↑ Tregs
↑ Th1, Th2, Th17 Tconvs
Improved
Rheumatoid arthritis
[98]
Single-centre, Phase I/II (n = 988; 100 HC, 655 ST, 233 ST + IL-2) 1 MIU/d for 5 days ↑ Tregs
↑ Th1, Th2 and Th17 Tconvs
Improved
SLE
[99]
Pilot study (n = 1 ST + IL-2) 4 cycles between 15 and 3 MIU/d for 5 days; separated by 9-16 days ↑ Tregs Improved NA
SLE
[100]
Single-centre, uncontrolled Phase I/IIa (n = 38) 3 cycles of 1 MIU every other day for 2 weeks followed by a 2-week break ↑ Tregs
↓ Tfh and Th17 Tconvs
Improved NCT02084238
SLE
[101]
Single-centre, uncontrolled Phase I/II (n = 5) 15 MIU/d for 5 days ↑ Tregs
↑ CD56bright NK cells
Not evaluated
SLE
[102]
Single-centre, uncontrolled, dose-adaptation Phase I/IIa (n = 12 ST+ IL-2) 4 cycles between 075 and 3 MIU/d for 5 days separated by 9-16 days ↑ Tregs
↑ eosinophils
↓ Tfh cells and B cells
Improved DRKS00004858
SLE
[103]
Single-centre, controlled Phase I/II (n = 30; 12 ST and 18 ST + IL-2) 3 cycles of 1 MIU every other day for 2 weeks followed by a 2-week break ↑ Tregs Improved
SLE
[104]
Single-centre, uncontrolled Phase I/II (n = 120; 70 HC, 50 rapamycin + IL-2) 100 WIU 3-5 days/month combined with rapamycin 05 mg every other day for 24 weeks ↑ Tregs
SLE
[105]
Single-centre, randomized, doble-blind, placebo-controlled Phase II (n = 60) 3 cycles of 1 MIU every other day for 2 weeks followed by a 2-week break ↑ Tregs
↑ NK cells and CD56bright
Improved NCT02465580
11 autoimmune diseases: RA, AS, SLE, psoriasis, Behcet’s disease, GPA, takayasu’s disease, CD, UC, AIH and sclerosing cholangitis [106] Multicentre, uncontrolled Phase I/IIa (n = 46; RA = 4, AS = 10, SLE = 6, psoriasis = 5, Behçet’s disease = 2, GPA = 1, Takayasu’s disease = 1, CD = 7, UC = 4, AIH = 2, sclerosing cholangitis = 4 1 MIU/d for 5 days, followed by fortnightly injections for 6 months ↑ Tregs
↑ CD56bright NK cells and eosinophils
Improved in AS, UC, SLE and psoriasis. NCT01988506
IL-2 combination therapies
T1D Single-centre, randomized, Phase II
(n = 24) Not yet recruiting
Cyclosporin orally 35 mg/kg twice a day for 2 months following by IL-2 1MIU/d for 5 days and then every week until 1 year NCT05153070
T1D Single-centre, randomized, Phase I/II
(n = 45) Not yet recruiting
Anti-thymocyte globulin 25 mg/kg in 2 infusions, 05 and 2 mg/kg at days 1 and 2
Adalimumab 50 mg/ month, for 1 year
IL-2 1 MIU/d for 5 days (days 10-14) and then every 2 weeks, for 52 weeks
Exenatide 2 mg SC weekly up to 52 weeks
NCT02586831
RA
[107
50 WIU IL-2 per day for a 5-day course with or without 160 mg Tocilizumab at the dosage of 160 mg at days 1-3 ↑ Treg:Th17 ratio Improved
T1D
[90]
Open-label Phase I 2-4 mg/day of oral rapamycin orally for 3 months in combination with 45 MIU of IL-2 three times per week for 4 weeks ↑ Tregs
↑ NK cells and eosinophils
Deterioration of β-cell function NCT00525889
SLE
[104]
100 WIU of IL-2 for 3 to 5 days/month in combination with 05 mg of oral rapamycin every other day ↑ Tregs,
↑ Treg:Th17 ratio
Improved
T1D
[108]
Open-label, dose-escalating, Phase I One infusion of autologous polyclonal Tregs in combination with 5-day courses of either 033 or 1 MIU of IL-2 ↑ Tregs,
↑ NK cells,
↑ MAIT cells,
↑ MAIT cells, CD8+ T cells
Deterioration of β-cell function NCT02772679

Low-dose IL-2 expands Tregs

The simplest and most uniform immunological outcome in all trials is an increase in the frequency and/or absolute number of Tregs in peripheral blood which was observed in almost all studies of low-dose subcutaneous IL-2 therapy, with the unique exception of a study in alopecia areata patients [84] where Tregs were recruited to and significantly increased within scalp skin lesions but not in the blood. Furthermore, most trials to date have measured expansion of Tregs only in peripheral blood without considering homing of Tregs to affected tissues. Access to tissue from the site of inflammation is major limitation in diseases such as T1D in which serious complication have been reported following biopsy of the pancreas (reviewed by [109]). However, site-specific Treg expansion should be considered as an informative outcome measure for low-dose IL-2 trials in diseases where accessing affected tissue carries low risk. Indeed, a study by [110] showed a significant reduction in Tregs (% of CD4+ T cells) infiltrating cutaneous lupus erythematosus skin lesions. As many low-dose IL-2 trials in SLE patients have reported improvement in skin rashes and alopecia [100, 102], it would be highly informative for future studies to consider measuring tissue-specific Treg expansion where possible.

Treg dose selection, response kinetics, and expanded Treg phenotype

IL-2 is known to have a very short half-life in human blood and so frequent dosing regimens are necessary [111]. Pre-clinical animal models are of little use in determining appropriate IL-2 doses for human trials as murine Tregs require much higher doses to stimulate in vivo expansion (as reviewed by [112]). This has resulted in a huge variation in individual and cumulative IL-2 doses, dosing frequency, and duration of IL-2 treatment between different studies making direct comparisons challenging. Overall, peripheral Tregs have been shown to increase in a dose and time-dependent manner and successive treatment cycles may have residual, cumulative effects [92]. In early studies, relatively high doses (1–3 MIU/m2) were used daily for prolonged periods of time resulting in a large increase in Treg frequency and number [82] but were associated with increases in other cell populations including effector CD4 T cells and NK cells. Subsequent studies suggested less frequent treatment may be optimal for maintaining selective Treg expansion resulting in protocols of daily cycles on 3–5 consecutive days followed by a rest period before repeating cycles [84, 86] or protocols based on an induction period of more intensive treatment followed by a maintenance phase [106]. Many of these protocols result in expansion of the number and frequency of Treg to a level that far exceeds normal ranges seen in disease free individuals raising concerns of generalized immunosuppression. An alternative approach to optimize expansion of Tregs within physiological levels, whilst limiting effects on other immune cells has been taken by researchers in T1D who have conducted a series of experimental medicine studies aimed at optimizing first the dose and then the frequency of administration using a response-adaptive trial design [93]. These studies identified a relatively low dose of 0.26 × 106 IU/m2 every 3 days was sufficient to maintain a 30% increase in Tregs with minimal effects on effector T cells [113] and is currently being tested in a Phase 2 clinical trial. In summary, the optimal regimen remains unclear and is likely to vary between conditions.

Many studies have performed detailed analysis of the phenotype and function of Tregs expanded by therapy. Most studies reported that in vivo expanded Tregs were skewed towards a memory phenotype displaying features characteristic of enhanced activation (higher expression of CD25, GITR, CTLA-4, Ki67, Helios, CD39, CD45R0, and lower CCR7) and have a higher suppressive capacity after the treatment [86, 92, 93, 101] Contrastingly, studies in GvHD showed a delayed proliferative response of naïve CD45RA+Ki67+ Tregs in response to treatment [114] and this was accompanied by an increase in recent thymic emigrant (RTE) Tregs (CD45RA+CD31+) after around 16 weeks of treatment which was especially prominent in pediatric patients [89]. This suggests that low-dose IL-2 may have the tissue-specific effect of increasing Treg thymic output and this may be of benefit as RTE Tregs were significantly lower at baseline in the GvHD patient cohort. Moreover, a recent study in alopecia areata patients noticed an increase in frequency and numbers only in the naïve (CD45RA+) Treg subset; however, no clinical benefit was derived from this therapy [85]. A trial comparing the effect of low-dose IL-2 across 11 autoimmune diseases found an increase in both memory and naïve Treg subsets [106] It remains to be seen whether dosing and treatment regimen can influence the phenotype of expanded Tregs in vivo.

Effect of low-dose IL-2 on other immune cells

As discussed above, many immune cell types, and some epithelial cells, express the IL-2 receptors. Although efforts have been made through dose selection to specifically target Tregs, and minimize activation of Tconvs, many other immune cell types have been shown to expand in response to low-dose IL-2 therapy. These most commonly include NK cells, of which the CD56bright subset appears to be particularly sensitive to treatment regardless of disease background due to their expression of CD25 [82, 92, 93, 101, 105] and these cells are thought to have immune regulatory properties. An increase in eosinophils is frequently observed which is likely a consequence of the ILC2-IL-5-eosinophil axis [45]. Both Todd et al. [93] and Rosenzwajg et al. [106] reported increased eosinophilia in patients with high eosinophil counts at baseline, meaning future patients could be stratified for risk of eosinophilia. Other common changes in cell frequencies include changes in subsets of Tconv helper subset frequencies [95–97], and reduced T follicular helper (Tfh) cell counts [100, 102] which could be linked to reduced CD19+ B cell counts (Table 1). Furthermore, a recent study in SLE reported a reduction in circulating anti-double-stranded DNA antibodies [105] in treated patients suggesting autoreactive B cells may also be inhibited by this therapy. Finally, a marked increase in regulatory CD8+CD25+ T cells has been recorded by some groups [82, 92], supporting the notion that this therapy skews towards a regulatory milieu which may induce suppressive properties in many immune cell types.

Therapeutic benefit

Many studies to date have shown an improvement in disease biomarkers during the course of low-dose IL-2 therapy, as measured by various scoring systems such as SLE responder index [100], disease activity score in psoriatic arthritis [97] and severity of alopecia tool [84], or metabolic parameters such as c-peptide decline in T1D [94] and liver enzyme and serum IgG levels in autoimmune hepatitis [87]. Despite this, very few trials to date have been designed to test the clinical efficacy of this therapy by the gold standard method of double blind, placebo-controlled trials with a pre-specified clinical efficacy outcome and no phase III trials have taken place to date.

Of the double blind, placebo-controlled trials conducted to date which have assessed clinical efficacy [85], and [94] did not see significant improvements in treated vs placebo groups in alopecia areata and T1D patients, respectively. However, these studies were small and therefore not powered to assess efficacy in large numbers of patients. A range of Phase II studies which aim to assess clinical efficacy are underway [105]. reported no significant improvement in SLE-4 score in treated vs placebo at 12 weeks (primary endpoint), but this did reach significance by week 24. Therefore, despite the robust Treg biological outcomes in every trial, clinical efficacy is yet to be proven however low-dose IL-2 continues to show promise as a future therapy.

Heterogeneous responsiveness and clinical response predictors

There is a noteworthy, heterogeneous response of Tregs to IL-2 between individuals, highlighting the importance of monitoring dose-response rates during treatment [91] and suggesting dose personalization may be important to optimize therapy. The clinical and biological response to IL-2 is likely to be complicated by disease duration and staging. In individuals with active and ongoing inflammation, response to therapy may be expected to be different to at risk individuals or patients in quiescent stages of disease. Similarly, the age of the recipient and state of thymic Treg output and differentiation status in the periphery is likely to affect responsiveness. Indeed, in clinical trials of low dose IL-2 in cGVHD, children achieved a higher Treg: Tconv ratio and better clinical responses compared to adults, suggesting that age-dependent intrinsic differences play an important role [89]. The cytokines soluble-IL-2Rα and VEGFR2 have been identified as clinical response predictors, distinguishing responders from non-responders to the therapy [94].

Safety and adverse events

Local reactions at injection sites and flu-like symptoms are the most commonly reported adverse events with no serious adverse events. However, IL-2 dose escalation above 1 MIU/day is not well tolerated in some patients and is associated with increased NK cell expansion and increased side-effect frequency [83, 91]. In addition to the promising safety and efficacy results to date, further trials are needed to validate long-term positive outcomes for autoimmune and inflammatory disease patients.

Combination therapies

Due to its positive impact on Tregs, low-dose IL-2 therapy has been trialed in combination with other immune-modulating or cellular therapies with an aim to restore immune regulation. A small study tested the efficacy of low-dose IL-2 therapy in combination with the anti-IL-6 receptor antibody tocilizumab in RA patients [107] as IL-6 is known to increase the Th17:Treg ratio and is a driver of this disease (Reviewed by [115]). This combination resulted in expansion of Tregs but not Th17 Tconvs, and reduced pain and swelling symptoms.

Combining low-dose IL-2 with other therapies which aim to “de-bulk” the Tconv pool by either removing or preventing the expansion of these cells is another strategy being considered. For example, a trial administering a short treatment of cyclosporin followed by low-dose IL-2 is currently recruiting newly diagnosed T1D patients (NCT05153070). Rapamycin (sirolimus) can limit Tconv proliferation in response to IL-2 through inhibiting mTOR (as described above) and also induce a suppressive phenotype in conventional CD4 T cells. A combination of IL-2 and rapamycin treatment has previously been shown to act synergistically in preventing the onset of β-cell destruction and dysglycaemia in non-obese diabetic (NOD) mice [116]). However in T1D patients, this combination therapy expanded Tregs but also caused a significant decline in β-cell function [90]. This was thought to be caused by either the rapamycin alone, or the treatment regimen in which low-dose IL-2 was given <2 weeks before the rapamycin. In contrast, this combination therapy showed efficacy in restoring Th17:Treg balance and reducing clinical disease scores in SLE patients [104]. Finally, a recent study by [108] was the first to trial low-dose IL-2 in combination with adoptive Treg therapy to treat T1D, however, this too failed to reach its secondary outcome measure of a reduction in c-peptide decline. Therefore, the clinical outcome of combination therapies are more variable depending on the disease background of treated patients.

Beyond wild-type IL-2

Overcoming the challenges of low-dose IL-2 therapy

Despite the early success of low-dose IL-2 trials in autoimmune diseases to date, there are two main limitations of this therapy as highlighted above: the short half-life of IL-2 requires frequent dosing that is low enough in individual and cumulative dosage to avoid Tconv activation, and off-target proliferative effects on other immune cells expressing IL-2 receptors. These drawbacks have inspired the development of new IL-2-engineering approaches which aim to increase the half-life of IL-2 and its specificity towards the high-affinity IL-2 receptor (Figure 2).

Figure 2.

Figure 2.

IL-2 engineering approaches. Several IL-2 technologies have been developed which act to increase the affinity of IL-2 to the high-affinity receptor complex, thus selectively expanding Tregs, and to increase its serum half-life in comparison to wild-type IL-2. Firstly, IL-2 can undergo PEGylation by attaching polyethyleneglycol chains to its surface. IL-2 muteins are generated by introducing point mutations into either the IL-2 protein or its receptor. IL-2 complexes are composed of IL-2 bound to either human or murine antibodies which act to block or conformationally alter binding sites for the IL-2 receptor-β (CD122). Finally, fusion proteins of IL-2 and the IL-2 receptor-α can also increase Treg selectivity and half-life. Figure created using BioRender.

IL-2 muteins

This novel technology works by introducing mutations into the IL-2 and/or IL-2R proteins to alter their affinity towards certain IL-2 receptor complexes (Figure 1). This technique was originally used to develop an IL-2 ‘superkine’ with an elevated binding affinity for IL-2Rβ (CD122) which improved in vivo anticancer responses [117]. Conversely, a study by Peterson et al. [118] showed that a novel IL-2 mutein/fusion protein IgG-(IL-2N88D)2 preferentially expanded Tregs in vivo in cynomolgus monkeys and humanised mice. Coupling of the IL-2N88D mutein to human IgG increased its in vivo serum half-life. A Phase I clinical trial of this drug (RO7049665) was completed in 2019; however, Phase II trials testing its clinical benefit in ulcerative colitis and autoimmune hepatitis have since been terminated early due to lack of efficacy (Table 2).

Table 2:

Engineered IL-2 trials

IL-2 engineering Agent Study phase Disease background Trial number Progress
Mutein Efavaleukin alfa Phase I/II GvHD NCT03422627 Ongoing
Mutein Efavaleukin alfa Phase I SLE NCT03451422 Ongoing
Mutein Efavaleukin alfa Phase II SLE NCT04680637 Ongoing
Mutein Efavaleukin alfa Phase II UC NCT04987307 Ongoing
Mutein Efavaleukin alfa Phase II RA NCT03410056 Terminated early
Mutein Efavaleukin alfa Phase I Healthy NCT04987333 Ongoing
Mutein RG-7835 Phase II UC NCT03943550 Terminated early
Mutein RG-7835 Phase I Healthy NCT03221179 Completed—no results posted
Mutein RG-7835 Phase II AIH NCT04790916 Terminated early as no efficacy shown in RA trial
Mutein CC-92252 Phase I Healthy/psoriasis NCT03971825 Terminated—did not meet progression criteria
PEGylated IL-2 NKTR-358 Phase I Healthy NCT04380324 Completed—no results posted
PEGylated IL-2 NKTR-358 Phase I SLE NCT03556007 Completed—no results posted
PEGylated IL-2 NKTR-358 Phase II SLE NCT04433585 Ongoing—recruiting
PEGylated IL-2 NKTR-358 Phase I Psoriasis NCT04119557 Completed—no results posted
PEGylated IL-2 NKTR-358 Phase I Eczema NCT04081350 Active—not recruiting
PEGylated IL-2 NKTR-358 Phase I Healthy NCT04133116 Completed—no results posted
PEGylated IL-2 NKTR-358 Phase I UC NCT04677179 Ongoing—recruiting

Table summarizing the completed and ongoing clinical trials using engineered IL-2 agents for the treatment of autoimmune diseases.

Recent studies using murine muteins in mouse model systems have showcased the wide therapeutic scope and selectivity of mutein therapies which could potentially be translated to humans. A study by Khoryati et al. [119] showed that Fc.Mut24, an IL-2 mutein fused to an IgG2a Fc domain, induced potent and specific Treg expansion, an enrichment of Tregs in the pancreas, and resolution of diabetes in NOD mice. Additionally, FC.Mut24 has shown benefit in mouse models of haemophilia (HaemA mice) [120] which are prone to formation of neutralising antibodies towards topical Factor VIII therapy. FC.Mut24 and subsequent Factor VIII gene therapy preferentially expanded Tregs and prevented anti-Factor VIII inhibitor formation in these mice [121]. have also demonstrated that antigen specific Tregs can be expanded in vivo using muteins such as these.

IL-2 mutein technology has been further developed through orthogonal IL-2/IL-2R pairs which cannot bind their native wild-type counterparts, giving a highly selective Treg activation system. In a murine mixed haematopoietic chimerism model, heart allograft acceptance and Treg expansion was significantly improved following adoptive transfer of orthogonal-CD122 transduced Tregs and administration of its paired orthogonal IL-2 [122].

To date, no IL-2 mutein therapies have been approved for use in humans but many clinical trials are underway testing their efficacy in classic autoimmune diseases. Efavaleukin alfa (Amgen) is currently in Phase I and Phase II trials (Table 2) testing this agent in healthy individuals, SLE, GvHD, ulcerative colitis, and rheumatoid arthritis, the latter of which has been terminated early.

PEGylation and IL-2 complexes

PEGylation increases the half-life of IL-2 and its selectivity for intermediate or high affinity complexes through attaching polyethylene glycol (PEG) chains to lysine residues on its surface. Dixit et al. [123] demonstrated that the PEGylated IL-2 molecule NKTR-358 has an increased selectivity for the high-affinity IL-2 receptor complex. When administered to cynomolgus monkeys, an increase in Treg expansion and in vitro suppressive capacity was observed. Furthermore, this treatment halted disease progression in a murine model of SLE.

Multiple clinical trials are underway using NKTR-358 (LY3471851; Nektar therapeutics; Eli Lilly) to treat human autoimmune and inflammatory diseases such as SLE, Psoriasis, Eczema, and UC (Table 2). Preliminary results from the SLE study (NCT03556007) have shown selective expansion of Tregs and a preliminary reduction in SLE clinical scores in some patients [124].

Forming complexes between IL-2 and anti-IL-2 antibodies serve to increase its half-life and can also block CD25 or CD122 binding sites on the IL-2 protein. When bound to IL-2, the murine mAb JES6-1A12 causes a conformational change which disrupts the interaction between wild-type IL-2 and CD122/CD132. IL-2-JES6-1A12 selectively expanded Tregs and improved symptoms in mouse models of asthma [125], experimental autoimmune encephalomyelitis (EAE [126];), transplantation [127], myasthenia gravis [128], and SLE [129]. IL-2-5344 has also shown efficacy in suppressing experimental food allergy in mice [130], and IL-2-5111.2 favored the expansion of Tregs in humanized mice and halted progression of T1D, EAE, and GvHD [131]. Furthermore, the human IL-2 complex IL-2-UFKA-20 preferentially expands Tregs in non-human primates promising good translatability for this complex [132].

IL-2 fusion proteins

Malek and colleagues were the first to develop an IL-2-CD25 fusion protein with selectivity for the high affinity IL-2 receptor complex and an increased half-life [133]. At lower doses than WT IL-2, this fusion protein has been shown to decrease the occurrence of diabetes in NOD mice [134] and decreased symptom severity in SLE mouse models [135]. A recent study has also showed that this fusion protein can induce a heightened Treg proliferative response compared to recombinant IL-2 [136].

In summary, IL-2 engineering approaches have shown promise in overcoming some of the challenges arisen from low-dose IL-2 therapies. However, further human trials are needed to assess the impact of drawbacks associated with these novel therapies. These include the need for higher much higher doses of agents which reduce signaling through the IL-2Rβ (CD122), potential immunogenicity of muteins, the hypothesized potential for non-covalently linked complexes to release free IL-2 in vivo (e.g. PEGylated IL-2), and off target effects on other cells which express the high-affinity IL-2 receptor.

Conclusion

To conclude, there is a strong rationale for targeting Tregs using low doses of IL-2 to treat autoimmune diseases given the evidence from GWAS studies, mechanistic in vitro Treg studies, and experiments in animal models. Clinical trials to date have proven that low doses of IL-2 are safe and universally expand Tregs across a range of autoimmune diseases. Therefore, it is clear that this drug is efficient at targeting Tregs; however, its specificity for Tregs is not optimal. Despite this, there are suggestions from some trials that other cell targets may contribute to efficacy such as Tfh cells, auto-antibody producing B cells, CD56Bright NK cells, and the thymus. Future clinical trials designed to evaluate clinical efficacy will shed light on whether this therapy can induce remission or halt the progression of autoimmune diseases such as SLE, GvHD, and T1D. These should also evaluate key unanswered questions such as whether expanded Tregs infiltrate disease-relevant tissues, how efficacy is affected by time-since disease onset, and would this be a financially viable treatment option for all autoimmune disease patients? Finally, IL-2 engineering approaches have successfully overcome some of the limitations of wild-type IL-2 such as increasing the half-life and specificity of IL-2 towards the high-affinity IL-2 receptor complex. Several clinical trials are underway to determine whether these drugs present an improved therapeutic avenue for autoimmune diseases than wild type IL-2 to restore immune regulation.

Acknowledgements

Not applicable.

Abbreviations

AIH

autoimmune hepatitis

ALS

amyotrophic lateral sclerosis

AS

ankylosing spondylitis

CD

Crohn’s disease

EAE

experimental autoimmune encephalomyelitis

FoxP3

Forkhead box protein P3

GPA

granulomatosis with polyangiitis

GvHD

graft versus host disease

GWAS

genome wise association

IL-2

interleukin 2

IL-2Rα

IL-2 receptor α-chain

IL-2Rγc

IL-2 receptor gamma-chain

IL-2Rβ

the IL-2 receptor β-chain

ILC2

type 2 innate lymphoid cells

IPEX

immune dysregulation polyendocrinopathy enteropathy X-linked syndrome

JAK

Janus kinase

NK cell

natural killer cell

NOD

non-obese diabetic

PEG

polyethylene glycol

PTEN

phosphatase and tensin homolog

PTPN2

protein phosphatase N2

RA

rheumatoid arthritis

RTE

recent thymic emigrant

SLE

systemic lupus erythematosus

SNP

single-nucleotide polymorphism

STAT5

signal transducer and activator of transcription 5

T1D

type 1 diabetes

Tconvs

conventional T cells

TCR

T cell receptor

Tfh

T follicular helper

Tregs

regulatory T cells

UB

ulcerative colitis

Contributor Information

Ffion Harris, Department of Immunobiology, Faculty of Life Sciences and Medicine, King’s College, London, UK.

Yoana Arroyo Berdugo, Department of Immunobiology, Faculty of Life Sciences and Medicine, King’s College, London, UK.

Timothy Tree, Department of Immunobiology, Faculty of Life Sciences and Medicine, King’s College, London, UK; National Institute of Health Research Biomedical Research Centre at Guy’s and St. Thomas’ National Health Service Foundation Trust, King’s College London, London, UK.

Ethical approval

The animal research adheres to the ARRIVE guidelines (https://arriveguidelines.org/arrive-guidelines)—not applicable.

Conflict of Interests

Tree has a patent (B75649EPD40021) relating to ld-IL-2 therapy for ALS and reports receiving research consultancy fees from Novartis and Quell Therapeutics.

Funding

This work is supported by an Innovative Medicines Initiative 2 Joint Undertaking under grant agreement no. 115797 (INNODIA), which receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA, JDRF, and The Leona M. and Harry B. Helmsley Charitable Trust. Flow cytometry studies and patient recruitment were supported by the T1DUK Immunotherapy Consortium funded by grants from Diabetes UK (Ref: 15/0005232 and 15/0005233). The authors acknowledge financial support from the Department of Health via the National Institute for Health Research (NIHR) comprehensive Biomedical Research Center award to Guy’s & St Thomas’ NHS Foundation Trust in partnership with King’s College London and King’s College Hospital NHS Foundation Trust. This work was also supported by Medical Research Council.

Data availability

Not applicable.

Author contributions

The authors contributed equally.

Clinical trial registration

Not applicable.

References

  • 1. 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. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7636184. [PubMed] [Google Scholar]
  • 2. Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA.. CD4+CD25high regulatory cells in human peripheral blood. J Immunol 2001, 167, 1245–53. doi: 10.4049/JIMMUNOL.167.3.1245. [DOI] [PubMed] [Google Scholar]
  • 3. Dieckmann D, Plottner H, Berchtold S, Berger T, Schuler G.. Ex vivo isolation and characterization of Cd4+Cd25+ T cells with regulatory properties from human blood. J Exp Med 2001, 193, 1303–10. doi: 10.1084/jem.193.11.1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH.. Identification and functional characterization of human Cd4+Cd25+ T cells with regulatory properties isolated from peripheral blood. J Exp Med 2001, 193, 1285–94. doi: 10.1084/jem.193.11.1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. 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, 1295–302. doi: 10.1084/jem.193.11.1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Stephens LA, Mottet C, Mason D, Powrie F.. Human CD4 + CD25 + thymocytes and peripheral T cells have immune suppressive activity in vitro. Eur J Immunol 2001, 31, 1247–54. doi:. [DOI] [PubMed] [Google Scholar]
  • 7. Taams LS, Smith J, Rustin MH, Salmon M, Poulter LW, Akbar AN.. Human anergic/suppressive CD4 + CD25 + T cells: a highly differentiated and apoptosis-prone population. Eur J Immunol 2001, 31, 1122–31. doi:. [DOI] [PubMed] [Google Scholar]
  • 8. Schmidt A, Oberle N, Krammer PH.. Molecular mechanisms of Treg-mediated T cell suppression. Front Immunol 2012, 3, 51. doi: 10.3389/fimmu.2012.00051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sojka DK, Huang YH, Fowell DJ.. Mechanisms of regulatory T-cell suppression – a diverse arsenal for a moving target. Immunology 2008, 124, 13–22. doi: 10.1111/j.1365-2567.2008.02813.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Fontenot JD, Gavin MA, Rudensky AY.. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003, 4, 330–6. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 11. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001, 27, 20–1. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 12. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 2001, 27, 68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 13. Brandhuber BJ, Boone T, Kenney WC, McKay DB.. Three-dimensional structure of interleukin-2. Science 1987, 238, 1707–9. doi: 10.1126/science.3500515. [DOI] [PubMed] [Google Scholar]
  • 14. Gillis S, Ferm MM, Ou W, Smith KA.. T cell growth factor: parameters of production and a quantitative microassay for activity. J Immunol 1978, 120, 2027–32. [PubMed] [Google Scholar]
  • 15. Kindler V, Matthes T, Jeannin P, Zubler RH.. Interleukin-2 secretion by human B lymphocytes occurs as a late event and requires additional stimulation after CD40 cross-linking. Eur J Immunol 1995, 25, 1239–43. doi: 10.1002/eji.1830250516. [DOI] [PubMed] [Google Scholar]
  • 16. Thornton AM, Donovan EE, Piccirillo CA, Shevach EM.. Cutting edge: IL-2 is critically required for the in vitro activation of CD4+CD25+ T cell suppressor function. J Immunol 2004, 172, 6519–23. doi: 10.4049/jimmunol.172.11.6519. [DOI] [PubMed] [Google Scholar]
  • 17. Hatakeyama M, Tsudo M, Minamoto S, Kono T, Doi T, Miyata T, et al. Interleukin-2 receptor β chain gene: generation of three receptor forms by cloned human α and β chain cDNA’s. Science 1989, 244, 551–6. doi: 10.1126/SCIENCE.2785715. [DOI] [PubMed] [Google Scholar]
  • 18. Takeshita T, Asao H, Ohtani K, Ishii N, Kumaki S, Tanaka N, et al. Cloning of the γ chain of the human IL-2 receptor. Science 1992, 257, 379–82. doi: 10.1126/SCIENCE.1631559. [DOI] [PubMed] [Google Scholar]
  • 19. Ye C, Brand D, Zheng SG.. Targeting IL-2: an unexpected effect in treating immunological diseases. Signal Transduct Targeted Ther 2018, 3. doi: 10.1038/s41392-017-0002-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, Horak I.. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 1993, 75, 253–61. doi: 10.1016/0092-8674(93)80067-o. [DOI] [PubMed] [Google Scholar]
  • 21. Suzuki H, Kündig TM, Furlonger C, Wakeham A, Timms E, Matsuyama T, et al. Deregulated T cell activation and autoimmunity in mice lacking interleukin-2 receptor beta. Science 1995, 268, 1472–6. doi: 10.1126/SCIENCE.7770771. [DOI] [PubMed] [Google Scholar]
  • 22. Willerford DM, Chen J, Ferry JA, Davidson L, Ma A, Alt FW.. Interleukin-2 receptor alpha chain regulates the size and content of the peripheral lymphoid compartment. Immunity 1995, 3, 521–30. doi: 10.1016/1074-7613(95)90180-9. [DOI] [PubMed] [Google Scholar]
  • 23. Morgan DA, Ruscetti FW, Gallo R.. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science 1976, 193, 1007–8. doi: 10.1126/science.181845. [DOI] [PubMed] [Google Scholar]
  • 24. Barron L, Dooms H, Hoyer KK, Kuswanto W, Hofmann J, O’Gorman WE, et al. Mechanisms of IL-2–dependent maintenance of functional regulatory T cells. J Immunol 2010, 185, 6426–30. doi: 10.4049/jimmunol.0903940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Sharfe N, Dadi HK, Shahar M, Roifman CM.. Human immune disorder arising from mutation of the α chain of the interleukin-2 receptor. Proc Natl Acad Sci USA 1997, 94, 3168–71. doi: 10.1073/pnas.94.7.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Zhang Z, Gothe F, Pennamen P, James JR, McDonald D, Mata CP, et al. Human interleukin-2 receptor β mutations associated with defects in immunity and peripheral tolerance. J Exp Med 2019, 216, 1311–27. doi: 10.1084/JEM.20182304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Roth TL, Puig-Saus C, Yu R, Shifrut E, Carnevale J, Li PJ, et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature 2018, 559, 405–9. doi: 10.1038/S41586-018-0326-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Van Zeebroeck L, Arroyo Hornero R, Côrte-Real BF, Hamad I, Meissner TB, Kleinewietfeld M.. Fast and efficient genome editing of human FOXP3+ regulatory T cells. Front Immunol 2021, 12, 655122. doi: 10.3389/fimmu.2021.655122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. 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, 167–78. doi: 10.1016/s1074-7613(02)00367-9. [DOI] [PubMed] [Google Scholar]
  • 30. Bayer AL, Yu A, Adeegbe D, Malek TR.. Essential role for interleukin-2 for CD4+CD25+ T regulatory cell development during the neonatal period. J Exp Med 2005, 201, 769–77. doi: 10.1084/jem.20041179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Setoguchi R, Hori S, Takahashi T, Sakaguchi S.. Homeostatic maintenance of natural Foxp3+ CD25+ CD4+ regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med 2005, 201, 723–35. doi: 10.1084/jem.20041982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Russell SM, Johnston JA, Noguchi M, Kawamura M, Bacon CM, Friedmann M, et al. Interaction of IL-2Rβ and γc chains with Jak1 and Jak3: Implications for XSCID and XCID. Science 1994, 266, 1042–5. doi: 10.1126/SCIENCE.7973658. [DOI] [PubMed] [Google Scholar]
  • 33. Bensinger SJ, Walsh PT, Zhang J, Carroll M, Parsons R, Rathmell JC, et al. Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J Immunol (Baltimore, MD: 1950) 2004, 172, 5287–96. doi: 10.4049/JIMMUNOL.172.9.5287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Gaffen SL. Signaling domains of the interleukin 2 receptor. Cytokine 2001, 14, 63–77. doi: 10.1006/cyto.2001.0862. [DOI] [PubMed] [Google Scholar]
  • 35. Kovanen PE, Young L, Al-Shami A, Rovella V, Pise-Masison CA, Radonovich MF, et al. Global analysis of IL-2 target genes: identification of chromosomal clusters of expressed genes. Int Immunol 2005, 17, 1009–21. doi: 10.1093/INTIMM/DXH283. [DOI] [PubMed] [Google Scholar]
  • 36. Walsh PT, Buckler JL, Zhang J, Gelman AE, Dalton NM, Taylor DK, et al. PTEN inhibits IL-2 receptor-mediated expansion of CD4+ CD25+ Tregs. J Clin Investig 2006, 116, 2521–31. doi: 10.1172/JCI28057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Crellin NK, Garcia RV, Levings MK.. Altered activation of AKT is required for the suppressive function of human CD4+CD25+ T regulatory cells. Blood 2007, 109, 2014–22. doi: 10.1182/blood-2006-07-035279. [DOI] [PubMed] [Google Scholar]
  • 38. Reif K, Burgering BMT, Cantrell DA.. Phosphatidylinositol 3-kinase links the interleukin-2 receptor to protein kinase b and p70 S6 kinase. J Biol Chem 1997, 272, 14426–33. doi: 10.1074/JBC.272.22.14426. [DOI] [PubMed] [Google Scholar]
  • 39. Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH.. RAFT1: A mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994, 78, 35–43. doi: 10.1016/0092-8674(94)90570-3. [DOI] [PubMed] [Google Scholar]
  • 40. Valmori D, Tosello V, Souleimanian NE, Godefroy E, Scotto L, Wang Y, et al. Rapamycin-mediated enrichment of T cells with regulatory activity in stimulated CD4+ T cell cultures is not due to the selective expansion of naturally occurring regulatory T cells but to the induction of regulatory functions in conventional CD4+ T cells. J Immunol 2006, 177, 944–9. doi: 10.4049/jimmunol.177.2.944. [DOI] [PubMed] [Google Scholar]
  • 41. Williams MA, Tyznik AJ, Bevan MJ.. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature 2006, 441, 890–3. doi: 10.1038/nature04790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56bright subset. Blood 2001, 97, 3146–51. doi: 10.1182/BLOOD.V97.10.3146. [DOI] [PubMed] [Google Scholar]
  • 43. Fehniger TA, Cooper MA, Nuovo GJ, Cella M, Facchetti F, Colonna M, et al. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell–derived IL-2: a potential new link between adaptive and innate immunity. Blood 2003, 101, 3052–7. doi: 10.1182/blood-2002-09-2876. [DOI] [PubMed] [Google Scholar]
  • 44. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of TH2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells. Nature 2010, 463, 540–4. doi: 10.1038/nature08636. [DOI] [PubMed] [Google Scholar]
  • 45. Van Gool F, Molofsky AB, Morar MM, Rosenzwajg M, Liang HE, Klatzmann D, et al. Interleukin-5-producing group 2 innate lymphoid cells control eosinophilia induced by interleukin-2 therapy. Blood 2014, 124, 3572–6. doi: 10.1182/BLOOD-2014-07-587493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Cheng G, Yu A, Malek TR.. T cell tolerance and the multi-functional role of IL-2R signaling in T regulatory cells. Immunol Rev 2011, 241, 63–76. doi: 10.1111/j.1600-065X.2011.01004.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Yu A, Snowhite I, Vendrame F, Rosenzwajg M, Klatzmann D, Pugliese A, et al. 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, 2172–83. doi: 10.2337/db14-1322. [DOI] [PubMed] [Google Scholar]
  • 48. Danke NA, Koelle DM, Yee C, Beheray S, Kwok WW.. Autoreactive T cells in healthy individuals. J Immunol (Baltimore, MD: 1950) 2004, 172, 5967–72. doi: 10.4049/jimmunol.172.10.5967. [DOI] [PubMed] [Google Scholar]
  • 49. Kim JM, Rasmussen JP, Rudensky AY.. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol 2007, 8, 191–7. doi: 10.1038/ni1428. [DOI] [PubMed] [Google Scholar]
  • 50. Hull CM, Peakman M, Tree TIM.. Regulatory T cell dysfunction in type 1 diabetes: what’s broken and how can we fix it?. Diabetologia 2017, 60, 1839–50. doi: 10.1007/s00125-017-4377-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Rajendeeran A, Tenbrock K.. Regulatory T cell function in autoimmune disease. J Translat Autoimmun 2021, 4, 2589–9090. doi: 10.1016/J.JTAUTO.2021.100130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Scheinecker C, Göschl L, Bonelli M.. Treg cells in health and autoimmune diseases: new insights from single cell analysis. J Autoimmun 2020, 110, 102376. doi: 10.1016/j.jaut.2019.102376. [DOI] [PubMed] [Google Scholar]
  • 53. Moradi B, Schnatzer P, Hagmann S, Rosshirt N, Gotterbarm T, Kretzer JP, et al. CD4+CD25+/highCD127low/- regulatory T cells are enriched in rheumatoid arthritis and osteoarthritis joints-analysis of frequency and phenotype in synovial membrane, synovial fluid and peripheral blood. Arthritis Res Ther 2014, 16, R97. doi: 10.1186/ar4545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Pan X, Yuan X, Zheng Y, Wang W, Shan J, Lin F, et al. Increased CD45RA+FoxP3low regulatory T cells with impaired suppressive function in patients with systemic lupus erythematosus. PLoS One 2012, 7, e34662. doi: 10.1371/JOURNAL.PONE.0034662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Li W, Deng C, Yang H, Wang G.. The regulatory T cell in active systemic lupus erythematosus patients: a systemic review and meta-analysis. Front Immunol 2019, 10, 159. doi: 10.3389/fimmu.2019.00159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Zhang X, Zhang X, Zhuang L, Xu C, Li TAO, Zhang G, et al. Decreased regulatory T-cell frequency and interleukin-35 levels in patients with rheumatoid arthritis. Exp Therapeutic Med 2018, 16, 5366–72. doi: 10.3892/etm.2018.6885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Jacquemin C, Augusto JF, Scherlinger M, Gensous N, Forcade E, Douchet I, et al. OX40L/OX40 axis impairs follicular and natural Treg function in human SLE. JCI Insight 2018, 3, e122167. doi: 10.1172/JCI.INSIGHT.122167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Legorreta-Haquet MV, Chávez-Rueda K, Chávez-Sánchez L, Cervera-Castillo H, Zenteno-Galindo E, Barile-Fabris L, et al. Function of Treg cells decreased in patients with systemic lupus erythematosus due to the effect of prolactin. Medicine 2016, 95, e2384. doi: 10.1097/MD.0000000000002384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Valencia X, Yarboro C, Illei G, Lipsky PE.. Deficient CD4+CD25high T regulatory cell function in patients with active systemic lupus erythematosus. J Immunol 2007, 178, 2579–88. doi: 10.4049/jimmunol.178.4.2579. [DOI] [PubMed] [Google Scholar]
  • 60. Flores-Borja F, Jury EC, Mauri C, Ehrenstein MR.. Defects in CTLA-4 are associated with abnormal regulatory T cell function in rheumatoid arthritis. Proc Natl Acad Sci USA 2008, 105, 19396–401. doi: 10.1073/pnas.0806855105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Sun H, Gao W, Pan W, Zhang Q, Wang G, Feng D, et al. Tim3 + Foxp3 + Treg cells are potent inhibitors of effector T cells and are suppressed in rheumatoid arthritis. Inflammation 2017, 40, 1342–50. doi: 10.1007/S10753-017-0577-6. [DOI] [PubMed] [Google Scholar]
  • 62. Sambucci M, Gargano F, De Rosa V, De Bardi M, Picozza M, Placido R, et al. FoxP3 isoforms and PD-1 expression by T regulatory cells in multiple sclerosis. Sci Rep 2018, 8, 1–9. doi: 10.1038/s41598-018-21861-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Guo H, Zheng M, Zhang K, Yang F, Zhang X, Han Q, et al. Functional defects in CD4+ CD25high FoxP3+ regulatory cells in ankylosing spondylitis. Sci Rep 2016, 6, 37559. doi: 10.1038/srep37559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Wang YY, Wang Q, Sun XH, Liu RZ, Shu Y, Kanekura T, et al. DNA hypermethylation of the forkhead box protein 3 (FOXP3) promoter in CD4+ T cells of patients with systemic sclerosis. Br J Dermatol 2014, 171, 39–47. doi: 10.1111/BJD.12913. [DOI] [PubMed] [Google Scholar]
  • 65. Brusko TM, Wasserfall CH, Clare-Salzler MJ, Schatz DA, Atkinson MA.. Functional defects and the influence of age on the frequency of CD4+ CD25+ T-cells in type 1 diabetes. Diabetes 2005, 54, 1407–14. doi: 10.2337/diabetes.54.5.1407. [DOI] [PubMed] [Google Scholar]
  • 66. Lindley S, Dayan CM, Bishop A, Roep BO, Peatman M, Tree TIM.. Defective suppressor function in CD4+CD25+ T-cells from patients with type 1 diabetes. Diabetes 2005, 54, 92–9. doi: 10.2337/DIABETES.54.1.92. [DOI] [PubMed] [Google Scholar]
  • 67. Long SA, Cerosaletti K, Bollyky PL, Tatum M, Shilling H, Zhang S, 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: 10.2337/DB09-0694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Yang JHM, Cutler AJ, Ferreira RC, Reading JL, Cooper NJ, Wallace C, et al. Natural variation in IL-2 sensitivity influences regulatory T cell frequency and function in individuals with long-standing type 1 diabetes. Diabetes 2015, 64, 3891–902. doi: 10.2337/DB15-0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Long SA, Cerosaletti K, Wan JY, Ho JC, Tatum M, Wei S, et al. An autoimmune-associated variant in PTPN2 reveals an impairment of IL-2R signaling in CD4+ T cells. Genes Immunity 2011, 12, 116–25. doi: 10.1038/gene.2010.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Barrett JC, Clayton DG, Concannon P, Akolkar B, Cooper JD, Erlich HA, et al. Genome-wide association study and meta-analysis finds over 40 loci affect risk of type 1 diabetes. Nat Genet 2009, 41, 703–7. doi: 10.1038/NG.381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Todd JA, Walker NM, Cooper JD, Smyth DJ, Downes K, Plagnol V, et al. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 2007, 39, 857–64. doi: 10.1038/ng2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Burton PR, Clayton DG, Cardon LR, Craddock N, Deloukas P, Duncanson A, et al. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007, 447, 661–78. doi: 10.1038/NATURE05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Jones BE, Maerz MD, Buckner JH.. IL-6: a cytokine at the crossroads of autoimmunity. Curr Opin Immunol 2018, 55, 9–14. doi: 10.1016/j.coi.2018.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hirano T, Matsuda T, Turner M, Miyasaka N, Buchan G, Tang B, et al. Excessive production of interleukin 6/B cell stimulatory factor-2 in rheumatoid arthritis. Eur J Immunol 1988, 18, 1797–802. doi: 10.1002/EJI.1830181122. [DOI] [PubMed] [Google Scholar]
  • 75. Linker-Israeli M, Deans RJ, Wallace DJ, Prehn J, Ozeri-Chen T, Klinenberg JR.. Elevated levels of endogenous IL-6 in systemic lupus erythematosus. A putative role in pathogenesis. J Immunol 1991, 147, 117–23. [PubMed] [Google Scholar]
  • 76. Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, et al. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med 2016, 8, 43. doi: 10.1186/S13073-016-0299-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Li Y, Wang HF, Li X, Li HX, Zhang Q, Zhou HW, et al. Disordered intestinal microbes are associated with the activity of systemic lupus erythematosus. Clin Sci (London, England: 1979) 2019, 133, 821–38. doi: 10.1042/CS20180841. [DOI] [PubMed] [Google Scholar]
  • 78. Liu X, Zhang J, Zou Q, Zhong B, Wang H, Mou F, et al. Lactobacillus salivarius isolated from patients with rheumatoid arthritis suppresses collagen-induced arthritis and increases Treg frequency in mice. Https://Home.Liebertpub.Com/Jir 2016, 36, 706–12. doi: 10.1089/JIR.2016.0057. [DOI] [PubMed] [Google Scholar]
  • 79. Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 1994, 271, 907–13. doi: 10.1001/JAMA.1994.03510360033032. [DOI] [PubMed] [Google Scholar]
  • 80. Grinberg-Bleyer Y, Baeyens A, You S, Elhage R, Fourcade G, Gregoire S, et al. IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J Exp Med 2010, 207, 1871–8. doi: 10.1084/JEM.20100209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Tang Q, Adams JY, Penaranda C, Melli K, Piaggio E, Sgouroudis E, et al. Central role of a defective interleukin-2 production in triggering islet autoimmune destruction. Immunity 2008, 28, 687–97. doi: 10.1016/J.IMMUNI.2008.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 2011, 365, 2067–77. doi: 10.1056/NEJMoa1105143. [DOI] [PubMed] [Google Scholar]
  • 83. Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med 2011, 365, 2055–66. doi: 10.1056/NEJMOA1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Castela E, Le Duff F, Butori C, Ticchioni M, Hofman P, Bahadoran P, et al. Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in Alopecia Areata. JAMA Dermatol 2014, 150, 748–51. doi: 10.1001/JAMADERMATOL.2014.504. [DOI] [PubMed] [Google Scholar]
  • 85. Le Duff F, Bouaziz JD, Fontas E, Ticchioni M, Viguier M, Dereure O, et al. Low-dose IL-2 for treating moderate to severe Alopecia Areata: a 52-week multicenter prospective placebo-controlled study assessing its impact on T regulatory cell and NK cell populations. J Investig Dermatol 2021, 141, 933–6. doi: 10.1016/J.JID.2020.08.015. [DOI] [PubMed] [Google Scholar]
  • 86. Camu W, Mickunas M, Veyrune JL, Payan C, Garlanda C, Locati M, et al. Repeated 5-day cycles of low dose aldesleukin in amyotrophic lateral sclerosis (IMODALS): a phase 2a randomised, double-blind, placebo-controlled trial. EBioMedicine 2020, 59, 102844. doi: 10.1016/J.EBIOM.2020.102844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Lim TY, Martinez-Llordella M, Kodela E, Gray E, Heneghan MA, Sanchez-Fueyo A.. Low-dose interleukin-2 for refractory autoimmune hepatitis. Hepatology 2018, 68, 1649–52. doi: 10.1002/hep.30059. [DOI] [PubMed] [Google Scholar]
  • 88. Kennedy-Nasser AA, Ku S, Castillo-Caro P, Hazrat Y, Wu MF, Liu H, et al. Ultra Low-dose IL-2 for GVHD prophylaxis after allogeneic hematopoietic stem cell transplantation mediates expansion of regulatory T cells without diminishing anti-viral and anti-leukemic activity. Clin Cancer Res 2014, 20, 2215–25. doi: 10.1158/1078-0432.CCR-13-3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Whangbo JS, Kim HT, Mirkovic N, Leonard L, Poryanda S, Silverstein S, et al. Dose-escalated interleukin-2 therapy for refractory chronic graft-versus-host disease in adults and children. Blood Adv 2019, 3, 2550–61. doi: 10.1182/BLOODADVANCES.2019000631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Long AS, Rieck M, Sanda S; Network, for D. T. and the I. T. Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function. Diabetes 2012, 61, 2340–8. doi: 10.2337/DB12-0049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Hartemann A, Bensimon G, Payan CA, Jacqueminet S, Bourron O, Nicolas N, et al. Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diab Endocrinol 2013, 1, 295–305. doi: 10.1016/S2213-8587(13)70113-X. [DOI] [PubMed] [Google Scholar]
  • 92. Rosenzwajg M, Churlaud G, Mallone R, Six A, Dérian N, Chaara W, et al. Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun 2015, 58, 48–58. doi: 10.1016/J.JAUT.2015.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Todd JA, Evangelou M, Cutler AJ, Pekalski ML, Walker NM, Stevens HE, et al. Regulatory T cell responses in participants with type 1 diabetes after a single dose of interleukin-2: a non-randomised, open label, adaptive dose-finding trial. PLoS Med 2016, 13, e1002139. doi: 10.1371/JOURNAL.PMED.1002139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Rosenzwajg M, Salet R, Lorenzon R, Tchitchek N, Roux A, Bernard C, et al. Low-dose IL-2 in children with recently diagnosed type 1 diabetes: a Phase I/II randomised, double-blind, placebo-controlled, dose-finding study. Diabetologia 2020, 63, 1808–21. doi: 10.1007/s00125-020-05200-w. [DOI] [PubMed] [Google Scholar]
  • 95. Zhang SX, Wang J, Sun HH, Zhang JQ, Liu GY, Luo J, et al. Circulating regulatory T cells were absolutely decreased in dermatomyositis/polymyositis patients and restored by low-dose IL-2. Ann Rheum Dis 2019, 80, e130. doi: 10.1136/ANNRHEUMDIS-2019-216246. [DOI] [PubMed] [Google Scholar]
  • 96. Miao M, Hao Z, Guo Y, Zhang X, Zhang S, Luo J, et al. Short-term and low-dose IL-2 therapy restores the Th17/Treg balance in the peripheral blood of patients with primary Sjögren’s syndrome. Ann Rheum Dis 2018, 77, 1838–40. doi: 10.1136/ANNRHEUMDIS-2018-213036. [DOI] [PubMed] [Google Scholar]
  • 97. Wang J, Zhang SX, Hao YF, Qiu MT, Luo J, Li YY, et al. The numbers of peripheral regulatory T cells are reduced in patients with psoriatic arthritis and are restored by low-dose interleukin-2. Therapeutic Adv Chronic Dis 2020, 11, 2040622320916014. doi: 10.1177/2040622320916014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Zhang SX, Wang J, Wang CH, Jia RH, Yan M, Hu FY, et al. Low-dose IL-2 therapy limits the reduction in absolute numbers of circulating regulatory T cells in rheumatoid arthritis. Therapeutic Adv Musculoskeletal Dis 2021, 13, 1759720X211011370. doi: 10.1177/1759720X211011370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Humrich JY, Spee-Mayer CV, Siegert E, Alexander T, Hiepe F, Radbruch A, et al. Rapid induction of clinical remission by low-dose interleukin-2 in a patient with refractory SLE. Ann Rheum Dis 2015, 74, 791–2. doi: 10.1136/ANNRHEUMDIS-2014-206506. [DOI] [PubMed] [Google Scholar]
  • 100. He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, et al. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med 2016, 22, 991–3. doi: 10.1038/nm.4148. [DOI] [PubMed] [Google Scholar]
  • 101. Von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis 2016, 75, 1407–15. doi: 10.1136/annrheumdis-2015-207776. [DOI] [PubMed] [Google Scholar]
  • 102. Humrich JY, von Spee-Mayer C, Siegert E, Bertolo M, Rose A, Abdirama D, et al. Low-dose interleukin-2 therapy in refractory systemic lupus erythematosus: an investigator-initiated, single-centre phase 1 and 2a clinical trial. Lancet Rheumatol 2019, 1, e44–54. doi: 10.1016/S2665-9913(19)30018-9. [DOI] [PubMed] [Google Scholar]
  • 103. Shao M, He J, Zhang R, Zhang X, Yang Y, Li C, et al. Interleukin-2 deficiency associated with renal impairment in systemic lupus Erythematosus. J Interferon Cytokine Res 2019, 39, 117–24. doi: 10.1089/jir.2018.0016. [DOI] [PubMed] [Google Scholar]
  • 104. Zhao C, Chu Y, Liang Z, Zhang B, Wang X, Jing X, et al. Low dose of IL-2 combined with rapamycin restores and maintains the long-term balance of Th17/Treg cells in refractory SLE patients. BMC Immunol 2019, 20, 32. doi: 10.1186/S12865-019-0305-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. He J, Zhang R, Shao M, Zhao X, Miao M, Chen J, et al. Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis 2020, 79, 141–9. doi: 10.1136/ANNRHEUMDIS-2019-215396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Rosenzwajg M, Lorenzon R, Cacoub P, Pham HP, Pitoiset F, El Soufi K, et al. Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial. Ann Rheum Dis 2019, 78, 209–17. doi: 10.1136/ANNRHEUMDIS-2018-214229. [DOI] [PubMed] [Google Scholar]
  • 107. Sheng-Xiao Z, Xiao-Wen M, Xiao-Qing L, Miao M, Xiao-Yan W, Hong-Qing N, et al. SAT0181 low dose interleukin-2 combined with tocilizumab selectively increases regulatory t cells helping refractory rheumatoid arthritis patients achieve remission more rapidly. Ann Rheum Dis 2017, 76, 839. doi: 10.1136/ANNRHEUMDIS-2017-EULAR.3611. [DOI] [Google Scholar]
  • 108. Dong S, Hiam-Galvez KJ, Mowery CT, Herold KC, Gitelman SE, Esensten JH, et al. The effect of low-dose IL-2 and Treg adoptive cell therapy in patients with type 1 diabetes. JCI Insight 2021, 6, e147474. doi: 10.1172/JCI.INSIGHT.147474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Atkinson MA. Pancreatic biopsies in type 1 diabetes: revisiting the myth of Pandora’s box. Diabetologia 2014, 57, 656–9. doi: 10.1007/s00125-013-3159-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Franz B, Fritzsching B, Riehl A, Oberle N, Klemke CD, Sykora J, et al. Low number of regulatory T cells in skin lesions of patients with cutaneous lupus erythematosus. Arthritis Rheum 2007, 56, 1910–20. doi: 10.1002/ART.22699. [DOI] [PubMed] [Google Scholar]
  • 111. Bindon C, Czerniecki M, Ruell P, Edwards A, McCarthy WH, Harris R, et al. Clearance rates and systemic effects of intravenously administered interleukin 2 (IL-2) containing preparations in human subjects. Br J Cancer 1983, 47, 123–33. doi: 10.1038/bjc.1983.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Klatzmann D, Abbas AK.. The promise of low-dose interleukin-2 therapy for autoimmune and inflammatory diseases. Nat Rev Immunol 2015, 15, 283–94. doi: 10.1038/nri3823. [DOI] [PubMed] [Google Scholar]
  • 113. Seelig E, Howlett J, Porter L, Truman L, Heywood J, Kennet J, et al. The DILfrequency study is an adaptive trial to identify optimal IL-2 dosing in patients with type 1 diabetes. JCI Insight 2018, 3, e99306. doi: 10.1172/JCI.INSIGHT.99306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Whangbo JS, Kim HT, Nikiforow S, Koreth J, Alho AC, Falahee B, et al. Functional analysis of clinical response to low-dose IL-2 in patients with refractory chronic graft-versus-host disease. Blood Adv 2019, 3, 984–94. doi: 10.1182/BLOODADVANCES.2018027474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Schinnerling K, Aguillón JC, Catalán D, Soto L.. The role of interleukin-6 signalling and its therapeutic blockage in skewing the T cell balance in rheumatoid arthritis. Clin Exp Immunol 2017, 189, 12–20. doi: 10.1111/cei.12966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Rabinovitch A, Suarez-Pinzon WL, James Shapiro AM, Rajotte RV, Power R.. Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. Diabetes 2002, 51, 638–45. doi: 10.2337/DIABETES.51.3.638. [DOI] [PubMed] [Google Scholar]
  • 117. Levin AM, Bates DL, Ring AM, Krieg C, Lin JT, Su L, et al. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine.’. Nature 2012, 484, 529–33. doi: 10.1038/nature10975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Peterson LB, Bell CJM, Howlett SK, Pekalski ML, Brady K, Hinton H, et al. A long-lived IL-2 mutein that selectively activates and expands regulatory T cells as a therapy for autoimmune disease. J Autoimmun 2018, 95, 1–14. doi: 10.1016/J.JAUT.2018.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Khoryati L, Pham MN, Sherve M, Kumari S, Cook K, Pearson J, et al. An IL-2 mutein engineered to promote expansion of regulatory T cells arrests ongoing autoimmunity in mice. Sci Immunol 2020, 5, eaba5264. doi: 10.1126/SCIIMMUNOL.ABA5264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Chen AC, Cai X, Li C, Khoryati L, Gavin MA, Miao CH.. A Treg-selective IL-2 mutein prevents the formation of factor VIII inhibitors in hemophilia mice treated with factor VIII gene therapy. Front Immunol 2020, 11, 638. doi: 10.3389/fimmu.2020.00638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Pham MN, Khoryati L, Jamison BL, Hayes E, Sullivan JM, Campbell DJ, et al. In vivo expansion of antigen-specific regulatory T cells through staggered Fc.IL-2 mutein dosing and antigen-specific immunotherapy. ImmunoHorizons 2021, 5, 782–91. doi: 10.4049/immunohorizons.2100051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Hirai T, Ramos TL, Lin PY, Simonetta F, Su LL, Picton LK, et al. Selective expansion of regulatory T cells using an orthogonal IL-2/IL-2 receptor system facilitates transplantation tolerance. J Clin Invest 2021, 131, e139991. doi: 10.1172/JCI139991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Dixit N, Fanton C, Langowski JL, Kirksey Y, Kirk P, Chang T, et al. NKTR-358: A novel regulatory T-cell stimulator that selectively stimulates expansion and suppressive function of regulatory T cells for the treatment of autoimmune and inflammatory diseases. Journal of Translational Autoimmunity 2021, 4, 100103. doi: 10.1016/J.JTAUTO.2021.100103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Fanton C, Furie R, Chindalore V, Levin R, Diab I, Dixit N, et al. Selective expansion of regulatory T cells by NKTR-358 in healthy volunteers and patients with systemic lupus erythematosus. J Transl Autoimmun 2022, 5, 100152. doi: 10.1016/J.JTAUTO.2022.100152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Wilson MS, Pesce JT, Ramalingam TR, Thompson RW, Cheever A, Wynn TA.. Suppression of murine allergic airway disease by IL-2: anti-IL-2 monoclonal antibody-induced regulatory T cells. J Immunol 2008, 181, 6942–54. doi: 10.4049/jimmunol.181.10.6942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Webster KE, Walters S, Kohler RE, Mrkvan T, Boyman O, Surh CD, et al. In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J Exp Med 2009, 206, 751–760. doi: 10.1084/JEM.20082824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Park YH, Koo SK, Kim Y, Kim HM, Joe IY, Park CS, et al. Effect of in vitroexpanded CD4(+)CD25(+)Foxp3(+) regulatory T cell therapy combined with lymphodepletion in murine skin allotransplantation. Clin Immunol 2010, 135, 43–54. doi: 10.1016/J.CLIM.2009.11.011. [DOI] [PubMed] [Google Scholar]
  • 128. Liu R, Zhou Q, La Cava A, Campagnolo DI, Van Kaer L, Shi FD.. Expansion of regulatory T cells via IL-2/anti-IL-2 mAb complexes suppresses experimental myasthenia. Eur J Immunol 2010, 40, 1577–89. doi: 10.1002/eji.200939792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Yan JJ, Lee JG, Jang JY, Koo TY, Ahn C, Yang J.. IL-2/anti-IL-2 complexes ameliorate lupus nephritis by expansion of CD4+CD25+Foxp3+ regulatory T cells. Kidney Int 2017, 91, 603–15. doi: 10.1016/j.kint.2016.09.022. [DOI] [PubMed] [Google Scholar]
  • 130. Smaldini PL, Trejo F, Cohen JL, Piaggio E, Docena GH.. Systemic IL-2/anti-IL-2Ab complex combined with sublingual immunotherapy suppresses experimental food allergy in mice through induction of mucosal regulatory T cells. Allergy 2018, 73, 885–95. doi: 10.1111/all.13402. [DOI] [PubMed] [Google Scholar]
  • 131. Trotta E, Bessette PH, Silveria SL, Ely LK, Jude KM, Le DT, et al. A human anti-IL-2 antibody that potentiates regulatory T cells by a structure-based mechanism. Nat Med 2018, 24, 1005–14. doi: 10.1038/S41591-018-0070-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Karakus U, Sahin D, Mittl PRE, Mooij P, Koopman G, Boyman O.. Receptor-gated IL-2 delivery by an anti-human IL-2 antibody activates regulatory T cells in three different species. Sci Transl Med 2020, 12, eabb9283. doi: 10.1126/scitranslmed.abb9283. [DOI] [PubMed] [Google Scholar]
  • 133. Ward NC, Yu A, Moro A, Ban Y, Chen X, Hsiung S, et al. IL-2/CD25: a long-acting fusion protein that promotes immune tolerance by selectively targeting the IL-2R on regulatory T cells. J Immunol 2018, 201, 2579–92. doi: 10.4049/JIMMUNOL.1800907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Ward NC, Lui JB, Hernandez R, Yu L, Struthers M, Xie J, et al. Persistent IL-2 receptor signaling by IL-2/CD25 fusion protein controls diabetes in NOD mice by multiple mechanisms. Diabetes 2020, 69, 2400–13. doi: 10.2337/DB20-0186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Xie JH, Zhang Y, Loubeau M, Mangan P, Heimrich E, Tovar C, et al. Mouse IL-2/CD25 fusion protein induces regulatory T cell expansion and immune suppression in preclinical models of systemic lupus erythematosus. J Immunol 2021, 207, 34–43. doi: 10.4049/jimmunol.2100078. [DOI] [PubMed] [Google Scholar]
  • 136. Moro A, Gao Z, Wang L, Yu A, Hsiung S, Ban Y, et al. Dynamic transcriptional activity and chromatin remodeling of regulatory T cells after varied duration of interleukin-2 receptor signaling. Nat Immunol 2022, 23, 802–13. doi: 10.1038/s41590-022-01179-1. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


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

RESOURCES