Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

In vivo Expansion of Naïve CD4+CD25high FOXP3+ Regulatory T Cells in Patients with Colorectal Carcinoma after IL-2 Administration

Abstract

Regulatory T cells (Treg cells) are increased in context of malignancies and their expansion can be correlated with higher disease burden and decreased survival. Initially, interleukin 2 (IL-2) has been used as T-cell growth factor in clinical vaccination trials. In murine models, however, a role of IL-2 in development, differentiation, homeostasis, and function of Treg cells was established. In IL-2 treated cancer patients a further Treg-cell expansion was described, yet, the mechanism of expansion is still elusive. Here we report that functional Treg cells of a naïve phenotype - as determined by CCR7 and CD45RA expression - are significantly expanded in colorectal cancer patients. Treatment of 15 UICC stage IV colorectal cancer patients with IL-2 in a phase I/II peptide vaccination trial further enlarges the already increased naïve Treg-cell pool. Higher frequencies of T-cell receptor excision circles in naïve Treg cells indicate IL-2 dependent thymic generation of naïve Treg cells as a mechanism leading to increased frequencies of Treg cells post IL-2 treatment in cancer patients. This finding could be confirmed in naïve murine Treg cells after IL-2 administration. These results point to a more complex regulation of Treg cells in context of IL-2 administration. Future strategies therefore might aim at combining IL-2 therapy with novel strategies to circumvent expansion and differentiation of naïve Treg cells.

Introduction

Human regulatory T cells (Treg cells) have been characterized as CD4+CD25high T cells with inhibitory function [1]. They are crucial for the preservation of T-cell homeostasis and self-tolerance and regulate the immune responses to alloantigens, pathogens and tumors [2]. Both in humans and animal models activation of Treg cells results in exertion of their full suppressive function [3], [4]. Natural Treg cells are generated in the thymus as a distinct lineage of anergic CD4+ T cells bearing self-reactive T-cell receptors, although cells with similar characteristics can also be generated in the periphery under appropriate conditions [5]. Typically, Treg cells express cytotoxic T-lymphocyte–associated-antigen 4 (CTLA-4) and glucocorticoid-induced tumor-necrosis-factor receptor-related protein (GITR), although both molecules can also be expressed by activated T cells [6], [7]. The transcription factor forkhead box P3 (FOXP3) has been demonstrated to be expressed exclusively on Treg cells in the mouse [2], while data concerning its expression in humans are not as clear-cut [8], [9]. In numerous murine tumor models increased frequencies of CD4+CD25high Treg cells seem to be a hallmark of tumor progression and metastasis [10], [11]. Moreover, efficient anti-tumor immune responses are induced by deletion of these cells resulting in complete tumor regression [12], [13]. In humans, we and others have demonstrated that CD4+CD25highFOXP3+ Treg cells are also expanded in patients with solid tumors and hematologic malignancies and contribute to the overall immunosuppression in these patients [14], [15]. Numerous animal models over the last years could demonstrate that increased numbers of Treg cells are beneficial for tumor growth while depletion of Treg cells can lead to tumor regression [16]. In humans, administration of an IL-2 immunotoxin to tumor patients results in decreased numbers of Treg cells and higher responses against simultaneously administered tumor peptides [17], [18]. Several studies over the last years have addressed the question of Treg-cell frequencies in colorectal cancer patients [19], [20], [21]. These reports could demonstrate increased number of FOXP3+ Treg cells in the peripheral blood, tumor-draining lymph nodes and in close tumor proximity [19], [20], [21]. Still, the question why Treg cells are expanded in human tumors remains elusive.

CD45RO expression has been primarily linked to Treg cells, which led to the assumption that Treg cells belong to the memory T-cell compartment [4], [22]. Recently, a Treg-cell population with a naïve phenotype (CCR7+CD45RA+) was identified in healthy individuals [23], [24]. These naïve Treg cells proliferated vigorously in response to auto-antigens suggesting that particularly this subpopulation was specific for self rather than foreign antigens [23]. Persistence of naïve Treg cells has been described throughout adult life [25], [26], although it must be pointed out that this pool of naïve Treg cells is relatively small in peripheral blood of healthy individuals [27]. In patients with multiple myeloma (MM), however, we could demonstrate an expansion of naïve Treg cells [28]. We could validate this finding for a number of hematologic malignancies and patients with solid tumors and even show strong suppressive function for the naïve Treg-cell population [29]. Comparison of levels of T-cell receptor excision circles (TREC) in Treg cells of healthy individuals addressed the diversity and developmental stage of thymic emigrants as well as peripheral blood Treg cells [30], [31]. As expected, TREC numbers were significantly higher in thymic emigrants compared to peripheral blood derived Treg cells, which supports thymic development of human CD4+CD25high Treg cells [31].

On the one hand, IL-2 is both important for the development and expansion of effector T cells and also critical in the context of immune tolerance [32]. On the other hand, experiments performed in murine models established that IL-2 and its downstream effector molecules are essential for the generation, maintenance, and function of Treg cells [33], [34], [35], [36]. Several studies have addressed the role of IL-2 administration on the frequency and function of human Treg cells in cancer patients and demonstrated that treatment with recombinant IL-2 induces an expansion of Treg cells in peripheral blood, thereby interfering with efficient anti-tumor immune responses [37], [38], [39], [40], [41], [42], [43]. These studies pointed to a peripheral expansion of Treg cells post IL-2 therapy [41]. It was further suggested that altered migratory behavior due to increased expression of CCR4 and CXCR4 on Treg cells might be associated with peripheral expansion and increased migration to the tumor site [41]. In two recent studies however, Correale et al. demonstrated that the combination of chemotherapy with GM-CSF and IL-2 administration leads to reduced Treg-cell numbers [44], [45]. These findings suggested a more complex regulation of Treg-cell frequency, distribution and function than previously thought. Moreover, it remains unclear whether different mechanisms might be responsible for frequency changes of Treg cells in these different patient populations treated with IL-2.

In the current study, we investigated the impact of IL-2 administration on the frequency and function of CD4+CD25highFOXP3+ Treg cells. We provide clear evidence for an increased expansion of naïve Treg cells particularly post IL-2 therapy as a major mechanism of overall Treg-cell expansion in these cancer patients. Our data clearly suggest an increase of newly generated Treg cells in cancer patients. These novel findings are of particular interest for strategies targeting Treg cells in cancer patients.

Results

Increased frequencies of CD4+CD25highFOXP3+ Treg cells in peripheral blood of patients with metastatic colorectal cancer

Within a clinical phase I/II combined chemoimmunotherapy trial of patients with metastatic colorectal cancer we assessed frequencies of FOXP3-expressing CD4+CD25high Treg cells in peripheral blood before initiation of therapy in comparison to healthy controls (Fig. 1A). The frequency of Treg cells in healthy donors (n = 22, 2.9%±1.2%) was comparable to previously published results (Fig. 1B) [46], [47]. In contrast, individuals with colorectal cancer assessed before initiation of treatment (n = 15, 4.7%±1.2%, p<0.001) showed significantly increased frequencies of Treg cells compared to healthy individuals (Fig. 1A and 1B). We also assessed previously described surface receptors associated with Treg cells including CTLA4 and GITR on CD4+CD25highFOXP3+ T cells in colorectal cancer patients as well as healthy individuals. As depicted in Figure 1C, we observed comparable expression of both molecules on Treg cells from healthy donors and colorectal cancer patients while percentages of CD4+CD25highFOXP3+ Treg cells expressing either molecule were significantly increased in colorectal cancer patients (CTLA4: 3.7%±1.2% vs. 1.5%±0.6%, p<0.001; GITR: 1.7%±0.8% vs. 0.5%±0.2%, p<0.001) (Fig. 1D).

thumbnail
Figure 1. Frequency of CD4+CD25highFOXP3+ Treg cells.

(A) Flow cytometric analysis of CD25 and FOXP3 on peripheral blood derived CD4+ T cells from a representative healthy individual (left panel) and a representative colorectal cancer patient before treatment (right panel). Numbers represent percentage of events within the gate. (B) Frequency of CD4+CD25highFOXP3+ Treg cells in 22 healthy donors and 15 colorectal cancer patients (CRC) before treatment. Each dot represents a single individual assessed in the respective group; mean expression (line) of all samples in each group is also shown (*, p<0.05, Student's t test). (C) CTLA4 (top) and GITR expression (bottom) in CD4+CD25highFOXP3+ Treg cells of healthy donors (left, grey fill) and colorectal cancer patients (right, grey fill). Isotype control (black line). (D) Frequency of CTLA4 (left) and GITR (right) expressing CD4+CD25highFOXP3+ Treg cells in healthy donors (white) and colorectal cancer patients (grey, CRC) before treatment. Shown here are median, 25th and 75th percentile (box), 10th and 90th percentile (whiskers) and outliers (dots), (*, p<0.05, Student's t test).

https://doi.org/10.1371/journal.pone.0030422.g001

Expansion of CD4+CD25highFOXP3+ Treg cells after chemoimmunotherapy including low-dose IL-2 administration

Next, we investigated changes in frequency of Treg cells in respect to IL-2 administration (Fig. 2). From the 15 patients included in the trial, 12 successfully completed the whole protocol, while 3 patients were rapidly progressing under therapy and deceased shortly thereafter. Blood samples from the 12 patients (P1–12) who completed the whole treatment were taken after completion of IL-2 treatment following vaccination. For the 3 rapidly progressing patients (P13–15) who were taken off study, Treg-cell frequencies were assessed at the last time point were material was available. As depicted for patient P11 in Figure 2A we observed an expansion of CD4+CD25highFOXP3+ Treg cells in the majority of patients. When assessing all patients, 11 colorectal cancer patients had higher frequencies post therapy while 2 (P3, P15) had lower frequencies at the second time point of analysis and two other patients (P4, P5) showed similar frequencies at both time points (Fig. 2B). No feature (laboratory test, treatment or clinical parameter) we have assessed so far showed an association with changes in Treg-cell frequency in these patients (data not shown). In addition, we did not observe any cytopenia greater grade 2 during therapy. Overall, in the majority of patients the frequency of Treg cells after combined chemoimmunotherapy was increased compared to the initial frequencies before treatment (5.8%±1.7% vs. 4.7%±1.2%, p<0.05) (Fig. 2C) as well as in comparison to healthy donors (5.8%±1.7% vs. 2.9%±1.2%, p<0.001). When assessing the absolute numbers of Treg cells we observed the same trend; total numbers of Treg cells were increased after chemoimmunotherapy (after: 29.2×106/l±20.5×106/l vs. before: 21.3×106/l±17.1×106/l, p<0.005) and only one of the rapidly progressing patients showed reduced numbers of Treg cells (Fig. 2D).

thumbnail
Figure 2. Analysis of frequencies of CD4+CD25highFOXP3+ Treg cells after chemoimmunotherapy.

(A) Flow cytometric analysis of CD25 and FOXP3 on peripheral blood derived CD4+ T cells from a representative colorectal cancer patient before (left panel) and after chemoimmunotherapy (right panel). Numbers represent percentage of events within the gate. (B) Serial analysis of frequencies of CD4+CD25highFOXP3+ Treg cells in colorectal cancer patients before and after chemoimmunotherapy. (C) Frequency of CD4+CD25highFOXP3+ Treg cells in 15 colorectal cancer patients before (light grey box) and after (dark grey box) chemoimmunotherapy. Shown here are median, 25th and 75th percentile (box), 10th and 90th percentile (whiskers) and outliers (dots), (*, p<0.05, Student's t test). (D) Serial analysis of total numbers of CD4+CD25highFOXP3+ Treg cells in colorectal cancer patients before and after chemoimmunotherapy.

https://doi.org/10.1371/journal.pone.0030422.g002

Among the 15 colorectal cancer patients, 4 patients had progressive disease at the time of analysis while 11 showed a response or stable disease. Comparing these two patient cohorts revealed no significant difference in the proportion of Treg cells, whereas both patient cohorts showed higher frequencies of Treg cells compared to healthy controls (data not shown). Next, we assessed if Treg-cell frequencies were indicative of longer freedom from treatment failure or overall survival or if these 2 parameters were linked to the expansion of Treg cells, yet no statistically significant correlation was detected (data not shown). Our data indicate that an expansion of CD4+CD25highFOXP3+ Treg cells occurred in the majority of colorectal cancer patients after IL-2 administration as part of combined chemoimmunotherapy.

Inhibitory function of CD4+CD25high Treg cells from colorectal cancer patients

In 4 colorectal cancer patients sufficient numbers of highly purified CD4+CD25high Treg cells (Fig. 3A and B) were isolated from peripheral blood by flow cytometric cell sorting before and after vaccination to analyze their inhibitory function in comparison to Treg cells from healthy controls (n = 4). Proliferation of allogeneic conventional CD4+CD25 T cells stimulated by beads coated with CD3 and CD28 mAbs was used as the read out to assess inhibitory function of CD4+CD25high Treg cells [28]. Proliferation of allogeneic conventional CD4+CD25 T cells was significantly inhibited when highly purified CD4+CD25high T cells from healthy donors were added at a 1∶1 ratio (white bar, Fig. 3C, p<0.001). On a cell-to-cell basis highly purified CD4+CD25high Treg cells from colorectal cancer patients (before initiation of therapy) showed an equally strong inhibitory function on conventional CD4+CD25 T-cell proliferation (dark grey bar, Fig. 3C, p<0.001). Titration experiments demonstrated that the inhibitory function of Treg cells from healthy individuals and colorectal cancer patients was comparable also at lower Treg∶Tconv ratios (data not shown). Moreover, after IL-2 treatment of colorectal cancer patients (after treatment), Treg cells had equal suppressive function on conventional CD4+CD25 T-cell proliferation when compared to Treg cells isolated before start of therapy (light grey bar, Fig. 3C, p<0.001). Taken together, these data suggest, that Treg cells from colorectal cancer patients have normal suppressive function.

thumbnail
Figure 3. Functional analysis of CD4+CD25highFOXP+ Treg cells.

(A) CD4+ cells were separated by flow cytometric cell sorting into conventional CD4+CD25 and regulatory CD4+CD25high T cells as defined by their expression of CD25. (B) Re-analysis of FOXP3 expression in CD4+CD25 Tconv (left, grey fill) and CD4+CD25high Treg cells (right, grey fill) post cell sorting. Isotype control (black line). (C) Reduction of proliferation of CD4+CD25 Tconv cells stimulated with beads coated with CD3 and CD28 mAbs (black bar) by highly purified CD4+CD25highFOXP3+ Treg cells from healthy donors (white bar) or colorectal cancer patients before (dark grey bar) and after therapy (light grey bar).

https://doi.org/10.1371/journal.pone.0030422.g003

Expansion of fully functional naïve Treg cells in colorectal cancer patients after chemoimmunotherapy

It has been previously reported by us and others that differential expression of the cell surface receptors CD45RA and CCR7 can be used to differentiate between naïve, central and effector memory Treg cells (Fig. 4A) [23], [28], [48]. This approach can be used to address the question, if naïve, central or effector memory Treg cells contribute to the overall expansion of Treg cells in patients with solid tumors and how treatment, particularly administration of IL-2 influences the frequency and function of naïve versus memory CD4+CD25highFOXP3+ Treg cells. In healthy individuals, naïve CCR7+CD45RA+ Treg cells were hardly detectable (Fig. 4B). Treg cells were almost exclusively of a memory phenotype (Fig. 4C and D) with a higher frequency of TEM cells (Fig. 4D). In contrast, in colorectal cancer patients we observed a significantly higher number of Treg cells with a CCR7+CD45RA+ naïve phenotype (Fig. 4B). This was further accompanied by an increase of Treg cells with a central memory phenotype in colorectal cancer patients prior to therapy (Fig. 4C) while effector memory Treg cell levels were comparable in colorectal cancer patients (prior to therapy) and healthy donors (Fig. 4D). However, most surprising, after IL-2 treatment, expansion of Treg cells almost exclusively occurred within the naïve Treg-cell population (Fig. 4B) while frequencies of central and effector memory Treg cells remained unchanged (Fig. 4C and D). To further characterize the increased subset of naïve CD4+CD25highFOXP3+ Treg cells, we assessed the expression of intracellular CTLA4 or GITR expression. Comparable to the data obtained for the total Treg-cell population both molecules were expressed at similar levels on a per cell basis on naïve Treg-cells from healthy individuals and cancer patients irrespective of IL-2 treatment (data not shown) while percentages of CD4+CD25highFOXP3+ Treg cells expressing either molecule were significantly increased in colorectal cancer patients (CTLA4: 0.31%±0.23% vs. 0.05%±0.01%, p<0.05; GITR: 0.10%±0.07% vs. 0.02%±0.01%, p<0.05) with a further increase after IL-2 administration (CTLA4: 0.78%±0.56% vs. 0.31%±0.23%, p<0.001; GITR: 0.24%±0.19% vs. 0.10%±0.07%, p<0.05) (Fig. S1).

thumbnail
Figure 4. Increase of naïve CD4+CD25highFOXP3+ Treg cells in colorectal cancer patients after chemoimmunotherapy.

(A) Strategy of flow cytometric analysis of CCR7 and CD45RA expression on the surface of CD4+CD25highFOXP3+ Treg cells as exemplified for a representative healthy donor (left) and a representative colorectal cancer patient (right). Frequencies of (B) CCR7+CD45RA+ naïve Treg cells (Tnaive), (C) CCR7+CD45RA central memory Treg cells (TCM), and (D) CCR7CD45RA effector memory Treg cells (TEM) were assessed in peripheral blood of colorectal cancer patients (n = 15) before (light grey bars) and after therapy (dark grey bars) as well as healthy individuals (white bars, n = 22). Significant differences (p<0.05, Student's t test) between healthy donors and colorectal cancer patients before and after chemoimmunotherapy are marked by an asterisk (*). Error bars represent SD. (E) Assessment of regulatory function of naïve and memory CD4+CD25high Treg cells sorted according to their CD45RA expression from colorectal cancer patients. Reduction of proliferation of CD4+CD25 Tconv cells stimulated with beads coated with CD3 and CD28 mAbs by highly purified naïve and memory CD4+CD25high Treg cells from colorectal cancer patients before and after therapy.

https://doi.org/10.1371/journal.pone.0030422.g004

Next, we assessed the suppressive function of naïve versus memory Treg-cell populations before and after therapy (Fig. 4E). We observed a clear inhibition of conventional CD4+CD25 T-cell proliferation induced by stimulation with CD3/CD28-coated beads by adding sorted Treg cells to the culture irrespective of the subtype (naïve vs. memory) used or the status of therapy (no therapy vs. chemoimmunotherapy) demonstrating that the different subtypes of Treg cells have full suppressive activity independent of IL-2 therapy. Taken together, these data demonstrate a significant expansion of naïve Treg cells post IL-2 treatment. Furthermore, these cells are similarly effective in suppressing conventional T-cell activation when analyzed on a cell-to-cell basis, suggesting an overall higher suppressive effect of these cells in colorectal cancer patients.

Thymic expansion of naïve CD4+CD25highFOXP3+ Treg cells in patients with colorectal cancer after IL-2 administration

As Treg cells with a naïve phenotype were increased in patients with colorectal cancer, particularly post IL-2 treatment, we were interested to assess whether the increase of Treg cells resulted from peripheral expansion or possibly thymic generation of CD4+CD25highFOXP3+ Treg cells. We previously reported that the level of TREC can be used as a marker to estimate the developmental vicinity of Treg cells to the thymus and their division history [28] and therefore applied TREC analysis to address this issue in relation to IL-2 treatment. PB derived CD4+ T cells from two colorectal cancer patients and two age-matched healthy individuals were sorted according to their CD25, CCR7 and CD45RA expression into the appropriate CD4+CD25high Treg-cell subsets, namely Tnaïve, TCM, and TEM (Fig. 5A and B). TREC values for the highly purified Treg-cell subsets were assayed by real-time PCR. As described before, CD4+CD25high Treg cells showed relatively low TREC contents in the naïve T-cell population and TREC levels were below detection threshold in the TCM and TEM subsets (Fig. 5C) [28]. These observations are in line with the concept of antigen-driven peripheral expansion of Treg cells in healthy individuals rather than recent thymic emigration. In contrast, the TREC content on the single cell level in naïve CD4+CD25high Treg cells in colorectal cancer patients was more than two-fold higher in average compared to healthy individuals before initiation of chemoimmunotherapy and even more increased after administration of IL-2 (>4–fold in average, Fig. 5C). These results strongly suggest that the expansion of Treg cells in colorectal cancer patients was nurtured by the generation of new Treg cells in the thymus and the further increase of Treg-cell frequencies post IL-2 administration was strongly associated with an increase of thymic emigrants of naïve Treg cells.

thumbnail
Figure 5. Replicative history of CD4+CD25highFOXP3+ Treg cell populations defined by the expression of CD45RA and CCR7.

CD4+CD25high Treg cells were isolated by flow cytometric cell sorting according to their expression of CD25 as well as CD45RA and CCR7 in three Treg-cell subsets, namely Tnaïve (CD45RA+CCR7+), TCM (CD45RACCR7+), and TEM cells (CD45RACCR7). (A) Strategy of flow cytometric analysis of CD4 and CD25 expression on the surface of CD4+ T cells as exemplified for a colorectal cancer patient. (B) Re-analysis of FOXP3 and CD25 expression (left) as well as CCR7 and CD45RA expression (right) in CD4+CD25high Treg cells. (C) Naïve, central and effector memory CD4+CD25high Treg cells from healthy donors and colorectal cancer patients before and after chemoimmunotherapy were assessed for TREC (T-cell receptor excision circle) content. Genomic DNA of sorted subsets was isolated, and the number of TREC was determined by quantitative real-time PCR. Data are shown as the mean values obtained for 2 independent healthy donors and 2 colorectal cancer patients. Error bars represent SD.

https://doi.org/10.1371/journal.pone.0030422.g005

Administration of IL-2 leads to an expansion of a population of murine “naïve” CD4+ CD25highFOXP3+ Treg cells

To demonstrate that IL-2 administration indeed leads to an increase of naïve Treg cells, we administered IL-2 or PBS i.p. in C57BL/6 mice for 10 days and assessed the frequency of CD4+CD25highFOXP3+ Treg cells in these animals after the treatment period. In comparison to PBS-treated animals a significant expansion of CD4+CD25highFOXP3+ Treg cells occurred after IL-2 administration in spleen, peripheral as well as mesenteric lymph nodes, peripheral blood, thymus, and liver (Fig. 6A). Of particular interest was the increase of Treg cells in the thymus, which could be indicative of an increased thymic output. As a first approximation for vicinity of T cells to thymic output expression of CD45RB on CD4+CD25highFOXP3+ Treg cells was assessed since up to now no true marker for murine naïve Treg cells has been established. We observed a significant increase of CD45RBhigh CD4+CD25highFOXP3+ Treg cells in spleen, peripheral as well as mesenteric lymph nodes, peripheral blood, thymus, and liver of IL-2 treated mice (Fig. 6B) clearly indicating that an increase of naïve Treg cells contributed to the overall expansion of Treg cells after IL-2 administration and that the overall Treg-cell expansion was related to elevated frequencies of naïve Treg cells in the thymus.

thumbnail
Figure 6. IL-2 administration leads to an expansion of “naïve” CD4+CD25highFOXP3+ Treg cells in C57BL/6 mice.

(A) Flow cytometric analysis of CD4 and FOXP3 expression in CD4+ T cells from untreated as well as IL-2-treated animals in spleen, peripheral and mesenteric lymph nodes, peripheral blood, thymus, and liver. Significant differences (p<0.05, Student's t test) between untreated and IL-2 treated animals are marked by an asterisk (*). (B) Analysis of “naïve” CD45RBhigh CD4+CD25highFOXP3+ Treg cells in spleen, peripheral and mesenteric lymph nodes, peripheral blood, thymus, and liver. Significant differences (p<0.05, Student's t test) between untreated and IL-2 treated animals are marked by an asterisk (*). Similar results were obtained in two independent experiments. (C) CD45RB+CD44lowCD62L+ naïve CD4+CD25 Tconv and CD4+CD25high Treg cells were isolated by flow cytometric cell sorting and assessed for TREC content. Genomic DNA of sorted subsets was isolated, and the number of TREC was determined by quantitative real-time PCR (n = 3, p<0.05, Student's t test). Error bars represent SD. Similar results were obtained in three independent experiments.

https://doi.org/10.1371/journal.pone.0030422.g006

To further delineate if IL-2 treatment induces generation of naïve Treg cells in the thymus we sorted CD45RB+CD44lowCD62L+ naïve Tconv and Treg cells from IL-2 as well as PBS treated animals (Fig. 6C) and assessed TREC levels in the naïve Tconv and Treg-cell population. We observed significantly higher levels of TREC in Tconv and Treg-cell populations after IL-2 administration (Fig. 6C) suggesting that IL-2 treatment indeed results in a higher thymic output of naïve Treg cells as observed in IL-2 treated human colorectal cancer patients.

Discussion

In the current study we addressed the question whether IL-2 treatment leads to an expansion of fully functional Treg cells in colorectal cancer patients and whether an increase of Treg cells in colorectal cancer patients is due to peripheral or thymic expansion. Using phenotypic, functional and molecular approaches we demonstrate an increase of CD4+CD25highFOXP3+ Treg cells in colorectal cancer patients already before initiation of an IL-2 containing chemoimmunotherapy regimen. The increase of Treg cells in these patients was further augmented after completion of treatment, clearly demonstrating that IL-2 treatment is associated with an increase in Treg-cell numbers an alarming effect which could potentially influence and dampen the immune response in an immunotherapy protocol. Treg cells expressed phenotypic markers associated with Treg cells (e.g. CTLA-4 and GITR) and suppressed the proliferation of CD4+CD25 Tconv cells. Furthermore, we can clearly show that Treg cells in colorectal cancer patients have normal suppressive function.

In addition, we demonstrate that the expansion of Treg cells in colorectal cancer patients was particularly prominent within the naïve CCR7+CD45RA+ CD4+CD25highFOXP3+ Treg cell population and to a lesser extent in central memory Treg cells, while there was no difference in Treg cells with an effector memory phenotype. Most surprising, expansion of Treg cells post IL-2 treatment was almost exclusively due to an increase of naïve Treg cells. Expansion of naïve Treg cells was further supported by the increase of TREC numbers that were significantly higher in naïve Treg cells from untreated colorectal cancer patients when compared to healthy controls and further increased post IL-2 treatment. To demonstrate that the observed results in humans are dependent on the IL-2 administration, we treated mice with IL-2 and could detect increased levels of Treg cells in these animals. In line with our results in humans, we observed thymic generation of naïve Treg cells after IL-2 treatment as shown by high TREC levels in the naïve Treg-cell population. These findings strongly suggest that Treg-cell expansion in colorectal cancer patients is due to higher thymic output which is further increased by IL-2 treatment.

Over the last years several murine studies showed the importance of IL-2 for Treg cells. IL-2 is a critical growth factor for murine Treg cells, it is responsible for maintenance and regulation of Treg cells in the periphery [33], [34], [35]. IL-2 seems to be involved in the generation of Treg cells during antigen-specific immune responses [49] and has been suggested to be involved in the suppressive function of Treg cells [50], [51]. In human Treg-cell biology, IL-2 is supposed to induce a peripheral expansion of CD4+CD25highFOXP3+ Treg cells as suggested by Wei et al. [41] while it has no effect on FOXP3 expression in conventional T cells. This has been further confirmed in a second study which reported that IL-2 induced STAT-dependent mechanisms are responsible for the selective expression of FOXP3 in Treg cells and in vivo expansion following IL-2 administration indicating a unique programming of CD4+CD25high Treg cells within the IL-2 signaling pathway [52].

Most recently increased frequencies of CD4+CD25highFOXP3+ Treg cells were reported for patients with renal cell carcinoma, malignant melanoma or ovarian cancer patients after IL-2 monotherapy [37], [39], [40], [41] and administration of IL-2 during immune reconstitution after chemotherapy in pediatric sarcomas led to a preferential expansion of Treg cells after cytoreductive chemptherapy [38]. In contrast, patients with metastatic colorectal cancer treated with a combined chemoimmunotherapy containing gemcitabine and FOLFOX-4 (oxaliplatin, fluorouracil, and folinic acid) polychemotherapy followed by the subcutaneous administration of GM-CSF and low-dose IL-2 showed clinical objective responses in the majority of patients associated with a significant reduction in CD4+CD25highFOXP3+ Treg cells as reported by Correale et al. [44], [45].

Our data however support an alternative outcome of combined chemoimmunotherapy as low-dose IL-2 in combination with a peptide-vaccination resulted in increased frequencies of CD4+CD25highFOXP3+ Treg cells, particularly naïve Treg cells.

These rather opposite results might be explained by the differences in the vaccination and chemotherapy protocols, e.g. high-dose vs. low-dose 5-FU, irinotecan vs. oxaliplatin, forgoing of gemcitabine, dosage and schedule of GM-CSF administration, or addition of CAP-1-peptide, in the time course of administration, in the patient cohort under study (metastatic colorectal cancer vs. unselected colorectal cancer), number of patients analyzed, and additional, yet unknown, confounding factors. Further conflicting might be technical issues as it has been previously stated by Baecher-Allan et al. that the assessment of human Treg cells is still difficult and the use of different assays sometimes makes it difficult to compare different studies [47]. Indeed, a recent study in patients with renal cell carcinoma or malignant melanoma suggested an enormously high proportion of naïve Treg cells [39]. However, the number of naïve Treg cells in the healthy control group was also reported to exceed 50% of all Treg cells, a frequency that could never be confirmed by us and others [24], [27].

Irrespective of these experimental differences in earlier studies, we clearly unraveled the expansion of naïve Treg cells to be one of the mechanisms leading to an overall expansion of fully functional Treg cells in colorectal cancer patients which was further augmented by IL-2 therapy. Over the last years differentiation of Treg cells into naïve, central and effector memory Treg cells according to their expression of CCR7 and CD45RA has been established for healthy individuals [23], [24], [25], [31], [53]. We have incorporated this strategy for the analysis of Treg cells in cancer patients and demonstrated an expansion of naïve Treg cells in human multiple myeloma patients and B-CLL patients [28]. Remarkably, in B cell malignancies, increase of Treg cells was associated with peripheral expansion of naïve Treg cells while in patients with colorectal cancer the expansion seems to be thymus dependent as determined by analysis of TREC as an approximation of adjacency of Treg cells to the thymus. Moreover, administration of IL-2 further augmented this increase. Two recent publications underlined the importance of IL-2 for the development of Treg cells in the thymus [54], [55], while other results support a more restricted role for IL-2 of regulating Treg cells in the periphery without alteration of the thymic output [34]. The latter proposition has been further strengthened by data obtained by Wei et al. who could demonstrate expansion of memory Treg cells in vitro by IL-2 treatment [41]. To address whether effects induced by IL-2 were limited to peripheral expansion of mainly memory Treg cells, we treated C57BL/6 mice with IL-2 and assessed the frequencies of Treg cells with a special focus on naïve Treg cells in the periphery and in the thymus. We observed an overall expansion of Treg cells in these animals and detected a particular enrichment of naïve Treg cells in the thymus and peripheral lymph nodes. By assessment of TREC in the naïve Treg-cell population we could demonstrate thymic generation of naïve Treg cells as a result of IL-2 adminstration, clearly suggesting that increased thymic output is an important mechanism leading to increased frequencies of naïve Treg cells post IL-2 treatment and these cells subsequently can differentiate into peripheral Treg cells with a memory phenotype. Whether peripheral expansion might also occur under these conditions might be further studied by administration of IL-2 to thymectomized animals.

The assessment of the source of expanded Treg cells in cancer patients is of particular importance as the mechanism of expansion, an augmented production of Treg cells in the thymus, expansion of Treg cells in the periphery, preferential migration of Treg cells to the tumor site, increased conversion of conventional T cells into Treg cells as well as diminished apoptosis and cell death of peripheral Treg cells, might influence the strategy to therapeutically target Treg cells to increase anti-tumor immunity. E.g. peripherally expanded Treg cells might be deleted by short-term use of cytotoxic agents such as denileukin diftitox while continuing thymic expansion would require either long-term treatment or deletion of the cause of thymic expansion of these inhibitory cells to induce a long-lasting reduction of Treg cells. Other mechanisms such as conversion of differentiated conventional T cells into Treg cells might also contribute to the overall expansion of Treg cells in cancer patients as well the preferential egress of activated Treg cell from the tumor microenvironment into the peripheral blood and will have to be addressed when targeting Treg cells [5].

Taken together, we demonstrate an in vivo expansion of fully functional CD4+CD25highFOXP3+ Treg cells in colorectal cancer patients due to an increase in naïve Treg cells with an increased TREC content. Moreover, naïve Treg cells with a further increase of TREC are expanded post IL-2 treatment clearly pointing to an increased thymic output of naïve Treg cells after IL-2 therapy, a mechanism also observed in mice in vivo post IL-2 treatment. This expansion of Treg cells post administration of IL-2 can potentially hinder an immune response towards co-administered anti-tumor reagents and should therefore be avoided when planning new cancer immunotherapy protocols. The existence of different mechanisms of expansion of Treg cells (thymic vs. peripheral expansion and conversion) highlights the complexity of regulation of these cells and cautions the use of simple strategies targeting these highly regulated cells in future cancer immunotherapy approaches.

Materials and Methods

Patients and clinical parameters

15 HLA-A2+ patients with primary metastatic colorectal cancer were enrolled in this Phase I/II combined chemoimmunotherapy with a HLA-A2 peptide derived from the carcinoembryonic antigen CEA (CAP-1) and irinotecan, 5-fluorouracil, and leucovorin after approval by the institutional review committee at the University of Cologne and the German Drug Administration [56]. All patients signed informed consent. Mean age was 56.2±13.1 years; 8 were male and 7 were female; 12 had colon while 3 had rectal cancer. Mean age for the corresponding healthy controls was 49.5±9.3 years with no significant differences in gender and age. Inclusion criteria required an age of between 18 and 75 years, positive HLA-A2 status, elevated serum CEA (>5 µg/l) and/or CEA-positive tumor, untreated metastatic disease, chemotherapy-free interval after adjuvant treatment of at least 6 months, Karnofsky index >70%, life expectancy of at least 3 months, sufficient bone marrow and liver function, HIV and hepatitis B and C negativity, absence of central nervous system metastases, no immunosuppressant medication, and negative pregnancy test. All patients received low-dose IL-2 (1×106 IU) post vaccination. For vaccination, several approaches were compared in this study, namely a vaccine containing only the CAP-1-peptide, or the CAP-1-peptide together with 50 µg GM-CSF (Novartis) or the CAP-1-peptide together with an oligonucleotide adjuvant (dSLIM, Mologen); as a fourth option, patients obtained autologous CAP-1-pulsed dendritic cells as a cellular vaccine. For the analysis of Treg-cell frequency and function post IL-2 treatment, no statistically significant influence of the different types of vaccinations could be established (data not shown). Two cycles of vaccination and IL-2 treatment were given 2 and 1 week before the first cycle of chemotherapy consisting of 80 mg/m2 irinotecan, 2,000 mg/m2 high-dose 5-FU, and 500 mg/m2 leucovorin (six weekly administrations). Alternating, two cycles of IL-2 and vaccination were combined with one cycle of chemotherapy and repeated three times. After the third cycle, patients were treated with IL-2 and CAP-1-peptide vaccination on a weekly schedule until progressive disease. At baseline and after three cycles of chemoimmunotherapy, patients underwent a leukapheresis to obtain PBMC for diagnostic and therapeutic purposes. PBMC were isolated using Ficoll/Hypaque (Amersham, Uppsala, Sweden) density centrifugation. Staging was performed according to the UICC classification for colorectal cancer. All patients were UICC stage IV. The treatment schedule is provided as Figure S2. Characteristics of the patients studied are summarized in Table 1.

Antibodies and FACS analysis

Phenotype of T cells was defined by flow cytometry using the following antibodies: CD4-FITC, CD45RA-PE-Cy-5, CD4-APC, CD4-APC-Cy-7 (all from Becton Dickinson PharMingen), CD25-PE-Cy7 (BDBiosciences), CCR7-FITC (R&D Systems) as well as the corresponding isotype control antibodies (BDPharMingen). Intracellular staining was performed with the following antibodies: FOXP3-PE or APC (eBioscience), GITR-FITC (R&D Systems), CTLA4-PE or with the appropriate isotype controls (BDPharMingen) [28], [46]. Cells were stained according to the manufacturer's recommendations.

Samples were acquired on a FACSCanto and analyzed with FlowJo software (TreeStar Inc.). Frequencies of CD4+CD25highFOXP3+ T cells are shown as percent values of CD4+ T cells.

Isolation of CD4+CD25high and CD4+CD25 T cells

For functional analysis, CD4+CD25high T cells were purified from PBMC. Briefly, CD4 MACS Beads (Miltenyi Biotec) were used for isolation of CD4+ T cells [28], [46]. After staining with CD25-PE, and CD4-APC (BDPharMingen) according to the manufacturer's recommendations, CD4+CD25high T cells were purified using a FACSDiVa Cell Sorter (BDBiosciences) and used for functional characterization. The CD4+CD25 T cells isolated from healthy individuals were used as effectors to assess Treg cell function independently of potential defects of conventional CD4+ T cells from colorectal cancer patients [28], [46]. The cells were re-analyzed for FOXP3 expression after sorting and routinely showed >95% purity.

Assessment of inhibitory function

To assess the suppressive activity of Treg cells, 5,6-Carboxyfluorescin-Diacetat-Succinimidyl-Ester (CFSE, Sigma-Aldrich) stained CD4+CD25 T cells (5×104/well) were stimulated with magnetic beads (Dynal Biotech) coated with 5% anti-CD3 (OKT3), 14% anti-CD28 (9.3) and 81% anti-MHC class I (W6/32) at a ratio of 3∶1 (cells∶beads) in X-VIVO 15 supplemented with 10% fetal calf serum, 100 U/ml penicillin/streptomycin and 2 mM glutamine (all from Invitrogen). PKH-26 (Sigma-Aldrich)-labelled allogeneic CD4+CD25high T cells or naive or memory CD4+CD25high T cells activated for 20 hours with 10 U/ml IL-2 (Proleukin®, Chiron) and 0.5 µg/ml anti-CD3 mAb in X-VIVO 15 (BioWhittakker) were added at a 1∶1 ratio to the culture and proliferation of CD4+CD25 T cells was determined by assessing CFSE dilution after four days of culture [28].

Isolation of CD4+CD25high T-cell subpopulations for assessment of T-cell receptor excision circles and functional characterization

Briefly, CD4 MACS Beads were used for isolation of CD4+ T cells [46]. After staining with CCR7-FITC, CD25-PE, CD45RA-PE-Cy-5, and CD4-APC, CD4+CD25high T cells and the respective T cell subsets, CCR7+CD45RA+ Tnaïve, CCR7+CD45RA TCM, and CCR7CD45RA TEM cells were purified using a FACSDiVa Cell Sorter and used for either functional characterization or assessment of TREC levels.

DNA was isolated from purified CD4+CD25, CD4+CD25low and CD4+CD25high Tnaïve, TCM, and TEM cells respectively using a DNA Isolation Kit (Roche Diagnostics) following the manufacturer's instructions.

Relative TREC levels were determined using real-time PCR with a LightCycler (Roche Diagnostics) based on specific primers and general fluorescence detection with SYBR Green. All PCR were performed using LightCycler-FastStart DNA Master SYBR Green I kit (Roche Diagnostics). All samples were studied in duplicate reactions using the human TREC primer kit (Search-LC). The number of TREC molecules in the sample was calculated as number of copies per 104 cells (detection limit ≥10 molecules).

Analysis of IL-2 effects on murine Treg cells

Female C57BL/6 mice of 7 weeks were obtained from Elevage Janvier (France) and maintained in our animal facility. In vivo experiments were approved by the Animal Care Commission of Nord-Rhein-Westfalia, Germany (TVZ 9.93.2.10.31.07.089). After either treatment with human IL-2 (1×105 IU/mouse/day, Proleukin) i.p. or PBS as control for 10 days [57], mice were sacrificed and their spleens, thymi, liver, and LN removed. Single-cell suspensions were prepared and stained for flow cytometric analysis. Antibodies used for staining were CD4-PE-Cy7, CD8α-Pacific Blue, CD45RB-APC-Cy7, CD25-Alexa 647, and FOXP3-Alexa 488 as well as appropriate isotype controls (all from BDBiosciences or BioLegend). Samples were acquired on a FACSCantoII and analyzed with FlowJo software. Frequencies of CD4+CD25highFOXP3+ Treg cells are shown as percent values of CD4+ T cells.

Isolation of CD4+CD45RB+CD44lowCD62L+ naïve murine T-cells for assessment of T-cell receptor excision circles

Briefly, CD4 MACS Beads were used for isolation of murine CD4+ T cells from the spleen after treatment with IL-2 as described above [46]. After staining with CD44-FITC, CD62L-PE, CD25-Alexa 647, CD45RB-APC-Cy-7, CD8α-PE-Cy-7, and CD4-Alexa 405, naïve CD45RB+CD44lowCD62L+ conventional CD4+CD25 T cells as well as naïve CD4+CD25+ Treg cells were purified using a FACSDiVa Cell Sorter and used for assessment of TREC levels.

Murine δRec-ψJα TRECs were determined using real-time quantitative polymerase chain reaction (PCR) as described before [58]. Briefly, cells were centrifuged and the pellet frozen at −80°C until analysis. DNA was isolated from purified naïve CD4+CD25 Tconv and naïve CD4+CD25high Treg cells using a DNA Isolation Kit (Roche Diagnostics) following the manufacturer's instructions. Relative TREC levels were determined using real-time PCR containing mδRec primer (5′-GGGCACACAGCAGCTGTG), ψJα primer (5′-GCAGGTTTTTGTAAAGGTGCTCA), and mδRec-ψJα fluorescent probe (5′-FAM-CACAAGCACCTGCACCCTGTGCA-TAMRA-3′). Lysates were separately subjected to amplification of the single-copy CD8β chain gene using a CD8β forward primer (5′-CAGGACCCCAAGGACAAGTACT-3′), CD8β reverse primer (5′-CACTTTCACCATACAAAACTCCTTTG-3′), and CD8β probe (5′-FAMTGAGTTCCTGGCCTCCTGGAGTTCTTC-TAMRA-3′). Reactions contained 0.5 µM of each primer, 0.3 µM fluorescent probe, and Platinum Quantitative PCR Supermix-UDG (Invitrogen) Amplifications were performed in triplicate on an LightCycler 480 II (Roche Diagnostics). Amplification conditions were 50°C for 2 minutes, 95°C for 5 minutes, then 40 cycles of 95°C for 15 seconds, and 60°C for 1 minute. Standards for murine TRECs (mδREC-ψJα) and CD8β were provided by Dr. Y.-W. Chu (Center for Cancer Research, National Institutes of Health). Standard curves were generated as described before [58]. TREC frequency (TREC molecules per 10 000 cells) was determined by normalizing the number of TRECs amplified in the real-time PCR reaction to the number of amplified CD8β molecules.

Statistical analysis

Comparison between paired or unpaired groups was performed using the appropriate Student's t-test. A p-value<0.05 was defined as statistically significant. All statistical analyses were performed using the SPSS statistical software package (SPSS 19, SPSS Inc.). Figures were created using SigmaPlot 12.0 (Systat Software Inc).

Supporting Information

Figure S1.

CTLA4 and GITR expression in naïve Treg cells. Frequency of CTLA4 (left) and GITR (right) expressing naïve CCR7+CD45RA+ CD4+CD25highFOXP3+ Treg cells in healthy donors (white) and colorectal cancer patients before (light grey) and after IL-2 administration (dark grey) before treatment. Shown here are median, 25th and 75th percentile (box), 10th and 90th percentile (whiskers) and outliers (dots), (*, p<0.05, Student's t test).

https://doi.org/10.1371/journal.pone.0030422.s001

(TIF)

Figure S2.

Therapy schedule. Patients were first randomized to receive CAP-1 and IL-2 with different adjuvants (dSLIM, GM-CSF, or none). Subsequently, they were randomized to receive their first vaccination with or without pulsed autologous dendritic cells. Vaccinations (V) and chemotherapy (Chemo) were given in an alternating schedule, starting with two vaccinations.

https://doi.org/10.1371/journal.pone.0030422.s002

(TIF)

Acknowledgments

We are indebted to our patients for their commitment to this study. We thank I. Büchmann, A. Dolf, and P. Wurst for excellent technical assistance, and J. Oldenburg and the Division of Transfusion Medicine for providing us with blood samples from healthy individuals. We thank Dr. Yu-Waye Chu (National Institutes of Health) for providing the TREC protocol and standards for murine TRECs and CD8β.

Author Contributions

Conceived and designed the experiments: MB JLS. Performed the experiments: MB BS MRW BA TG EE SC AL JLS. Analyzed the data: MB JLS. Contributed reagents/materials/analysis tools: EE PAK. Wrote the paper: MB BS MRW BA TG SC AL JLS EE PAK.

References

  1. 1. Sakaguchi S, Miyara M, Costantino CM, Hafler DA (2010) FOXP3+ regulatory T cells in the human immune system. Nature reviews Immunology 10: 490–500.
  2. 2. Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6: 345–352.
  3. 3. Shevach EM (2001) Certified professionals: CD4(+)CD25(+) suppressor T cells. J Exp Med 193: F41–46.
  4. 4. Taams LS, Smith J, Rustin MH, Salmon M, Poulter LW, et al. (2001) Human anergic/suppressive CD4(+)CD25(+) T cells: a highly differentiated and apoptosis-prone population. Eur J Immunol 31: 1122–1131.
  5. 5. Vukmanovic-Stejic M, Zhang Y, Cook JE, Fletcher JM, McQuaid A, et al. (2006) Human CD4+ CD25hi Foxp3+ regulatory T cells are derived by rapid turnover of memory populations in vivo. J Clin Invest 116: 2423–2433.
  6. 6. Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, et al. (2000) Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 192: 303–310.
  7. 7. McHugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, et al. (2002) CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity 16: 311–323.
  8. 8. Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, et al. (2007) Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol 19: 345–354.
  9. 9. Tran DQ, Ramsey H, Shevach EM (2007) Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood 110: 2983–2990.
  10. 10. Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, et al. (2011) Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature 470: 548–553.
  11. 11. Kortylewski M, Xin H, Kujawski M, Lee H, Liu Y, et al. (2009) Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment. Cancer cell 15: 114–123.
  12. 12. Ghiringhelli F, Larmonier N, Schmitt E, Parcellier A, Cathelin D, et al. (2004) CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol 34: 336–344.
  13. 13. Shimizu J, Yamazaki S, Sakaguchi S (1999) Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. J Immunol 163: 5211–5218.
  14. 14. Beyer M, Schultze JL (2006) Regulatory T cells in cancer. Blood 108: 804–811.
  15. 15. Beyer M, Schultze JL (2008) Immunoregulatory T cells: role and potential as a target in malignancy. Curr Oncol Rep 10: 130–136.
  16. 16. Beyer M, Schultze JL (2009) Regulatory T cells: major players in the tumor microenvironment. Current pharmaceutical design 15: 1879–1892.
  17. 17. Mahnke K, Schonfeld K, Fondel S, Ring S, Karakhanova S, et al. (2007) Depletion of CD4+CD25+ human regulatory T cells in vivo: kinetics of Treg depletion and alterations in immune functions in vivo and in vitro. International journal of cancer Journal international du cancer 120: 2723–2733.
  18. 18. Morse MA, Hobeika AC, Osada T, Serra D, Niedzwiecki D, et al. (2008) Depletion of human regulatory T cells specifically enhances antigen-specific immune responses to cancer vaccines. Blood 112: 610–618.
  19. 19. Clarke SL, Betts GJ, Plant A, Wright KL, El-Shanawany TM, et al. (2006) CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS ONE 1: e129.
  20. 20. Yaqub S, Henjum K, Mahic M, Jahnsen FL, Aandahl EM, et al. (2008) Regulatory T cells in colorectal cancer patients suppress anti-tumor immune activity in a COX-2 dependent manner. Cancer Immunol Immunother 57: 813–821.
  21. 21. Ling KL, Pratap SE, Bates GJ, Singh B, Mortensen NJ, et al. (2007) Increased frequency of regulatory T cells in peripheral blood and tumour infiltrating lymphocytes in colorectal cancer patients. Cancer Immun 7: 7.
  22. 22. Thornton AM, Shevach EM (1998) CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med 188: 287–296.
  23. 23. Valmori D, Merlo A, Souleimanian NE, Hesdorffer CS, Ayyoub M (2005) A peripheral circulating compartment of natural naive CD4 Tregs. J Clin Invest 115: 1953–1962.
  24. 24. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, et al. (2009) Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30: 899–911.
  25. 25. Santner-Nanan B, Seddiki N, Zhu E, Quent V, Kelleher A, et al. (2008) Accelerated age-dependent transition of human regulatory T cells to effector memory phenotype. Int Immunol 20: 375–383.
  26. 26. Seddiki N, Santner-Nanan B, Tangye SG, Alexander SI, Solomon M, et al. (2006) Persistence of naive CD45RA+ regulatory T cells in adult life. Blood 107: 2830–2838.
  27. 27. Beyer M, Schultze JL (2007) CD4+CD25highFOXP3+ regulatory T cells in peripheral blood are primarily of effector memory phenotype. J Clin Oncol 25: 2628–2630.
  28. 28. Beyer M, Kochanek M, Giese T, Endl E, Weihrauch MR, et al. (2006) In vivo peripheral expansion of naive CD4+CD25high FoxP3+ regulatory T cells in patients with multiple myeloma. Blood 107: 3940–3949.
  29. 29. Beyer M, Classen S, Endl E, Kochanek M, Weihrauch MR, et al. (2011) Comparative Approach to Define Increased Regulatory T Cells in Different Cancer Subtypes by Combined Assessment of CD127 and FOXP3. Clinical & developmental immunology 2011: 734036.
  30. 30. Hori S, Haury M, Lafaille JJ, Demengeot J, Coutinho A (2002) Peripheral expansion of thymus-derived regulatory cells in anti-myelin basic protein T cell receptor transgenic mice. Eur J Immunol 32: 3729–3735.
  31. 31. Kasow KA, Chen X, Knowles J, Wichlan D, Handgretinger R, et al. (2004) Human CD4+CD25+ regulatory T cells share equally complex and comparable repertoires with CD4+CD25− counterparts. J Immunol 172: 6123–6128.
  32. 32. Malek TR, Bayer AL (2004) Tolerance, not immunity, crucially depends on IL-2. Nat Rev Immunol 4: 665–674.
  33. 33. Bayer AL, Yu A, Adeegbe D, Malek TR (2005) Essential role for interleukin-2 for CD4(+)CD25(+) T regulatory cell development during the neonatal period. J Exp Med 201: 769–777.
  34. 34. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY (2005) A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 6: 1142–1151.
  35. 35. Setoguchi R, Hori S, Takahashi T, Sakaguchi S (2005) 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 201: 723–735.
  36. 36. Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ (2002) Interleukin 2 signaling is required for CD4(+) regulatory T cell function. J Exp Med 196: 851–857.
  37. 37. Ahmadzadeh M, Rosenberg SA (2006) IL-2 administration increases CD4+ CD25(hi) Foxp3+ regulatory T cells in cancer patients. Blood 107: 2409–2414.
  38. 38. Zhang H, Chua KS, Guimond M, Kapoor V, Brown MV, et al. (2005) Lymphopenia and interleukin-2 therapy alter homeostasis of CD4+CD25+ regulatory T cells. Nat Med 11: 1238–1243.
  39. 39. Cesana GC, DeRaffele G, Cohen S, Moroziewicz D, Mitcham J, et al. (2006) Characterization of CD4+CD25+ regulatory T cells in patients treated with high-dose interleukin-2 for metastatic melanoma or renal cell carcinoma. J Clin Oncol 24: 1169–1177.
  40. 40. Sosman JA, Carrillo C, Urba WJ, Flaherty L, Atkins MB, et al. (2008) Three phase II cytokine working group trials of gp100 (210M) peptide plus high-dose interleukin-2 in patients with HLA-A2-positive advanced melanoma. J Clin Oncol 26: 2292–2298.
  41. 41. Wei S, Kryczek I, Edwards RP, Zou L, Szeliga W, et al. (2007) Interleukin-2 administration alters the CD4+FOXP3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res 67: 7487–7494.
  42. 42. van der Vliet HJ, Koon HB, Yue SC, Uzunparmak B, Seery V, et al. (2007) Effects of the administration of high-dose interleukin-2 on immunoregulatory cell subsets in patients with advanced melanoma and renal cell cancer. Clin Cancer Res 13: 2100–2108.
  43. 43. Jensen HK, Donskov F, Nordsmark M, Marcussen N, von der Maase H (2009) Increased intratumoral FOXP3-positive regulatory immune cells during interleukin-2 treatment in metastatic renal cell carcinoma. Clin Cancer Res 15: 1052–1058.
  44. 44. Correale P, Cusi MG, Tsang KY, Del Vecchio MT, Marsili S, et al. (2005) Chemo-immunotherapy of metastatic colorectal carcinoma with gemcitabine plus FOLFOX 4 followed by subcutaneous granulocyte macrophage colony-stimulating factor and interleukin-2 induces strong immunologic and antitumor activity in metastatic colon cancer patients. J Clin Oncol 23: 8950–8958.
  45. 45. Correale P, Tagliaferri P, Fioravanti A, Del Vecchio MT, Remondo C, et al. (2008) Immunity Feedback and Clinical Outcome in Colon Cancer Patients Undergoing Chemoimmunotherapy with Gemcitabine+FOLFOX followed by Subcutaneous Granulocyte Macrophage Colony-Stimulating Factor and Aldesleukin (GOLFIG-1 Trial). Clin Cancer Res 14: 4192–4199.
  46. 46. Beyer M, Kochanek M, Darabi K, Popov A, Jensen M, et al. (2005) Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine. Blood 106: 2018–2025.
  47. 47. Baecher-Allan C, Viglietta V, Hafler DA (2004) Human CD4+CD25+ regulatory T cells. Semin Immunol 16: 89–98.
  48. 48. Tosello V, Odunsi K, Souleimanian NE, Lele S, Shrikant P, et al. (2008) Differential expression of CCR7 defines two distinct subsets of human memory CD4+CD25+ Tregs. Clin Immunol 126: 291–302.
  49. 49. Knoechel B, Lohr J, Kahn E, Bluestone JA, Abbas AK (2005) Sequential development of interleukin 2-dependent effector and regulatory T cells in response to endogenous systemic antigen. J Exp Med 202: 1375–1386.
  50. 50. Thornton AM, Donovan EE, Piccirillo CA, Shevach EM (2004) Cutting edge: IL-2 is critically required for the in vitro activation of CD4+CD25+ T cell suppressor function. J Immunol 172: 6519–6523.
  51. 51. Pandiyan P, Zheng L, Ishihara S, Reed J, Lenardo MJ (2007) CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nat Immunol 8: 1353–1362.
  52. 52. Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, et al. (2006) IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 108: 1571–1579.
  53. 53. Seddiki N, Santner-Nanan B, Tangye SG, Alexander SI, Solomon M, et al. (2005) Persistence of naive CD45RA+ regulatory T cells in adult life. Blood.
  54. 54. Watanabe N, Wang YH, Lee HK, Ito T, Wang YH, et al. (2005) Hassall's corpuscles instruct dendritic cells to induce CD4(+)CD25(+) regulatory T cells in human thymus. Nature 436: 1181–1185.
  55. 55. Burchill MA, Yang JY, Vogtenhuber C, Blazar BR, Farrar MA (2007) IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3(+) regulatory T cells. Journal of Immunology 178: 280–290.
  56. 56. Weihrauch MR, Ansen S, Jurkiewicz E, Geisen C, Xia Z, et al. (2005) Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP-1 peptide and irinotecan, 5-fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res 11: 5993–6001.
  57. 57. Chen X, Oppenheim JJ, Winkler-Pickett RT, Ortaldo JR, Howard OM (2006) 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 36: 2139–2149.
  58. 58. Chu YW, Memon SA, Sharrow SO, Hakim FT, Eckhaus M, et al. (2004) Exogenous IL-7 increases recent thymic emigrants in peripheral lymphoid tissue without enhanced thymic function. Blood 104: 1110–1119.