The prognosis of follicular lymphoma (FL) patients is suspected to be influenced by tumor-infiltrating regulatory T cells (Treg). The mechanism of Treg enrichment in FL and their impact on malignant FL B cells remains to be elucidated. We analyzed 46 fresh lymph node biopsy samples, including FL (n = 20), diffuse large B-cell lymphoma (n = 10), classical Hodgkin lymphoma (n = 9), and reactive lymphadenitis (n = 7). Using multicolor flow cytometry and cell sorting, we observed an accumulation of CD25highCD127low/neg Tregs in FL tissues. These Tregs comprised activated ICOS+ Tregs that were able to suppress not only conventional T cells, but also FL B cells. These FL B cells were able to express ICOSL in vitro and to generate CD25highFoxP3high Tregs expressing ICOS. Treg generation was associated with ICOS/ICOSL engagement and was abrogated by antagonist anti-ICOS and anti-ICOSL antibodies. Interactions between Tregs and FL B cells resulted in ICOSL downregulation on FL B cells. Our results highlight a key role for Tregs in FL pathogenesis and suggest that targeting the ICOS/ICOSL pathway may be a promising immunotherapy for FL treatment. Cancer Res; 76(16); 4648–60. ©2016 AACR.

Follicular lymphoma (FL) is usually considered as an indolent disease, but some patients present resistance to treatment and/or transformation into aggressive lymphoma (1). The clinical outcome of FL patients was shown to be influenced by tumor-infiltrating macrophages and T cells (2–5), including CD4+ regulatory T cells (Treg).

Tregs comprise natural and inducible Tregs, that can modulate immune responses by suppressive mechanisms (6). Accumulation of Tregs was found in various hematologic and nonhematologic malignancies (6). In contrast with solid tumors, in which Tregs suppress antitumor immunity and promote tumor progression (7–10), high amounts of intratumoral FoxP3+ Tregs were associated with a favorable outcome in FL (11–13). This result appears paradoxical, because Tregs from FL tissues were shown to inhibit the function of antitumor CD4+ and CD8+ T cells (14–16). Of note, a similar favorable influence of Tregs has been also reported in classical Hodgkin lymphoma a lymphoma subtype in which the microenvironment is known to play a key role (17). Nonetheless, the favorable prognostic value of Tregs in FL remains controversial because it was not clearly confirmed in recent studies (18–20).

Tregs lack CD127 (IL7 receptor α chain) and usually express CD25 (IL2 receptor α chain), GITR (glucocorticoid-induced TNFR-related protein), CTLA-4 (cytotoxic T lymphocyte associated antigen 4), and CD45RO (21). Expression of the transcription factor FoxP3 (forkhead box P3) is considered as a crucial feature for the development and function of Tregs (21). Lymphoma B cells produce the CCL22 chemokine inducing Treg recruitment (15) and converting conventional T cells (Tconv) to Tregs (22–24). Cell contacts were shown to be required for this conversion, which involves CD70, CD80, and CD86 expression on B cells (22, 24).

ICOS (inducible T cell costimulator, CD278)/ICOSL pathway–driven generation of Tregs has been recently demonstrated in the periphery (25) and microenvironment of solid tumors (26–28). ICOS, a costimulatory molecule of the CD28 family, is expressed on activated T cells. ICOS is involved in T-cell responses upon engagement with its ligand, ICOSL (ICOSLG, B7-H2, CD275), which is normally expressed on B cells, dendritic cells, and monocytes (29). ICOS expression confers an activated phenotype and a strong suppressive capacity to intratumoral Tregs (30). Particular Tregs displaying strong expression of ICOS and CXCR5 and inhibiting CD4+ effector T cells were recently identified in FL tissues, and called follicular regulatory T cells (TFR; ref. 31). This subset is reminiscent of TFR observed in normal follicles in mice (32, 33). Nonetheless, the hypothesis of a direct suppressive function of Tregs on FL B cells has not been fully investigated. These observations prompted us to clarify the role of Tregs in FL, using a combination of flow cytometry, cell sorting, and IHC on tissue samples. As a control group, we analyzed tissue samples from not only benign lymphadenitis, but also lymphoma subtypes in which the microenvironment is suspected to play a significant role, especially cHL, and to a lesser extent diffuse large B-cell lymphoma (DLBCL).

Patients

Fresh biopsy samples from 46 patients were collected at diagnosis. Samples were classified as FL (n = 20), DLBCL (n = 10), cHL (n = 9), and reactive lymphadenitis (rL; n = 7) according to the World Health Organization classification (IARC, Lyon, 2008). DLBCL cases were considered as de novo cases, due to the lack of previous history of FL and absence of FL histologic features. All patients gave informed consent and the study was approved by the ethical board of the Paoli-Calmettes and Albert Bonniot institute.

Cell lines

Burkitt lymphoma cell line Raji and COS-7 cells were obtained from American Type Culture Collection (ATCC). Raji and COS-7 cells were respectively cultured in RPMI 1640 Medium (Life Technologies) supplemented with 10% fetal calf serum (FCS; Lonza) and in DMEM (Life Technologies) supplemented with 10% FCS. These cell lines were not authenticated at the time we performed the study.

Cell isolation and purification

Sample tissues were mechanically disrupted and passed through a nylon filter (BD Bioscience) to obtain mononuclear cells (MC), which were frozen or used immediately for functional assay.

B cells and CD4+ T cells were obtained from MCs by negative selection (Stemcell Technologies). To isolate Tregs and Tconv, purified intratumoral CD4+ T cells were labeled with anti-CD4-PC7, anti-CD25-APC, anti-CD127-FITC, and a viability marker (live/dead aqua) and sorted by FACs (BD Biosciences). To separate ICOS+ and ICOSneg CD4+ T cells, intratumoral CD4+ T cells were labeled with anti-ICOS-PE and live/dead aqua.

Antibodies and flow cytometry

Antibodies are detailed in Supplementary Table S1. To analyze intracellular markers like FoxP3, Helios, Bcl-2, and Ki67, cells were fixed/permeabilized after surface staining and then incubated with specific antibodies using Foxp3 staining buffer set (eBioscience). Data were acquired on LSRII (BD Biosciences) and analyzed using FACS Diva and FlowJo softwares.

Mice model

Naïve wild-type FVB/N (Charles River Laboratory) were used at 6 weeks of age. Mice were maintained in the pathogen-free animal facility "AniCan" at the Cancer Research Center of Lyon. Experiments were conducted in accordance with the European and French laws and were validated by the local animal ethical evaluation committee (CECCAP ethical committee agreement no. CECCAPP-2013-010 and registered at the MESR, #02312.01).

Mice were injected with either anti-mICOS mAb (17G9, rat IgG2) or isotype control (BioXcell; 100 μg/mL) by i.p. route and sacrificed after 24 hours or 48 hours. Blood was collected by intracardiac function, and lymph node and spleen were subjected to mechanical disaggregation to obtain homogenous single-cell suspension. Membrane multiparametric flow cytometry stainings (CD45-AF-700, CD3-APC-H7, CD19-BV605, ICOS-L-PE, and live/dead Aqua) were performed to evaluate ICOS expression on CD19+ B cells after gating on viable cells (live/dead Aquaneg). A multistaining including an IgG2a-PE in place of anti-ICOSL Mab (FMO) was used as a negative control. Flow cytometry analyses were performed on an LSR-II (Becton Dickinson), and results were analyzed using FlowJo software.

T-cell proliferation and suppression assays

To study proliferative capacity, CellTrace violet (Life Technologies)–labeled Tregs and Tconv were activated by CD3/CD28 dynabeads (Life Technologies; 1 bead for 1 cell) in the presence of recombinant human IL2 (rIL2, 100 IU/mL; Chiron) for 5 days. To evaluate the suppressive effects of Tregs on CD4+ Tconv, CellTrace violet–labeled Tconv were activated by CD3/CD28 dynabeads in the presence of rIL2 (100 IU/mL) and Tregs for 4 days. Cells were harvested and stained with anti-CD2-PECF-594 and live/dead near-IR. Proliferation was evaluated by Division index (DI) using the proliferation tool of FlowJo software. Supernatants of coculture were collected and IFNγ was further measured by ELISA (BD Biosciences).

B-cell suppression assay

FL B cells purified from tissues were stained with CellTrace violet and were cultured with autologous or allogeneic Tregs (1/1 ratio) in the presence of rIL2 (100 IU/mL) for 5 days. Anti-IgA/G/M (10 μg/mL; Jackson ImmunoReseach) and CpG-B (2 μg/mL; Invivogen) were added to stimulate FL B cells as previously described (34). To study T-cell–dependent suppression of Tregs, an equal number of Tconv and/or Tregs were added to stimulated B cells. Supernatant of coculture was collected to measure IL6 by ELISA (R&D Systems).

Kinetics of ICOSL expression on lymphoma B cells

Purified FL B cells or total MCs were cultured in culture medium. Cells were harvested and stained with live/dead aqua, anti-CD19-FITC, anti-ICOSL-PE, and anti-CD86-APC before and during culture. FL B cells were cultured overnight until ICOSL was expressed. Then, different ratios of intratumoral ICOS+ or ICOSneg CD4+ T cells were added on the B cell culture. In some experiments, anti-ICOS–blocking antibody (314.8 clone, produced in our laboratory) or isotype control (Ctrl) IgG1 (Sigma) were added at 10 μg/mL.

Treg induction assay

CellTrace violet-labeled intratumoral CD4+ T cells were cultured alone or with autologous or allogeneic FL B cells expressing ICOSL at 1.5/1 ratio in the presence of rIL2 (100 IU/mL). Anti-ICOS (314.8 clone) or anti-ICOSL (MIH.12 clone, eBioscience) blocking antibodies or isotype Ctrl IgG1 were added at 10 μg/mL for each. After 5 to 6 days, cells were harvested and surface stained with anti-CD4-PC7, anti-CD19-PECF-594, anti-CD25-APC, anti-ICOS-PE, and live/dead near-IR before fixation/permeabilization and intracellular staining with FoxP3-FITC.

Cells harvested after 5 to 6 days of coculture were restimulated with PMA (10 ng/mL; Sigma) and Ionomycin (1 μg/mL, Sigma) for 4 hours. After stimulation, cells were stained with anti-CD4-V450, anti-CD19-PECF-594, anti-CD25-PC7, anti-FoxP3-PE, anti-IFNγ-FITC, anti-IL2-APC and live/dead near-IR.

IHC

IHC was performed on whole sections of paraffin blocks used for diagnosis. After dewaxing and pressure-cooker antigen retrieval, mAbs directed against ICOS (clone sp98; Abcam), and FoxP3 (clone 236A/E7; eBioscience) were incubated in an automated immunostainer (Dako) using a standard avidin biotin peroxidase technique. Double staining experiments were performed using the K5361 Envision G/2 Doublestain System (Dako) according to the supplier's instructions. ICOSL IHC was performed on frozen tissue sections using the Envision-Flex K8000 kit (Dako) and the Autostainer Plus device (Dako). The anti-ICOSL mAb HIL-270 (provided by Pr. Richard Kroczek) was diluted at 10 μg/mL and incubated for 45 minutes at room temperature. As a control for the specificity of the HIL-270 ICOSL mAb, paraffin-embedded cell blocks were constructed using ICOSL-transfected COS-7 cells and parental COS-7 cells as negative control. Briefly, COS-7 cells transfected for 48 hours with βDNA4 vector containing full-length human ICOSL (produced in our laboratory) using X-tremGENE 9 DNA Transfection reagent (Roche) according to the manufacturer's instructions. After 48 hours, transfected and parental COS-7 cells were detached and prefixed in 4% formaldehyde solution and then suspended in agarose 1% LMT to form a cell-agarose gel, which was fixed in 4% formaldehyde solution for 24 hours. After fixation, the cell-agarose gel was fixed, dehydrated, paraffin impregnated on a Leica ASP300S processor (Leica Biosystems), and finally embedded in paraffin using a Leica Histo-Embedded processor. The method for ICOSL IHC on paraffin embedded cell blocks was similar to that described for ICOS and FoxP3. As expected, positive HIL-270 immunostaining was present only in ICOSL-transfected cells. For all antibodies, including HIL-270, a control was also done by omitting the primary mAb on serial sections of frozen tissues analyzed, which resulted in a lack of positive signal.

Quantitative RT-PCR

Total RNA was extracted from purified FL B cells and Burkitt lymphoma cell line Raji with TRIzol reagent (Life Technologies) according to the manufacturer's protocol and was then reverse transcribed into cDNA using M-MLV Reverse Transcriptase (Life Technologies). Quantitative PCR was performed using Taqman Universal PCR Master Mix and primers as follows: glyceraldehyde-3-phosphate dehydrogenase (GAPDH; Unigene reference: Hs.544577) and human ICOSL (Hs. 14155; Life Technologies). Gene expression was measured using the 7900HT Fast Real-Time PCR System (Life Technologies). For each sample, mRNA expression was normalized to GAPDH as a reference gene.

Statistical analysis

Quantitative variables were expressed as mean ± SEM. Statistical analysis was performed with GraphPad Prism 5 software (GraphPad software) using the Mann–Whitney and Wilcoxon nonparametric t test (*, P < 0.05; **, P < 0.01).

Intratumoral T cells with CD4+CD25highCD127low/– profile have characteristics of effector Tregs

Coexpression of CD25high and CD127low/neg was used to identify and isolate Tregs in FL tissue samples (Fig. 1A). CD25highCD127low/neg cells expressed high levels most Treg markers such as FoxP3 (86% ± 9.1%, mean ± SD), Helios (84% ± 7.6%; Fig. 1B) and strongly expressed other Tregs markers such as GITR (72 ± 13.4), surface CTLA-4 (32.8% ± 11%), and CD39 (80.2% ± 16.1%; Fig. 1B). Most CD25highCD127low/neg Tregs expressed CD27 (99.2% ± 1.1%) and were CD45RA-negative (3.2% ± 1.6%; Fig. 1B), suggesting a memory T-cell phenotype. Tconv were identified as CD25neg/low among viable CD4+ T cells (Fig. 1A). When compared to Tconv, purified CD25highCD127low/neg Tregs had a low proliferation rate in response to CD3/CD28 stimulation (87% and 10% for Tconv and Tregs, respectively; Fig. 1C). Tconv proliferation and IFNγ production was strongly inhibited when CD25highCD127low/neg cells were added in culture (Fig. 1D). Taken together, these results demonstrate that CD25highCD127low/neg T cells have the phenotypic and functional profile of effector Tregs.

Figure 1.

Phenotypic and functional characterization of CD25highCD127low/neg T cells. MCs in FL samples were used to characterize CD25highCD127low/neg T cells. A, gating strategy to identify Tregs and Tconv cells. CD4+ and CD8+ T cells were identified from viable CD3+ T cells (live/deadneg CD3+), then Tregs were defined as CD25highCD127low/neg while Tconv were CD25neg/low among CD4+ T cells. B, representative histograms for expression of Foxp3, Helios, and surface GITR, CTLA-4, CD39, CD45RA, and CD27 on CD8+ T cells, Tconv, and Tregs (n = 8–10). C and D, intratumoral Tregs and Tconv were sorted for functional assay. C, proliferation of Tconv and Tregs with stimulation by CD3/CD28 dynabeads (white) or without stimulation (gray) for 5 days (n = 2). D, CellTrace violet-labeled Tconv were cocultured with different ratios of autologous Tregs in the presence of CD3/CD28 dynabeads for 4 days (n = 3). Proliferation (top) and IFNy secretion measured in supernatant by ELISA (bottom) of Tconv. Proliferation was evaluated by division index (DI) using the proliferation tool of FlowJo software.

Figure 1.

Phenotypic and functional characterization of CD25highCD127low/neg T cells. MCs in FL samples were used to characterize CD25highCD127low/neg T cells. A, gating strategy to identify Tregs and Tconv cells. CD4+ and CD8+ T cells were identified from viable CD3+ T cells (live/deadneg CD3+), then Tregs were defined as CD25highCD127low/neg while Tconv were CD25neg/low among CD4+ T cells. B, representative histograms for expression of Foxp3, Helios, and surface GITR, CTLA-4, CD39, CD45RA, and CD27 on CD8+ T cells, Tconv, and Tregs (n = 8–10). C and D, intratumoral Tregs and Tconv were sorted for functional assay. C, proliferation of Tconv and Tregs with stimulation by CD3/CD28 dynabeads (white) or without stimulation (gray) for 5 days (n = 2). D, CellTrace violet-labeled Tconv were cocultured with different ratios of autologous Tregs in the presence of CD3/CD28 dynabeads for 4 days (n = 3). Proliferation (top) and IFNy secretion measured in supernatant by ELISA (bottom) of Tconv. Proliferation was evaluated by division index (DI) using the proliferation tool of FlowJo software.

Close modal

Intratumoral accumulation of ICOS+ Tregs is a specific feature of FL

The mean percentage of Tregs within CD4+ T cells was significantly increased in FL tissues as compared to rL (7.5-fold, P = 0.0005), cHL (2.6-fold, P = 0.003) and DLBCL (2.3-fold, P = 0.01; Fig. 2A). Both Tregs/Tconv and Tregs/TCD8+ ratios were significantly higher in FL than in other samples (Fig. 2B). ICOS was mostly expressed in CD4+ T cells, especially in Tregs (65.3% ± 14.4%), as compared with other tumor infiltrating lymphocytes (TIL) like CD8+ T cells, γδ T cells, and natural killer cells (Fig. 2C). Accordingly, the percentage of ICOS+ Tregs among CD4+ T cells was significantly increased in FL tumors (Fig. 2D). Like the majority of Tregs in FL, ICOS+ Tregs expressed CXCR5 (mean ± SD: 83% ± 7.5%; Fig. 2E). When compared to ICOSneg Tregs, ICOS+ Tregs in FL tissues displayed significant upregulation of surface CTLA-4 (40.1% ± 10.1%), GITR (83.6% ± 10.2%), CD39 (85% ± 15.9%), PD1 (90.4% ± 7.9%) as well as higher Ki67 expression (19.1% ± 7.2%; Fig. 2E). These data indicate a specific accumulation of activated and expanding ICOS+ Tregs in the FL microenvironment.

Figure 2.

Frequency and phenotype of Tregs in FL. MCs extracted from lymphoma and rL samples were stained with antibodies and analyzed by flow cytometry. A, percentage of Tregs among CD4+ T cells in FL compared with controls. B, ratio of Tregs to Tconv (left) and to TCD8 (right) in FL compared with controls. C, representative histogram for ICOS expression (white) and isotype control (gray) on different lymphocytes, including Tregs, Tconv, TCD8+, Tγδ, and NK. Mean of percentage of 14 FL samples was indicated. D, percentage of Tregs expressing ICOS among CD4+ T cells in FL compared with controls. E, representative dot plots for expression of various molecules on ICOS+ Tregs and ICOSneg Tregs from FL samples (n = 9–10). Mann–Whitney nonparametric t tests were performed (*, P ≤ 0.05; **, P < 0.01; ***, P < 0.001).

Figure 2.

Frequency and phenotype of Tregs in FL. MCs extracted from lymphoma and rL samples were stained with antibodies and analyzed by flow cytometry. A, percentage of Tregs among CD4+ T cells in FL compared with controls. B, ratio of Tregs to Tconv (left) and to TCD8 (right) in FL compared with controls. C, representative histogram for ICOS expression (white) and isotype control (gray) on different lymphocytes, including Tregs, Tconv, TCD8+, Tγδ, and NK. Mean of percentage of 14 FL samples was indicated. D, percentage of Tregs expressing ICOS among CD4+ T cells in FL compared with controls. E, representative dot plots for expression of various molecules on ICOS+ Tregs and ICOSneg Tregs from FL samples (n = 9–10). Mann–Whitney nonparametric t tests were performed (*, P ≤ 0.05; **, P < 0.01; ***, P < 0.001).

Close modal

ICOSL is occasionally expressed in neoplastic B cells from DLBCL, but is downregulated in FL B cells

To identify a putative cell population responsible for ICOS+ Tregs accumulation in fresh FL tissues, we assessed the expression of ICOSL and CD86 on FL B cells, reactive T cells, myeloid dendritic cells (mDC) and plasmacytoid DC (pDC). FL B cells were identified as CD3neg/CD16neg/CD56neg/CD20+, because most of these cells in FL samples were shown to be neoplastic B cells due to coexpression of CD10 and Bcl-2 (Supplementary Fig. S1A). pDCs were identified as lineageneg/HLA-DR+/CD33neg/+/CD123+ and mDCs as lineageneg/HLA-DR+/CD33++/CD123neg (Fig. 3A). CD86 was strongly expressed on mDCs (90.6% ± 10.1%), pDCs (21.8% ± 15.9%), and FL B cells (52.6% ± 27.3%), but ICOSL was weak on all cell populations (Fig. 3B) including reactive T cells (Supplementary Fig. S1A). Surprisingly, only ICOSL expression was rapidly upregulated on FL B cells after a few hours of culture in vitro of purified FL B cells (Fig. 3C). ICOSL expression was weak on FL B cells after purification (1.7% ± 1.6%), but it was induced by short in vitro culture (15.1% ± 18.0% after 2 hours of culture) and increased during culture (45.1% ± 34.6% at 48 hours). Furthermore, ICOSL upregulation on FL B cells was also observed in vitro culture of total MCs (Supplementary Fig. S1B). We then examined the expression of ICOSL transcripts on purified FL B cells by quantitative RT-PCR. The Burkitt lymphoma cell line Raji was used as a positive control due to its high ICOSL expression detected by flow cytometry (Supplementary Fig. S1C). ICOSL transcripts were detectable on purified FL B cells and Raji cell line but its mRNA level was 2-fold higher on FL B cells than on positive control Raji (Fig. 3D). In contrast to FL B cells, ICOSL was expressed on lymphoma B cells from some DLBCL samples, whereas ICOS was absent on TILs (Supplementary Fig. S1D).

Figure 3.

ICOSL and CD86 expression on immune cells in FL. MCs extracted from lymphoma samples were stained with antibodies and analyzed by flow cytometry. A, gating strategy used to identify lymphoma B cells and DCs: lymphoma B cells were identified as CD3negCD16negCD56negCD20+. Within DCs defined as lineage (CD3/CD16/CD56/CD20/CD14)neg HLA DR+, pDCs were CD33+/negCD123+, whereas mDCs were CD33++CD123neg. B, representative histogram for ICOSL and CD86 expression (white) as compared with isotype control (gray) on mDCs, pDCs, and lymphoma B cells from 14 samples of FL. Mean of percentage was indicated. C, purified FL B cells were cultured in vitro and percentage of expression of ICOSL and CD86 were analyzed at different times (n = 5). D, mRNA from purified FL B cells (n = 5) and Raji cell line were extracted and expression of ICOSL transcripts was detected by quantitative RT-PCR. The amount of ICOSL transcripts on purified FL B cells was compared with value of the positive control Raji used as a calibrator.

Figure 3.

ICOSL and CD86 expression on immune cells in FL. MCs extracted from lymphoma samples were stained with antibodies and analyzed by flow cytometry. A, gating strategy used to identify lymphoma B cells and DCs: lymphoma B cells were identified as CD3negCD16negCD56negCD20+. Within DCs defined as lineage (CD3/CD16/CD56/CD20/CD14)neg HLA DR+, pDCs were CD33+/negCD123+, whereas mDCs were CD33++CD123neg. B, representative histogram for ICOSL and CD86 expression (white) as compared with isotype control (gray) on mDCs, pDCs, and lymphoma B cells from 14 samples of FL. Mean of percentage was indicated. C, purified FL B cells were cultured in vitro and percentage of expression of ICOSL and CD86 were analyzed at different times (n = 5). D, mRNA from purified FL B cells (n = 5) and Raji cell line were extracted and expression of ICOSL transcripts was detected by quantitative RT-PCR. The amount of ICOSL transcripts on purified FL B cells was compared with value of the positive control Raji used as a calibrator.

Close modal

ICOSL IHC was performed on 16 frozen samples, including human tonsils (n = 2), reactive lymph node (n = 1), low-grade follicular lymphoma (n = 10), and DLBCL (n = 3). ICOSL immunostaining was absent or faint in neoplastic follicles in the 10 FL cases analyzed (Fig. 4). Internal positive controls were mainly located outside the B-cell follicles and often displayed morphologic features of macrophages with large cytoplasm and oval nucleus (Fig. 4). Capillary venules also displayed positive signals of variable intensity (Fig. 4) in accordance with a previous report (35). A few ICOSL+ cells are also present in the interfollicular areas of benign tonsils and lymphadenitis, whereas normal B cells in reactive follicles appeared mainly negative. In contrast, DLBCL cells displayed heterogeneous ICOSL positivity in two of three analyzed cases (data not shown).

Figure 4.

IHC features of ICOS, Foxp3, and ICOSL expression. Serial sections from a representative formalin-fixed paraffin-embedded FL case show the predominant intrafollicular localization of ICOS+ cells (A and B) and the presence of FoxP3+ cells within neoplastic follicles (FOL) of the same tumor area (C). C, FoxP3-positive cells were also observed within interfollicular areas (star). Flow cytometry indicated that this FL tissue sample contained 35% of ICOS+ CD4+ T cells and 15% of CD25highCD127low/neg Tregs among CD4+ T cells. D, double staining experiments highlight FoxP3/ICOS coexpression in TILs localized within follicles (black arrows) and intermingled with ICOS+ TILs (red arrow) and FoxP3+ TILs (brown arrow). E, ICOSL immunodetection on a frozen FL sample shows no detectable positivity in FL cells (FOL), whereas positive signals are seen outside neoplastic follicles, especially on venules (arrow). The negative control of the same FL case is shown in F. G and H, high-power view shows that some ICOSL-positive cells display morphologic features of reactive macrophages with large cytoplasm and oval or reniform nucleus (arrows).

Figure 4.

IHC features of ICOS, Foxp3, and ICOSL expression. Serial sections from a representative formalin-fixed paraffin-embedded FL case show the predominant intrafollicular localization of ICOS+ cells (A and B) and the presence of FoxP3+ cells within neoplastic follicles (FOL) of the same tumor area (C). C, FoxP3-positive cells were also observed within interfollicular areas (star). Flow cytometry indicated that this FL tissue sample contained 35% of ICOS+ CD4+ T cells and 15% of CD25highCD127low/neg Tregs among CD4+ T cells. D, double staining experiments highlight FoxP3/ICOS coexpression in TILs localized within follicles (black arrows) and intermingled with ICOS+ TILs (red arrow) and FoxP3+ TILs (brown arrow). E, ICOSL immunodetection on a frozen FL sample shows no detectable positivity in FL cells (FOL), whereas positive signals are seen outside neoplastic follicles, especially on venules (arrow). The negative control of the same FL case is shown in F. G and H, high-power view shows that some ICOSL-positive cells display morphologic features of reactive macrophages with large cytoplasm and oval or reniform nucleus (arrows).

Close modal

Intrafollicular localization of FoxP3+ and ICOS+ cells in FL cases

FoxP3 and ICOS IHC were performed on 6 FL and DLBCL cases that were also analyzed by flow cytometry. IHC results confirmed flow cytometry data, because numerous ICOS+ were present in FL cases, with a predominant intrafollicular localization (Fig. 4). Variable amounts of FoxP3+ cells were also observed within neoplastic follicles of FL cases (Fig. 4). In addition, ICOS+ /FoxP3+ TILs could be detected in FL cases using double staining experiments and were located within the follicles (Fig. 4), which implies vicinity and possible interaction with neoplastic B cells.

ICOS/ICOSL interaction induced downregulation of ICOSL on FL B cells

ICOSL downregulation through ICOS binding was previously described in normal murine B cells (36) and in human pDC (27). We evaluated the impact of FL T cells on ICOSL expression by cocultures of conditioned FL B cells with either autologous intratumoral ICOS+ or ICOSneg CD4+ T cells at different ratios during 24 hours. An important decrease of ICOSL expression on FL B cells (about 67% at 1:1 ratio after 24 hours of coculture) was observed in the presence of ICOS+ CD4+ T cells (Fig. 5A). A high level of ICOSL expression could be restored on FL B cells using an antagonist anti-ICOS antibody (314.8; Fig. 5B). Similar results were also observed in both in vitro culture of total MCs and in vivo using normal murine model (Supplementary Fig. S2). In contrast, no alteration of CD86 expression on FL B cells was observed (Fig. 5B). These data suggest that ICOS/ICOSL interaction is involved in downregulating ICOSL expression on FL B cells.

Figure 5.

Kinetic for expression of ICOSL and CD86 on FL B cells in vitro culture. Purified FL B cells were cocultured in vitro with intratumoral T cells, and their expression of ICOSL and CD86 was evaluated by flow cytometry. A, ICOSL expression on FL B cells at 24 hours in the presence of different ratios of autologous intratumoral ICOS+ CD4+ T cells or ICOSneg CD4+ T cells (n = 3). B, representative dot plots for ICOSL and CD86 expression on purified FL B cells at 24 hours in coculture with autologous intratumoral ICOS+ CD4+ T cells (CD4+ T- vs. B-cell ratio 1:1) in the presence of anti-ICOS antagonist (clone 314.8) or isotype Ctrl IgG1. Median of MFI of three independent experiments was indicated.

Figure 5.

Kinetic for expression of ICOSL and CD86 on FL B cells in vitro culture. Purified FL B cells were cocultured in vitro with intratumoral T cells, and their expression of ICOSL and CD86 was evaluated by flow cytometry. A, ICOSL expression on FL B cells at 24 hours in the presence of different ratios of autologous intratumoral ICOS+ CD4+ T cells or ICOSneg CD4+ T cells (n = 3). B, representative dot plots for ICOSL and CD86 expression on purified FL B cells at 24 hours in coculture with autologous intratumoral ICOS+ CD4+ T cells (CD4+ T- vs. B-cell ratio 1:1) in the presence of anti-ICOS antagonist (clone 314.8) or isotype Ctrl IgG1. Median of MFI of three independent experiments was indicated.

Close modal

ICOS/ICOSL interaction induced enrichment of CD25high FoxP3high Tregs in FL

We next explored the possible causes of Treg accumulation in FL tissues. We cocultured purified FL B cells conditioned to express ICOSL by in vitro culture, with intratumoral autologous or allogeneic CD4+ T cells in the presence of rIL2 for 5 to 6 days. Intratumoral CD4+ T cells were labeled with CellTrace violet prior coculture to evaluate the division rate. We first observed an enhancement of CD25 and FoxP3 expression on CD4+ T cells (data not shown). Consequently, the number of Tregs, defined by CD25 and FoxP3 coexpression, was significantly increased in the presence of FL B cells (median, 33.5% versus 20.0%; n = 6; P = 0.03; Fig. 6A, left). These Tregs comprised two distinct subsets of CD25+FoxP3+ and CD25highFoxP3high Tregs (Fig. 6A, right). Median percentages of CD25+FoxP3+ Tregs were 29.2% following coculture with FL B cells, versus 19.9% (P = 0.03) without FL B cells. The number of CD25highFoxP3high Tregs was virtually undetectable without FL B cells, but clearly increased during coculture. Their median percentage was 3.0% and 9.4% per total CD4+ T cells and per total Tregs, respectively (P = 0.03). Proliferation of Tregs in culture with FL B cells was significant, reaching 47.9% and 20.5% of CD25highFoxP3high Tregs and CD25+FoxP3+ Tregs, respectively (Fig. 6B, top). ICOS expression was markedly higher on CD25highFoxP3high Tregs than on CD25+FoxP3+ Tregs [mean fluorescence intensity (MFI): 1451 vs. 573, respectively; Fig. 6B, bottom]. After restimulation of cell suspension using PMA/ionomycin, intracellular cytokines were detected (Fig. 6C). FoxP3neg CD4+ T cells strongly secreted IL2 (median 23%) and IFNγ (median: 13.4%). In contrast, both putative Treg subsets showed weak IL2 and IFNγ secretion. Median percentages among CD25+FoxP3+ Tregs were 5.3% and 1.9% for IL2 and IFNγ, respectively. Median percentages among CD25highFoxP3high Tregs were 4.6% and 1.1% for IL2 and IFNγ, respectively. These data indicate that both cell populations were indeed Tregs rather than activated T cells. Thus, FL B cells expressing ICOSL could favor intratumoral enrichment of ICOS+ Tregs.

Figure 6.

Treg enrichment driven by FL B cells dependent on ICOS/ICOSL interaction. CellTrace violet-labeled intratumoral CD4+ T cells were cultured alone or with FL B cells expressing ICOSL, and cell suspension was stained for CD25, FoxP3, ICOS after 5 to 6 days of culture. A, frequency among CD4+ T cells of total Tregs (n = 6; left) and each Treg subset from a representative case (right). B, representative histogram for proliferation (top) and ICOS expression (bottom) of CD25+FoxP3+ Tregs and CD25highFoxP3high Tregs in coculture with FL B cells. C, production of intracellular IL2 and IFNγ on FoxP3neg CD4+ T cells, CD25+FoxP3+ Tregs, and CD25highFoxP3highTregs in response to PMA/ionomycin stimulation after 5 to 6 days of coculture (n = 2–4). D, percentage of CD25+FoxP3+Tregs (left) and CD25highFoxP3highTregs (right) among CD4+ T cells in coculture with FL B cells in the presence of neutralizing anti-ICOS or anti-ICOSL antibodies or isotype Ctrl IgG1. Statistical analyses were done using Wilcoxon nonparametric t tests (*, P < 0.05).

Figure 6.

Treg enrichment driven by FL B cells dependent on ICOS/ICOSL interaction. CellTrace violet-labeled intratumoral CD4+ T cells were cultured alone or with FL B cells expressing ICOSL, and cell suspension was stained for CD25, FoxP3, ICOS after 5 to 6 days of culture. A, frequency among CD4+ T cells of total Tregs (n = 6; left) and each Treg subset from a representative case (right). B, representative histogram for proliferation (top) and ICOS expression (bottom) of CD25+FoxP3+ Tregs and CD25highFoxP3high Tregs in coculture with FL B cells. C, production of intracellular IL2 and IFNγ on FoxP3neg CD4+ T cells, CD25+FoxP3+ Tregs, and CD25highFoxP3highTregs in response to PMA/ionomycin stimulation after 5 to 6 days of coculture (n = 2–4). D, percentage of CD25+FoxP3+Tregs (left) and CD25highFoxP3highTregs (right) among CD4+ T cells in coculture with FL B cells in the presence of neutralizing anti-ICOS or anti-ICOSL antibodies or isotype Ctrl IgG1. Statistical analyses were done using Wilcoxon nonparametric t tests (*, P < 0.05).

Close modal

When neutralizing anti-ICOS and anti-ICOSL mAbs were added in coculture, there were not significant change in the percentage of CD25+FoxP3+ Tregs among CD4+ T cells (Fig. 6D, left), whereas both mAbs induced a significant reduction in the percentage of CD25highFoxP3high Tregs (Fig. 6D, right). No impact was observed after the addition of an isotype control IgG1. These results indicate that generation of CD25highFoxP3high Tregs expressing ICOS is strongly dependent on ICOSL expression by FL B cells.

Intratumoral Tregs directly inhibit FL B cells for both T-cell–dependent and T-cell–independent responses

Our observation of Tregs overrepresentation in FL tumors prompted us to test their capacity to inhibit FL B cells. We selected IL6 as one of the main B cell–produced cytokine (37). We first tested whether Tregs could act on B cells activated in a T-cell–independent way. FL B cells were activated by CpG-B and anti-IgA/G/M prior to culture with Tregs at 1:1 ratio. We observed a significant attenuation of CD80 and CD86 expression on FL B cells. This was evidenced by the percentage for CD80 (approximately 30% of inhibition, P = 0.0078, n = 9) and by MFI for CD80 and CD86 (about 35% and 31% inhibition for CD80 and CD86, respectively (Fig. 7A). Similarly, Tregs abrogated IL6 secretion in the supernatant of FL B cells (median percentage of 26% inhibition, P = 0.0078, n = 8; Fig. 7B).

Figure 7.

Inhibition of FL B-cell responses by intratumoral Tregs. CellTrace violet-labeled FL B cells activated by anti-IgA/G/M and CpG-B were cocultured with Tregs for direct suppression assay (A and B) or Tconv with or without Tregs for indirect suppression assay (C and D) during 5 days. A and C, representative histogram for the percentage of CD80 (white) versus isotype Ctrl (gray; top) and MFI of CD80 and CD86 expression (bottom) on FL B cells were evaluated by flow cytometry (n = 9). B and D, IL6 secretion in supernatant of coculture measured by ELISA (n = 8). All data were normalized to “B-alone” condition that is considered as 100%. Wilcoxon nonparametric t tests were used: *, P < 0.05; **, P < 0.01.

Figure 7.

Inhibition of FL B-cell responses by intratumoral Tregs. CellTrace violet-labeled FL B cells activated by anti-IgA/G/M and CpG-B were cocultured with Tregs for direct suppression assay (A and B) or Tconv with or without Tregs for indirect suppression assay (C and D) during 5 days. A and C, representative histogram for the percentage of CD80 (white) versus isotype Ctrl (gray; top) and MFI of CD80 and CD86 expression (bottom) on FL B cells were evaluated by flow cytometry (n = 9). B and D, IL6 secretion in supernatant of coculture measured by ELISA (n = 8). All data were normalized to “B-alone” condition that is considered as 100%. Wilcoxon nonparametric t tests were used: *, P < 0.05; **, P < 0.01.

Close modal

Tconv in FL samples could also be suppressed by Tregs (Fig. 1D). About a quarter (25.2% ± 16.1%, n = 12) of these Tconv were TFH cells characterized by ICOS and CXCR5 coexpression (data not shown). We next searched for a putative suppression of Tconv-driven B-cell responses by Tregs. For this purpose, stimulated B cells were cocultured with Tconv, with or without Tregs at 1:1 ratio. Tconv induced a significant increase of both CD80 and CD86 expression on stimulated B cells, and of IL6 secretion (median increase of 67%, 121%, and 49% for CD80 MFI, CD86 MFI, and IL6, respectively). B-cell responses were inhibited after addition of Tregs (inhibition of 44%, 53%, and 39% for CD80 MFI, CD86 MFI, and IL6, respectively; Fig. 7C and D).

Of note, there was also a tendency for Tregs to suppress B-cell proliferation, although it was not statistically significant for all samples (Supplementary Fig. S3). These data suggest that intratumoral Tregs can directly or indirectly suppress FL B-cell responses upon activation.

Clarifying the impact of Tregs on FL pathogenesis is a critical issue for clinical implications. However, the lack of strictly a specific Treg marker may result in discrepancies between studies focusing on Tregs in human. FoxP3, which is considered as the most specific Treg marker, was reported to be transiently expressed in activated CD4+ and CD8+ T cells (38, 39), which, in contrast to Tregs, produce effector cytokines like IL2 and IFNγ (23, 39, 40). Using the CD25high/CD127low/neg combination, we could isolate T cells displaying Treg phenotypic markers and strong suppressive activity. Most CD25highCD127low/neg T cells were FoxP3high and hardly secreted IL2 and IFNγ, further supporting their Tregs' nature. Thus, our results corroborate previous investigations (41, 42) suggesting that the CD25highCD127low/neg combination is useful for Tregs' identification.

In this study, we confirm using multiparameter flow cytometry that not only Tregs, but also ICOS+ Tregs, are overrepresented in FL tumors. We observed that ICOS+ Tregs expressed high levels of CXCR5, PD1, GITR, and CTLA-4, and thus fit the profile of TFR that were described in mice and humans (32, 33) and in FL tissues as well (31). Of note, we demonstrate herein that FL B cells are able to promote generation of these intratumoral ICOS+/CD25highFoxP3high Tregs via ICOS/ICOSL interaction, which induced ICOSL downregulation on FL B cells. Furthermore, we provide evidence of a strong suppressive effect of intratumoral Tregs on malignant B cells.

Several studies have shown a favorable outcome for FL patients with high amounts of intratumoral Tregs, although this remains controversial (18, 43). These observations support the hypothesis that Tregs could control malignant FL cells, but no demonstration of such functional interaction was reported so far. In nontumoral human tonsils, B-cell responses were shown to be inhibited by Tregs (44, 45). We report herein that Tregs are able to suppress the activation process and cytokine secretion of FL B cells, either directly or indirectly.

Tregs are known to use various mechanisms to suppress immune responses, including cell-to-cell contact (45). Additionally, Tregs are known to kill B cells through Fas–FasL interaction or through the release of cytolytic molecules (46, 47). So, further study is needed to identify mechanisms involved in direct FL B cell inhibition. The indirect suppressive effect of Tregs upon FL B cells is in accordance with a report showing that TFH cells support FL B-cell activity via IL4 and CD40L (31). Our data suggest that Tregs are able to inhibit IL4/CD40L-producing T cells, probably including TFH, which could hamper FL growth.

Nonetheless, the favorable prognostic value of high numbers of Tregs in FL tumors remains debated (11–13). In mouse models, depletion of Tregs induced decreased lymphoma burden (48). Unfavorable outcome was reported for FL patients with high levels of peripheral blood Tregs (49). A possible explanation that may reconcile the latter studies with our results may reside in the polarization of Tregs into distinct functional subsets. Tregs expressing Tbet, IRF4, STAT3, and Bcl6 appear to control Th1-, Th2-, Th17-, and TFH-mediated immune responses, respectively (32, 50–52). Therefore, it is possible that intratumoral ICOS+ Tregs include multiple subsets, with opposite effects on tumor growth. The balance between these antagonist Tregs subsets may vary according to complex cues from the microenvironment. In this extent, it is noteworthy that we also observed a significant Tregs' increase in tissue samples from cHL, a lymphoma subtype that is known to undergo, like FL, crucial influences from the microenvironment, including a suspected favorable effect of Treg accumulation. This raises the question as to whether similar signaling networks could regulate Tregs in FL and cHL. The dependence of Tregs' status on the microenvironment of particular lymphoma subtypes is also supported by the previous observation that impoverishment of the Treg pool in angio-immunoblastic T-cell lymphoma (AITL) resulted from a proinflammatory microenvironment involving Th17 cells (53, 54).

We observed an increase in the amount of Tregs, including ICOS+/CD25highFoxp3high Tregs, among CD4+ T cells in coculture with FL B cells. This result is consistent with recent reports of Treg differentiation induced by malignant B cells (22–24). Tregs' increase could be due to either Tconv conversion or Tregs expansion. Our observation of significant Ki67 expression in ICOS+ Tregs favors the view of Tregs' expansion. On the other hand, it is also possible that lymphoma cells could convert Tconv to Tregs, as previously reported (22, 23). Tregs enrichment in FL tumors probably result from the combination of diverse mechanisms, including conversion and expansion, as well as migration of Tregs induced by CCL17- and CCL22-secreting FL B cells (55).

Survival, expansion, and suppressive functions of human Tregs are known to be strongly dependent on ICOS costimulation in physiologic settings (25). Accordingly, we show that coculture of ICOSL+ FL B cells with T cells induced generation of ICOS+/CD25highFoxP3high Tregs. Using neutralizing antibodies, we show that ICOS/ICOSL interaction is critical for this induction. ICOSL expression on FL B cells was downregulated following interaction with ICOS+ T cells, which may explain why ICOSL was undetectable in vivo. It remains, however, difficult to decipher what could be the primum movens of the cascade of ICOS/ICOSL interactions in vivo. Whatever the triggering event could be in FL, such a mechanism is globally reminiscent to that described in other human cancers, in which ICOS+ Tregs' enrichment is triggered by ICOSL produced by pDCs or tumor cells (26–28). Thus, our findings provide a basis to target the ICOS/ICOSL pathway for the development of future immunotherapies in FL patients.

No potential conflicts of interest were disclosed.

Conception and design: K.-S. Le, S. Pastor, C. Caux, C. Ménétrier-Caux, L. Xerri, D. Olive

Development of methodology: K.-S. Le, M.-L. Thibult, S. Just-Landi, S. Pastor, F. Gondois-Rey, C. Caux, D. Olive

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.-L. Thibult, F. Broussais, R. Bouabdallah, R. Colisson, D. Leroux, L. Xerri, D. Olive

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K.-S. Le, F. Gondois-Rey, S. Granjeaud, R. Colisson, C. Ménétrier-Caux, L. Xerri, D. Olive

Writing, review, and/or revision of the manuscript: K.-S. Le, R. Bouabdallah, C. Caux, C. Ménétrier-Caux, L. Xerri, D. Olive

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.-S. Le, M.-L. Thibult, S. Just-Landi, R. Bouabdallah, R. Colisson, D. Olive

Study supervision: L. Xerri, D. Olive

Other (performed experiments): K.-S. Le

The authors thank Pr. Thierry Fest (CHU de Rennes, Rennes, France), Pr. Richard Kroczek (Molecular Immunology, Robert Koch-Institute, Berlin, Germany), Cytometry platform, Experimental Histo-Pathology (ICEP) platform, IBiSA Cancer Immunomonitoring platform, and Department of Biopathology for their help.

This work was supported by grants from Institute National du cancer (INCa), Agence Nationale de la Recherche (ANR-11-EMMA-0045), Canceropole PACA, the pharmaceutical company GlaxoSmithKline (GSK) and by fellowships (K.-S. Le) from Ministère de l'Enseignement Supérieur et de la Recherche and the Ligue Nationale contre le Cancer (LIGUE). D. Olive team was labeled “Equipe FRM DEQ 201 40329534.” D. Olive is a senior scholar of the Institut Universitaire de France.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Freedman
A
. 
Follicular lymphoma: 2014 update on diagnosis and management
.
Am J Hematol
2014
;
89
:
429
36
.
2.
Dave
SS
,
Wright
G
,
Tan
B
,
Rosenwald
A
,
Gascoyne
RD
,
Chan
WC
, et al
Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells
.
N Engl J Med
2004
;
351
:
2159
69
.
3.
Farinha
P
,
Masoudi
H
,
Skinnider
BF
,
Shumansky
K
,
Spinelli
JJ
,
Gill
K
, et al
Analysis of multiple biomarkers shows that lymphoma-associated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL)
.
Blood
2005
;
106
:
2169
74
.
4.
Lee
AM
,
Clear
AJ
,
Calaminici
M
,
Davies
AJ
,
Jordan
S
,
MacDougall
F
, et al
Number of CD4+ cells and location of forkhead box protein P3-positive cells in diagnostic follicular lymphoma tissue microarrays correlates with outcome
.
J Clin Oncol
2006
;
24
:
5052
9
.
5.
Alvaro
T
,
Lejeune
M
,
Salvado
MT
,
Lopez
C
,
Jaen
J
,
Bosch
R
, et al
Immunohistochemical patterns of reactive microenvironment are associated with clinicobiologic behavior in follicular lymphoma patients
.
J Clin Oncol
2006
;
24
:
5350
7
.
6.
Mougiakakos
D
,
Choudhury
A
,
Lladser
A
,
Kiessling
R
,
Johansson
CC
. 
Regulatory T cells in cancer
.
Adv Cancer Res
2010
;
107
:
57
117
.
7.
Curiel
TJ
,
Coukos
G
,
Zou
L
,
Alvarez
X
,
Cheng
P
,
Mottram
P
, et al
Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival
.
Nat Med
2004
;
10
:
942
9
.
8.
Petersen
RP
,
Campa
MJ
,
Sperlazza
J
,
Conlon
D
,
Joshi
MB
,
Harpole
DH
 Jr
, et al
Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients
.
Cancer
2006
;
107
:
2866
72
.
9.
Gobert
M
,
Treilleux
I
,
Bendriss-Vermare
N
,
Bachelot
T
,
Goddard-Leon
S
,
Arfi
V
, et al
Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome
.
Cancer Res
2009
;
69
:
2000
9
.
10.
Kobayashi
N
,
Hiraoka
N
,
Yamagami
W
,
Ojima
H
,
Kanai
Y
,
Kosuge
T
, et al
FOXP3+ regulatory T cells affect the development and progression of hepatocarcinogenesis
.
Clin Cancer Res
2007
;
13
:
902
11
.
11.
Carreras
J
,
Lopez-Guillermo
A
,
Fox
BC
,
Colomo
L
,
Martinez
A
,
Roncador
G
, et al
High numbers of tumor-infiltrating FOXP3-positive regulatory T cells are associated with improved overall survival in follicular lymphoma
.
Blood
2006
;
108
:
2957
64
.
12.
Tzankov
A
,
Meier
C
,
Hirschmann
P
,
Went
P
,
Pileri
SA
,
Dirnhofer
S
. 
Correlation of high numbers of intratumoral FOXP3+ regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin's lymphoma
.
Haematologica
2008
;
93
:
193
200
.
13.
Wahlin
BE
,
Aggarwal
M
,
Montes-Moreno
S
,
Gonzalez
LF
,
Roncador
G
,
Sanchez-Verde
L
, et al
A unifying microenvironment model in follicular lymphoma: outcome is predicted by programmed death-1–positive, regulatory, cytotoxic, and helper T cells and macrophages
.
Clin Cancer Res
2010
;
16
:
637
50
.
14.
Hilchey
SP
,
De
A
,
Rimsza
LM
,
Bankert
RB
,
Bernstein
SH
. 
Follicular lymphoma intratumoral CD4+CD25+GITR+ regulatory T cells potently suppress CD3/CD28-costimulated autologous and allogeneic CD8+CD25 and CD4+CD25 T cells
.
J Immunol
2007
;
178
:
4051
61
.
15.
Yang
ZZ
,
Novak
AJ
,
Stenson
MJ
,
Witzig
TE
,
Ansell
SM
. 
Intratumoral CD4+CD25+ regulatory T-cell-mediated suppression of infiltrating CD4+ T cells in B-cell non-Hodgkin lymphoma
.
Blood
2006
;
107
:
3639
46
.
16.
Yang
ZZ
,
Novak
AJ
,
Ziesmer
SC
,
Witzig
TE
,
Ansell
SM
. 
Attenuation of CD8(+) T-cell function by CD4(+)CD25(+) regulatory T cells in B-cell non-Hodgkin's lymphoma
.
Cancer Res
2006
;
66
:
10145
52
.
17.
Chetaille
B
,
Bertucci
F
,
Finetti
P
,
Esterni
B
,
Stamatoullas
A
,
Picquenot
JM
, et al
Molecular profiling of classical Hodgkin lymphoma tissues uncovers variations in the tumor microenvironment and correlations with EBV infection and outcome
.
Blood
2009
;
113
:
2765
3775
.
18.
Farinha
P
,
Al-Tourah
A
,
Gill
K
,
Klasa
R
,
Connors
JM
,
Gascoyne
RD
. 
The architectural pattern of FOXP3-positive T cells in follicular lymphoma is an independent predictor of survival and histologic transformation
.
Blood
2010
;
115
:
289
95
.
19.
Koch
K
,
Hoster
E
,
Unterhalt
M
,
Ott
G
,
Rosenwald
A
,
Hansmann
ML
, et al
The composition of the microenvironment in follicular lymphoma is associated with the stage of the disease
.
Hum Pathol
2012
;
43
:
2274
81
.
20.
Smeltzer
JP
,
Jones
JM
,
Ziesmer
SC
,
Grote
DM
,
Xiu
B
,
Ristow
KM
, et al
Pattern of CD14+ follicular dendritic cells and PD1+ T cells independently predicts time to transformation in follicular lymphoma
.
Clin Cancer Res
2014
;
20
:
2862
72
.
21.
Sakaguchi
S
,
Miyara
M
,
Costantino
CM
,
Hafler
DA
. 
FOXP3+ regulatory T cells in the human immune system
.
Nat Rev Immunol
2010
;
10
:
490
500
.
22.
Yang
ZZ
,
Novak
AJ
,
Ziesmer
SC
,
Witzig
TE
,
Ansell
SM
. 
CD70+ non-Hodgkin lymphoma B cells induce Foxp3 expression and regulatory function in intratumoral CD4+CD25 T cells
.
Blood
2007
;
110
:
2537
44
.
23.
Ai
WZ
,
Hou
JZ
,
Zeiser
R
,
Czerwinski
D
,
Negrin
RS
,
Levy
R
. 
Follicular lymphoma B cells induce the conversion of conventional CD4+ T cells to T-regulatory cells
.
Int J Cancer
2009
;
124
:
239
44
.
24.
Yang
ZZ
,
Novak
AJ
,
Ziesmer
SC
,
Witzig
TE
,
Ansell
SM
. 
Malignant B cells skew the balance of regulatory T cells and TH17 cells in B-cell non-Hodgkin's lymphoma
.
Cancer Res
2009
;
69
:
5522
30
.
25.
Ito
T
,
Hanabuchi
S
,
Wang
YH
,
Park
WR
,
Arima
K
,
Bover
L
, et al
Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery
.
Immunity
2008
;
28
:
870
80
.
26.
Martin-Orozco
N
,
Li
Y
,
Wang
Y
,
Liu
S
,
Hwu
P
,
Liu
YJ
, et al
Melanoma cells express ICOS ligand to promote the activation and expansion of T-regulatory cells
.
Cancer Res
2010
;
70
:
9581
90
.
27.
Faget
J
,
Bendriss-Vermare
N
,
Gobert
M
,
Durand
I
,
Olive
D
,
Biota
C
, et al
ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells
.
Cancer Res
2012
;
72
:
6130
41
.
28.
Conrad
C
,
Gregorio
J
,
Wang
YH
,
Ito
T
,
Meller
S
,
Hanabuchi
S
, et al
Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells
.
Cancer Res
2012
;
72
:
5240
9
.
29.
Carreno
BM
,
Collins
M
. 
The B7 family of ligands and its receptors: new pathways for costimulation and inhibition of immune responses
.
Annu Rev Immunol
2002
;
20
:
29
53
.
30.
Strauss
L
,
Bergmann
C
,
Szczepanski
MJ
,
Lang
S
,
Kirkwood
JM
,
Whiteside
TL
. 
Expression of ICOS on human melanoma-infiltrating CD4+CD25highFoxp3+ T regulatory cells: implications and impact on tumor-mediated immune suppression
.
J Immunol
2008
;
180
:
2967
80
.
31.
Ame-Thomas
P
,
Le Priol
J
,
Yssel
H
,
Caron
G
,
Pangault
C
,
Jean
R
, et al
Characterization of intratumoral follicular helper T cells in follicular lymphoma: role in the survival of malignant B cells
.
Leukemia
2012
;
26
:
1053
63
.
32.
Chung
Y
,
Tanaka
S
,
Chu
F
,
Nurieva
RI
,
Martinez
GJ
,
Rawal
S
, et al
Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions
.
Nat Med
2011
;
17
:
983
8
.
33.
Linterman
MA
,
Pierson
W
,
Lee
SK
,
Kallies
A
,
Kawamoto
S
,
Rayner
TF
, et al
Foxp3+ follicular regulatory T cells control the germinal center response
.
Nat Med
2011
;
17
:
975
82
.
34.
Maby-El Hajjami
H
,
Ame-Thomas
P
,
Pangault
C
,
Tribut
O
,
DeVos
J
,
Jean
R
, et al
Functional alteration of the lymphoma stromal cell niche by the cytokine context: role of indoleamine-2,3 dioxygenase
.
Cancer Res
2009
;
69
:
3228
37
.
35.
Khayyamian
S
,
Hutloff
A
,
Buchner
K
,
Grafe
M
,
Henn
V
,
Kroczek
RA
, et al
ICOS-ligand, expressed on human endothelial cells, costimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells
.
Proc Natl Acad Sci U S A
2002
;
99
:
6198
203
.
36.
Watanabe
M
,
Takagi
Y
,
Kotani
M
,
Hara
Y
,
Inamine
A
,
Hayashi
K
, et al
Down-regulation of ICOS ligand by interaction with ICOS functions as a regulatory mechanism for immune responses
.
J Immunol
2008
;
180
:
5222
34
.
37.
Bao
Y
,
Cao
X
. 
The immune potential and immunopathology of cytokine-producing
B cell subsets: a comprehensive review
.
J Autoimmun
2014
;
55
:
10
23
.
38.
Walker
MR
,
Kasprowicz
DJ
,
Gersuk
VH
,
Benard
A
,
Van Landeghen
M
,
Buckner
JH
, et al
Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25 T cells
.
J Clin Invest
2003
;
112
:
1437
43
.
39.
Gavin
MA
,
Torgerson
TR
,
Houston
E
,
DeRoos
P
,
Ho
WY
,
Stray-Pedersen
A
, et al
Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development
.
Proc Natl Acad Sci U S A
2006
;
103
:
6659
64
.
40.
Kryczek
I
,
Liu
R
,
Wang
G
,
Wu
K
,
Shu
X
,
Szeliga
W
, et al
FOXP3 defines regulatory T cells in human tumor and autoimmune disease
.
Cancer Res
2009
;
69
:
3995
4000
.
41.
Shen
LS
,
Wang
J
,
Shen
DF
,
Yuan
XL
,
Dong
P
,
Li
MX
, et al
CD4(+)CD25(+)CD127(low/−) regulatory T cells express Foxp3 and suppress effector T cell proliferation and contribute to gastric cancers progression
.
Clin Immunol
2009
;
131
:
109
18
.
42.
Seddiki
N
,
Santner-Nanan
B
,
Martinson
J
,
Zaunders
J
,
Sasson
S
,
Landay
A
, et al
Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells
.
J Exp Med
2006
;
203
:
1693
700
.
43.
Sweetenham
JW
,
Goldman
B
,
LeBlanc
ML
,
Cook
JR
,
Tubbs
RR
,
Press
OW
, et al
Prognostic value of regulatory T cells, lymphoma-associated macrophages, and MUM-1 expression in follicular lymphoma treated before and after the introduction of monoclonal antibody therapy: a Southwest Oncology Group Study
.
Ann Oncol
2010
;
21
:
1196
202
.
44.
Lim
HW
,
Hillsamer
P
,
Banham
AH
,
Kim
CH
. 
Cutting edge: direct suppression of B cells by CD4+ CD25+ regulatory T cells
.
J Immunol
2005
;
175
:
4180
3
.
45.
Lim
HW
,
Hillsamer
P
,
Kim
CH
. 
Regulatory T cells can migrate to follicles upon T cell activation and suppress GC-Th cells and GC-Th cell-driven B cell responses
.
J Clin Invest
2004
;
114
:
1640
9
.
46.
Janssens
W
,
Carlier
V
,
Wu
B
,
VanderElst
L
,
Jacquemin
MG
,
Saint-Remy
JM
. 
CD4+CD25+ T cells lyse antigen-presenting B cells by Fas-Fas ligand interaction in an epitope-specific manner
.
J Immunol
2003
;
171
:
4604
12
.
47.
Lindqvist
CA
,
Christiansson
LH
,
Thorn
I
,
Mangsbo
S
,
Paul-Wetterberg
G
,
Sundstrom
C
, et al
Both CD4+ FoxP3+ and CD4+ FoxP3− T cells from patients with B-cell malignancy express cytolytic markers and kill autologous leukaemic B cells in vitro
.
Immunology
2011
;
133
:
296
306
.
48.
Dou
AX
,
Feng
LL
,
Liu
XQ
,
Wang
X
. 
Cyclic adenosine monophosphate involvement in low-dose cyclophosphamide-reversed immune evasion in a mouse lymphoma model
.
Cell Mol Immunol
2012
;
9
:
482
8
.
49.
Voo
KS
,
Foglietta
M
,
Percivalle
E
,
Chu
F
,
Nattamai
D
,
Harline
M
, et al
Selective targeting of Toll-like receptors and OX40 inhibit regulatory T-cell function in follicular lymphoma
.
Int J Cancer
2014
;
135
:
2834
46
.
50.
Koch
MA
,
Tucker-Heard
G
,
Perdue
NR
,
Killebrew
JR
,
Urdahl
KB
,
Campbell
DJ
. 
The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation
.
Nat Immunol
2009
;
10
:
595
602
.
51.
Zheng
Y
,
Chaudhry
A
,
Kas
A
,
deRoos
P
,
Kim
JM
,
Chu
TT
, et al
Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses
.
Nature
2009
;
458
:
351
6
.
52.
Chaudhry
A
,
Rudra
D
,
Treuting
P
,
Samstein
RM
,
Liang
Y
,
Kas
A
, et al
CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner
.
Science
2009
;
326
:
986
91
.
53.
Tripodo
C
,
Gri
G
,
Piccaluga
PP
,
Frossi
B
,
Guarnotta
C
,
Piconese
S
, et al
Mast cells and Th17 cells contribute to the lymphoma-associated pro-inflammatory microenvironment of angioimmunoblastic T-cell lymphoma
.
Am J Pathol
2010
;
177
:
792
802
.
54.
Gaulard
P
,
de Leval
L
. 
The microenvironment in T-cell lymphomas: emerging themes
.
Semin Cancer Biol
2014
;
24
:
49
60
.
55.
Rawal
S
,
Chu
F
,
Zhang
M
,
Park
HJ
,
Nattamai
D
,
Kannan
S
, et al
Cross talk between follicular Th cells and tumor cells in human follicular lymphoma promotes immune evasion in the tumor microenvironment
.
J Immunol
2013
;
190
:
6681
93
.

Supplementary data