Several reports have shown that prophylactic depletion of regulatory T cells (Treg) using various monoclonal antibodies (mAb) in mice can stimulate potent antitumor immune responses and prevent tumor development. These same depletion methods do not significantly suppress tumor growth in a therapeutic setting. Although different strategies to deplete FoxP3+ Treg have been used, no study has systematically compared these qualitatively for the effector mechanisms they each liberate. Herein, using prophylactic depletion of FoxP3+ Tregs with either anti-CD4, anti-CD25, or anti-FR4 mAbs, we have compared the cellular and effector requirements for elimination of the renal carcinoma RENCA and prevention of methylcholanthrene-induced fibrosarcoma. Collectively from these two models, it was clear that CD8+ T cells and natural killer cells played an important role downstream of Treg depletion. However, whereas all three mAbs quantitatively depleted FoxP3+ T cells to a similar extent, subtle differences in the downstream mechanisms of tumor control existed for all three approaches. In general, neutralization of any lymphocyte subset or effector mechanism was insufficient to alter tumor suppression initiated by Treg depletion, and in some settings, the neutralization of multiple effector mechanisms failed to prevent tumor rejection. These studies reveal that Tregs control multiple redundant elements of the immune effector response capable of inhibiting tumor initiation and underscore the importance of effectively targeting these cells in any cancer immunotherapy. Cancer Res; 70(7); 2665–74

A number of studies have now documented tumor rejection following the prophylactic depletion of regulatory T cells (Treg; refs. 14). In some models, Treg depletion alone is sufficient for complete tumor eradication, whereas in others, Treg depletion acts as an adjuvant, enhancing vaccination and cytokine treatment. How these antitumor immune responses released by Treg depletion are coordinated is less clear. Tumor rejection triggered by anti-CD25 has been reported to involve CD4+ and CD8+ T cells, depending on the tumor model (1, 2), whereas in another study, peptide vaccination induced tumor-specific CD4+ T cells, making IFN-γ sufficient for CT26 tumor rejection (5). IFN-γ was also important for rejection of Meth A sarcomas induced by treatment with the agonistic α-GITR antibody DTA-1, which is thought to induce tumor immunity by abrogating Treg activity (6). The suppressive activity of Treg on CD8+ T cells has been examined in a model that used a HA-expressing derivative of CT26 called CT44 (7). Here Treg did not influence CD8+ T-cell expansion or cytokine secretion but rather inhibited their cytotoxic activity in vitro. Depletion of the entire CD4+ T-cell subset can also induce tumor rejection in some circumstances, for example, both Ag104Ld (4) and Ad5E1/ras (8) tumors were rejected by mice depleted of CD4+ T cells and in both cases tumor rejection was dependent on CD8+ T cells. More recently, evidence points to a critical role for Tregs in dampening natural killer cell effector functions in vitro and in vivo (9). The mechanisms seemed to involve membrane-bound transforming growth factor-β on Treg downregulating NKG2D expression on NK cells (10, 11) or Treg preventing dendritic cell exposure to interleukin-15Rα (IL-15Rα) and dendritic cell–mediated NK cell proliferation in vivo (12). Treg cells were recently reported to constitutively express high amounts of folate receptor 4 (FR4) and administration of anti-FR4 monoclonal antibody (mAb) specifically reduced Treg cells, provoking effective tumor immunity in tumor-bearing mice, but the mechanisms of rejection were not characterized (13).

Because therapeutic CD25+ Treg depletion has failed to consistently cause suppression of established tumors, even when used in combination with other immunotherapies (14), we have chosen to study the mechanisms of tumor suppression caused by the early absence of Treg at the time of tumor or carcinogen inoculation. We have focused on the effector cells, cytokines, and cytotoxic molecules required for successful tumor cell elimination following Treg depletion. The experimental renal carcinoma RENCA and spontaneous fibrosarcomas induced by the chemical carcinogen methylcholanthrene (MCA) are both well characterized for host adaptive (CD8+ CTL) and innate (NK/NKT) immune mechanisms that control their respective outgrowth in mice (1519). In addition, MCA-induced fibrosarcomas were strikingly infiltrated with FoxP3+ Tregs (20), and anti-CD25 mAb significantly reduced MCA-induced fibrosarcoma incidence in an IFN-γ–dependent fashion (21, 22).

Here we show that RENCA and MCA-induced fibrosarcomas are both suppressed following a variety of strategies (anti-CD4, anti-FR4, and anti-CD25) that partially deplete Tregs. Quantitatively these strategies depleted FoxP3+ T cells and triggered tumor rejection to a similar extent, but qualitatively each method revealed distinct tumor rejection mechanisms, with no single lymphocyte subset or effector molecule completely accounting for the tumor immunity in these mouse models of cancer.

Mice and tumor cell lines

Wild-type (WT) and gene-targeted BALB/c and C57BL/6 mice were either purchased from the Walter and Eliza Hall Institute Animal Facility or bred and maintained in house. BALB/c and C57BL/6 gene-targeted strains were either derived on a pure background (C57BL/6 perforin, pfp) or backcrossed to the appropriate strains for 10 to 12 generations (Jα18, IFN-γ, TRAIL, DR5, IL-4, IL-13, granzyme B, pfp × IFN-γ). Adult mice (age, 6–12 wk) were used in all experiments, unless otherwise indicated, and experiments were conducted under specific pathogen-free conditions according to the Peter MacCallum Cancer Centre Animal Experimental Ethics Committee guidelines. The BALB/c-derived renal adenocarcinoma cell line RENCA was maintained in RPMI 1640 supplemented with 10% FCS, l-glutamine, and penicillin/streptomycin.

Tumor models

Groups of BALB/c or BALB/c gene–targeted mice were injected s.c. with 1 × 105 RENCA cells in RPMI media alone. The depletion schedule and dosage of Tregs with anti-CD4, anti-CD25, or anti-FR4 are outlined in Fig. 1A. Primary tumor growth was measured with digital calipers, and the tumor area was calculated by multiplying the shortest and longest axis of the tumor. Mice were culled when tumors reached 100 mm2 in the area. Groups of 20 male B6 WT and other gene-targeted mice were inoculated s.c. in the hind flank with 400 μg of 3-MCA (Sigma-Aldrich) in 0.1 mL of corn oil. Development of fibrosarcomas was monitored periodically over the course of 56 to 250 d. Tumors, with a diameter of >3 mm and showing progressive growth, were recorded as positive. The depletion schedule and dosage of Tregs with either rat anti-mouse CD4 (GK1.5), rat anti-mouse CD8 (53-6.7), rat anti-mouse CD25 (PC61), rat anti-mouse FR4 (Th6), or the control rat antibody MAC4 (cIg; all purified in house) are outlined in Fig. 5A and Supplementary Fig. S6A. Anti-ASGM1 was purchased from Wako Pure Chemicals.

Figure 1.

Mice depleted of CD4+, FR4+, or CD25+ T cells reject RENCA tumors. The standard mAb and tumor challenge regimen used in all experiments (unless otherwise noted) is depicted in A. Groups of mice received 500 μg (days −1, 0, and +1), thereafter followed by 250 μg of anti-CD4 (i.p.) as indicated, 500 μg of anti-CD25 (i.p.), or 10 μg of anti-FR4 (i.v.) on days indicated. All mice received 105 RENCA tumor cells s.c. on day 0. B, representative graph of the mean tumor growth of the various groups (calculated by the product of two perpendicular axes, mm2) ± SEM and plotted versus time. Number of cured mice in parentheses. C, the survival of all mAb-treated mice pooled from all mice (n = 77–170; pooled from 14–34 experiments) and plotted as percentage survival over time. D, depicts the growth of RENCA tumors in BALB/c WT, CD1d−/−, and Jα18−/− mice. WT, CD1d−/−, and Jα18−/− mice were challenged with 105 RENCA cells, and tumor growth was subsequently monitored by caliper square measurements. Data are presented as mean tumor area (mm2) ± SEM (n = 5 mice per group). Statistical differences in survival between tumor-bearing mice treated with the indicated antibodies and nontreated tumor-bearing mice were determined by log-rank test; *, P < 0.05.

Figure 1.

Mice depleted of CD4+, FR4+, or CD25+ T cells reject RENCA tumors. The standard mAb and tumor challenge regimen used in all experiments (unless otherwise noted) is depicted in A. Groups of mice received 500 μg (days −1, 0, and +1), thereafter followed by 250 μg of anti-CD4 (i.p.) as indicated, 500 μg of anti-CD25 (i.p.), or 10 μg of anti-FR4 (i.v.) on days indicated. All mice received 105 RENCA tumor cells s.c. on day 0. B, representative graph of the mean tumor growth of the various groups (calculated by the product of two perpendicular axes, mm2) ± SEM and plotted versus time. Number of cured mice in parentheses. C, the survival of all mAb-treated mice pooled from all mice (n = 77–170; pooled from 14–34 experiments) and plotted as percentage survival over time. D, depicts the growth of RENCA tumors in BALB/c WT, CD1d−/−, and Jα18−/− mice. WT, CD1d−/−, and Jα18−/− mice were challenged with 105 RENCA cells, and tumor growth was subsequently monitored by caliper square measurements. Data are presented as mean tumor area (mm2) ± SEM (n = 5 mice per group). Statistical differences in survival between tumor-bearing mice treated with the indicated antibodies and nontreated tumor-bearing mice were determined by log-rank test; *, P < 0.05.

Close modal

Statistical analysis

Statistical significance of tumor-free survival was assessed using a log-rank test (P < 0.05).

Treg depletion causes RENCA tumor rejection

To investigate if Tregs influence the growth of RENCA tumors, three depletion strategies were tested: anti-CD4, anti-CD25, or anti-FR4 mAb treatments. An experimental overview outlining treatment schedules and tumor challenges is shown in Fig. 1A. Multiple anti-CD4 injections were used to optimally deplete the larger number of CD4+ T cells, and this protocol was used throughout the RENCA studies. All three strategies equivalently depleted FoxP3+ cells in the lymph node (Supplementary Fig. S1), spleen, and blood (data not shown). Optimal depletion was observed at day 3 after the last mAb administration, and depletion was effective for up to 7 days in each case with respective FoxP3+ and FoxP3 cells returning equivalently from days 7 to 14 (data not shown). Anti-CD4 was most effective in depleting FoxP3+CD4+ cells but also depleted most other CD4+ T cells including FoxP3-CD4+ T cells (Supplementary Fig. S1C). Anti-CD25 and anti-FR4 were approximately equivalent in depleting FoxP3+ Treg, but anti-FR4 also depleted many FoxP3-FR4+ T cells (∼23–5%), comprising largely CD4+ T cells (Supplementary Figs. S1 and S2). Smaller populations of FoxP3 CD8+ T cells and FR4+TCRβ cells were also depleted (Supplementary Fig. S2); however, notably FR4 was only expressed on a very small population of NK cells (<10%; ref. 13). Anti-CD4, anti-CD25, and anti-FR4 mAb treatments were all able to stimulate rejection of RENCA tumors compared with untreated mice (Fig. 1B and C) or cIg-treated mice (data not shown). Consistently, tumors grew larger before regressing within 3 weeks in mice treated with anti-CD4 or anti-CD25 compared with those receiving anti-FR4 (Fig. 1B). Although the rate of tumor regression was variable between individual mice, no relapses were observed and no overt signs of autoimmunity were noted (over a 1-year observation period, data not shown; Fig. 1B). Over a large number of mice (n = 77–114) observed, anti-CD4 was more powerful in causing rejection of RENCA than anti-CD25 or anti-FR4 (Fig. 1C). A single day −1 and days −1, 0, and +1 protocol of anti-CD4 also enabled complete rejection of all RENCA tumors (data not shown). In some tumor models, anti-CD4 treatment can induce tumor rejection by depleting regulatory CD1d-restricted T cells (23); however, growth of RENCA tumors in CD1d−/− and Jα18−/− mice was equivalent to that in WT mice (Fig. 1D). Treg depletion failed to stimulate antitumor immunity when used therapeutically (starting on day 10 post tumor challenge) against RENCA (Supplementary Fig. S3).

CD8+ cells are required for RENCA tumor rejection following Treg depletion

To determine how RENCA tumors were rejected following Treg depletion, mice were first depleted of CD8+ T cells coincident with anti-CD4, anti-FR4, or anti-CD25 treatments and then challenged with RENCA. None of the 10 mice treated with anti-CD4 plus anti-CD8, 1 of the 10 anti-FR4 plus anti-CD8, and 0 of the18 mice treated with anti-CD25 plus anti-CD8 were able to reject RENCA tumors, showing that CD8+ T cells were essential for RENCA tumor rejection (all P < 0.0001 compared with Treg depletion alone; Fig. 2A). By contrast, sustained NK cell depletion alone had little or no effect on the ability of anti-CD4–treated or anti-FR4–treated mice to reject tumors. Interestingly, NK cell–depleted tumor-bearing mice treated with anti-CD25 were less able to reject tumors compared with tumor-bearing mice treated with only anti-CD25 (P < 0.0001); however, none of the nontreated tumor-bearing mice survived (P < 0.0001; Fig. 2B). This suggested that NK cells alone were not essential for tumor rejection following anti-CD4 or anti-FR4 treatment but were partially required following anti-CD25 treatment. Depletion of both NK cells and CD8+ T cells abrogated all protection from RENCA afforded by anti-CD4, anti-FR4, or anti-CD25 treatments (Fig. 2C). A role for CD8+ T cells was further supported by showing that anti-CD4–mediated, anti-FR4–mediated, and anti-CD25–mediated protection from RENCA tumor was associated with an increase in the proportion of tumor-infiltrating CD8+ T cells compared with control immunoglobulin-treated tumors (Supplementary Fig. S4A and B). We monitored the ex vivo restimulated cytolytic activity of tumor draining CD8+ T cells after anti-CD4, anti-FR4, and anti-CD25 treatments, and it was clear that all three treatments significantly enhanced the tumor-specific cytotoxic activity of CD8+ T cells compared with equivalent control mice bearing RENCA tumors (Supplementary Fig. S4C).

Figure 2.

CD8+ T cells are required for tumor rejection. Groups of BALB/c mice were injected s.c. with 105 RENCA cells and treated with mAbs, as outlined in Fig. 1A. A, the indicated groups of mice were additionally depleted of CD8+ T cells by i.p. injection of 100 μg of anti-CD8 on days −1, 0, and 1 and twice weekly thereafter. B, the indicated groups of mice were alternatively depleted of NK cells by i.v. injection of 100 μg of anti-ASGM1 on days −1 and 0 and every 7 d thereafter until tumors regressed or mice were culled. C, the indicated groups of mice were depleted of both CD8+ T and NK cells as in A and B. The survival of all antibody-treated mice was calculated from multiple experiments (pooled from 2 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between tumor-bearing mice treated with the indicated antibodies and tumor-bearing mice depleted of CD8+ T cells and/or NK cells and treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Figure 2.

CD8+ T cells are required for tumor rejection. Groups of BALB/c mice were injected s.c. with 105 RENCA cells and treated with mAbs, as outlined in Fig. 1A. A, the indicated groups of mice were additionally depleted of CD8+ T cells by i.p. injection of 100 μg of anti-CD8 on days −1, 0, and 1 and twice weekly thereafter. B, the indicated groups of mice were alternatively depleted of NK cells by i.v. injection of 100 μg of anti-ASGM1 on days −1 and 0 and every 7 d thereafter until tumors regressed or mice were culled. C, the indicated groups of mice were depleted of both CD8+ T and NK cells as in A and B. The survival of all antibody-treated mice was calculated from multiple experiments (pooled from 2 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between tumor-bearing mice treated with the indicated antibodies and tumor-bearing mice depleted of CD8+ T cells and/or NK cells and treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Close modal

Effector cytokines required for rejection of RENCA tumors following Treg depletion

Next, the role of the major Th1 and Th2 polarizing cytokines was investigated. All anti-CD4–treated IFN-γ−/− (8 of 8) and WT (114 of 114) mice were able to reject RENCA tumors, whereas control mice of both genotypes (24 of 24 and 170 of 170, respectively) all succumbed to tumor growth (Fig. 3A). Similarly, the majority of anti-FR4–treated IFN-γ−/− (11 of 12) and WT (58 of 77) mice were able to reject RENCA tumors (Fig. 3A). In contrast, whereas 90% (72 of 80) WT mice treated with anti-CD25 were able to reject RENCA tumors, no IFN-γ−/− mice treated with anti-CD25 (0 of 15 mice) survived tumor challenge (P < 0.0001 compared with WT mice treated with anti-CD25; Fig. 3A). Thus, whereas IFN-γ was required for tumor rejection following anti-CD25 treatment, it was dispensable for tumor clearance following anti-CD4 or anti-FR4 treatment.

Figure 3.

IL-4, IL-13, and IFN-γ are required for tumor rejection following anti-CD25 but not anti-CD4 or anti-FR4 treatment. Groups of BALB/c WT (A–C), IFN-γ−/− (A), IL-4−/− (B), or IL-13−/− (C) mice were injected s.c. with 105 RENCA cells on day 0. In all panels, WT and gene-targeted mice were treated as described in Fig. 1A. The survival of WT or gene-targeted mice after mAb treatment was calculated from multiple experiments (pooled from 1 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between WT tumor-bearing mice treated with the indicated antibodies and gene-targeted tumor-bearing mice treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Figure 3.

IL-4, IL-13, and IFN-γ are required for tumor rejection following anti-CD25 but not anti-CD4 or anti-FR4 treatment. Groups of BALB/c WT (A–C), IFN-γ−/− (A), IL-4−/− (B), or IL-13−/− (C) mice were injected s.c. with 105 RENCA cells on day 0. In all panels, WT and gene-targeted mice were treated as described in Fig. 1A. The survival of WT or gene-targeted mice after mAb treatment was calculated from multiple experiments (pooled from 1 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between WT tumor-bearing mice treated with the indicated antibodies and gene-targeted tumor-bearing mice treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Close modal

Anti-CD4 was able to induce rejection of RENCA tumors in 100% of IL-4–deficient and IL-13–deficient mice (Fig. 3B and C, respectively), whereas anti-FR4 caused reduced rejection of RENCA tumors in IL-4−/− (45.5%, 10 of 22; P = 0.00053) and IL-13−/− (52.9%, 9 of 17; P = 0.02923) mice. Whereas anti-CD25 treatment was able to induce RENCA tumor rejection in 90% of WT mice (72 of 80), only 18.2% (2 of 11) of IL-4−/− mice and 18.2% (2 of 11) of IL-13−/− mice treated in parallel rejected their tumors (P < 0.0001 and P < 0.0001, respectively; Fig. 3B and C). Thus, Th2 cytokines are key for rejection of RENCA tumors following anti-CD25, partially important after anti-FR4 and not relevant after anti-CD4 treatment. Collectively, these data showed that tumor rejection following anti-CD25 treatment required a combination of both Th1 and Th2 cytokines (IFN-γ, IL-4, and IL-13), whereas tumor rejection following anti-CD4 occurred in the absence of the major Th1 and Th2 polarizing cytokines. In contrast, tumor rejection following anti-FR4 treatment did not require IFN-γ but was partially dependent on Th2 cytokines (IL-4, IL-13).

The role of cytotoxic effector molecules in tumor rejection

Perforin was found to be dispensable for RENCA tumor rejection in anti-CD25–treated mice (Fig. 4A). A minor reduction in tumor rejection was noted in either anti-CD4–treated or anti-FR4–treated pfp−/− mice compared with WT mice (Fig. 4A; P < 0.0002 and P = 0.0104, respectively); however, anti-FR4–treated pfp−/− mice were still able to mediate tumor rejection compared with control-treated mice (P < 0.0001). Tumors grew at a similar rate in WT and pfp−/− mice, and all control-treated mice succumbed to tumor growth with similar kinetics. Very similar results were obtained in TRAIL−/− mice, wherein anti-CD4 and anti-CD25 treatments were able to induce RENCA tumor rejection almost equivalently to WT mice, providing further evidence that this effector pathway was also not essential (Fig. 4B).

Figure 4.

Differential requirements for perforin and TRAIL in tumor rejection following anti-CD4, anti-FR4, or anti-CD25 treatment. Groups of BALB/c WT (A and B), pfp−/− (A), or TRAIL−/− (B) mice were injected s.c. with 105 RENCA cells on day 0. In all panels WT and gene-targeted mice were treated as described in Fig. 1A. The survival of WT or gene-targeted mice after mAb treatment was calculated from multiple experiments (pooled from 1 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between WT tumor-bearing mice treated with the indicated antibodies and gene-targeted tumor-bearing mice treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Figure 4.

Differential requirements for perforin and TRAIL in tumor rejection following anti-CD4, anti-FR4, or anti-CD25 treatment. Groups of BALB/c WT (A and B), pfp−/− (A), or TRAIL−/− (B) mice were injected s.c. with 105 RENCA cells on day 0. In all panels WT and gene-targeted mice were treated as described in Fig. 1A. The survival of WT or gene-targeted mice after mAb treatment was calculated from multiple experiments (pooled from 1 to 34 experiments) and is plotted as percentage survival over time. Statistical differences in survival between WT tumor-bearing mice treated with the indicated antibodies and gene-targeted tumor-bearing mice treated with the indicated antibodies were determined by log-rank test; *, P < 0.05.

Close modal

We next used three strategies to investigate the contribution of death receptor signaling to tumor regression: blocking antibodies, RENCA cells overexpressing the antiapoptotic molecule FLIP (24), and where possible gene-targeted mice. In WT mice, treatment with a combination of anti-FasL, anti-TRAIL, and anti-tumor necrosis factor-α (TNF-α; called anti-FTT) had no significant effect on tumor rejection following either anti-CD4 or anti-CD25 treatment, implying that FasL, TRAIL, and TNF-α were not essential for tumor clearance (Supplementary Fig. S5A). Anti-CD25–treated mice were able to reject RENCA-FLIP and RENCA-Flag tumors with similar efficiency, whereas anti-CD4 was able to induce the rejection of all RENCA-Flag tumors and most RENCA-FLIP tumors (not statistically different, P > 0.05; Supplementary Fig. S5B). These results suggested that tumor rejection following anti-CD25 or anti-CD4 treatment occurred independently of death receptor signaling.

As expected, anti-CD25–treated pfp−/−IFN-γ−/− mice failed to effectively eliminate RENCA tumors, with only 2 of 17 mice surviving tumor challenge (P < 0.0001 compared with anti-CD25–treated WT mice tested in parallel), in keeping with a requirement for IFN-γ in tumor rejection following anti-CD25 treatment (Supplementary Fig. S6A). In contrast, all (10 of 10) anti-CD4–treated pfp−/−IFN-γ−/− mice were able to reject RENCA tumors. The effect of the simultaneous blockade of the pfp, IFN-γ, FasL, TRAIL, and TNF-α pathways on tumor clearance was also investigated, and anti-CD4 mAb–treated mice were still able to reject RENCA tumors (Supplementary Fig. S6B) and RENCA-FLIP and RENCA-Flag tumors (Supplementary Fig. S6C). Thus, following CD4 depletion, tumor rejection still occurred in the collective absence of all five effector molecules.

Treg depletion suppresses the development of MCA-induced sarcoma

To investigate if Tregs also influence the development of spontaneous tumors, two variations of the same three depletion strategies (anti-CD4, anti-CD25, or anti-FR4 mAb treatment) were tested (experimental overview outlining treatment schedules and MCA challenges; Fig. 5A and Supplementary Fig. S7A). As shown in BALB/c mice, similarly effective depletions of populations of FoxP3+ T cells were observed for each treatment schedule in C57BL/6 spleen, lymph nodes, and blood (data not shown). Anti-CD4, anti-CD25, and anti-FR4 mAb treatments were all able to prevent the development of MCA-induced sarcomas compared with control mAb (Fig. 5B–D and Supplementary Fig. S7B–D). Consistently, anti-FR4 reduced sarcoma formation regardless of whether it was given before MCA inoculation or twice weekly for 4 weeks from the point of MCA inoculation (Fig. 5D and Supplementary Fig. S7D). Anti-CD4 and anti-CD25 were also effective (Fig. 5B and C and Supplementary Fig. S7B and C), but anti-CD25 seemed less effective when given in a prolonged fashion (Supplementary Fig. S7C). No overt signs of autoimmunity were noted (over a 1-year observation period) in any of these mice that remained tumor free (data not shown).

Figure 5.

Treg depletion suppresses the development of MCA-induced sarcoma. Groups of male C57BL/6 mice (n = 20) were injected s.c. with MCA (400 μg) on day 0. The schedules for depleting Tregs on days −1 and 0 (A) with anti-CD4 (B), anti-CD25 (C), anti-FR4 (D), or cIg (B–D) mAbs are depicted. Anti-CD4 and anti-CD25 mAbs were injected i.p., whereas anti-FR4 mAbs were injected i.v. Tumor growth was monitored weekly by caliper square measurements, and the percentage of tumor-free mice of each group of mice is shown plotted against days after MCA inoculation. Statistical differences in the proportion of tumor-free mice treated with anti-CD4, anti-CD25, or anti-FR4 and tumor-free mice treated with cIg (groups of 20 mice per treatment) were determined by log-rank test; *, P < 0.05.

Figure 5.

Treg depletion suppresses the development of MCA-induced sarcoma. Groups of male C57BL/6 mice (n = 20) were injected s.c. with MCA (400 μg) on day 0. The schedules for depleting Tregs on days −1 and 0 (A) with anti-CD4 (B), anti-CD25 (C), anti-FR4 (D), or cIg (B–D) mAbs are depicted. Anti-CD4 and anti-CD25 mAbs were injected i.p., whereas anti-FR4 mAbs were injected i.v. Tumor growth was monitored weekly by caliper square measurements, and the percentage of tumor-free mice of each group of mice is shown plotted against days after MCA inoculation. Statistical differences in the proportion of tumor-free mice treated with anti-CD4, anti-CD25, or anti-FR4 and tumor-free mice treated with cIg (groups of 20 mice per treatment) were determined by log-rank test; *, P < 0.05.

Close modal

CD8+ and NK cells collectively account for MCA-induced sarcoma suppression following anti-FR4 treatment

Anti-FR4 prevented MCA-induced sarcoma in mice depleted of either CD8+ T cells (Fig. 6A) or NK cells (Fig. 6B), showing that neither CD8+ T cells nor NK cells were essential for tumor rejection stimulated by Treg depletion. By contrast, combined CD8+ T-cell and NK cell depletion resulted in a complete loss of the protection afforded by anti-FR4–mediated Treg depletion (Fig. 6C). Type I NKT cells are thought to be upstream of NK cells in natural host immune protection from MCA-induced sarcoma (18), but interestingly anti-FR4–mediated Treg depletion in type I NKT cell–deficient Jα18−/− mice was still effective in affording mice protection from MCA-induced sarcoma (Fig. 6D). Interestingly, combined CD8+ T-cell and NK cell depletion also resulted in a complete loss of the protection afforded by anti-CD25–mediated (Supplementary Fig. S8A–D) and anti-CD4–mediated Treg depletion (data not shown). Thus following Treg depletion, either CD8+ T cells or NK cells can act to prevent MCA-induced sarcomas.

Figure 6.

CD8+ and NK cells collectively account for MCA-induced sarcoma suppression following anti-FR4 treatment. Groups of male C57BL/6 or C57BL/6 Jα18−/− mice (n = 20) were injected s.c. with MCA (400 μg) and treated i.v. with anti-FR4 mAbs [on days −1 (50 μg) and 0 (10 μg), as outlined in Fig. 5A]. The indicated groups of mice were additionally depleted of CD8+ T cells by i.p. injection of 100 μg of anti-CD8 on days −1, 0, and +1 and thereafter twice weekly for 4 wk (A); NK cells by i.p. injection of 100 μg of anti-ASGM1 on days −1, 0, and +1 and thereafter weekly for 4 wk (B); or CD8+ T cells and NK cells together (C). D, type I NKT cell–deficient C57B/L6 Jα18−/− mice were treated with control immunoglobulin or anti-FR4 on days −1 and 0 as above. Mice were monitored for tumor development, and proportion of tumor-free mice is plotted against days after MCA inoculation. Statistical differences in the proportions of tumor-free mice between groups depleted of CD8+ T cells or NK cells treated with anti-CD4, anti-CD25, or anti-FR4 and tumor-free mice depleted of CD8+ T cells or NK cells treated with cIg (groups of 20 mice per treatment) were determined by log-rank test; *, P < 0.05.

Figure 6.

CD8+ and NK cells collectively account for MCA-induced sarcoma suppression following anti-FR4 treatment. Groups of male C57BL/6 or C57BL/6 Jα18−/− mice (n = 20) were injected s.c. with MCA (400 μg) and treated i.v. with anti-FR4 mAbs [on days −1 (50 μg) and 0 (10 μg), as outlined in Fig. 5A]. The indicated groups of mice were additionally depleted of CD8+ T cells by i.p. injection of 100 μg of anti-CD8 on days −1, 0, and +1 and thereafter twice weekly for 4 wk (A); NK cells by i.p. injection of 100 μg of anti-ASGM1 on days −1, 0, and +1 and thereafter weekly for 4 wk (B); or CD8+ T cells and NK cells together (C). D, type I NKT cell–deficient C57B/L6 Jα18−/− mice were treated with control immunoglobulin or anti-FR4 on days −1 and 0 as above. Mice were monitored for tumor development, and proportion of tumor-free mice is plotted against days after MCA inoculation. Statistical differences in the proportions of tumor-free mice between groups depleted of CD8+ T cells or NK cells treated with anti-CD4, anti-CD25, or anti-FR4 and tumor-free mice depleted of CD8+ T cells or NK cells treated with cIg (groups of 20 mice per treatment) were determined by log-rank test; *, P < 0.05.

Close modal

Effector mechanisms required for suppression of MCA-induced sarcoma following anti-FR4 treatment

Anti-FR4 treatment prevented MCA-induced sarcoma in pfp−/− (Supplementary Fig. S9A), granzyme B−/− (Supplementary Fig. S9B), DR5−/− (Supplementary Fig. S9C), TNF-α−/− (Supplementary Fig. S9D), and IL-4−/− (Supplementary Fig. S9E) mice, showing that neither pfp, granzyme B, TRAIL, TNF-α, nor IL-4 was essential for tumor rejection stimulated by Treg depletion. By contrast, loss of IFN-γ (Supplementary Fig. S9F) somewhat reduced the effectiveness of Treg depletion, and mice deficient for both pfp and IFN-γ showed no protection afforded by anti-FR4–mediated Treg depletion (Supplementary Fig. S9G). Given the importance of IL-4 and IFN-γ in anti-CD25–mediated rejection of RENCA, we also examined MCA sarcoma induction in these mice following anti-CD25 treatment. Clearly, like anti-FR4 treatment, IFN-γ, but not IL-4, was essential for the prevention of MCA-induced sarcomas stimulated by anti-CD25 (Supplementary Fig. S10A and B).

In this study we examined the mechanism of host immune suppression of experimental RENCA tumors or chemical carcinogen–induced fibrosarcomas following Treg depletion. Three methods of early depletion of regulatory FoxP3+ T cells were used (anti-CD4, anti-FR4, or anti-CD25 treatment) and collectively, while it was clear that CD8+ T cells and NK cells played an important role downstream of Treg depletion, each method revealed subtly different effector pathways of tumor rejection. The tumors used in this study did not express FR4, CD4, or CD25, and thus, the effect of the mAbs was not directly on the tumor but rather on the host (data not shown). In all cases, CD8+ T cells were required for RENCA tumor rejection; however, following anti-CD25 treatment tumor rejection required an unusual combination of IFN-γ, IL-4, and IL-13 and occurred independently of the tested death-inducing pathways (perforin, TRAIL, FasL, and TNF-α). By contrast, tumor rejection following anti-CD4 or anti-FR4 depletion did not require any of the cytokines examined (IFN-γ, IL-4, and IL-13 were dispensable). Tumor rejection following anti-FR4 depletion seemed to partially involve perforin and TRAIL pathways, but neither pathway was critical. Despite the large number of cytokines and effector pathways tested, the current study failed to determine how RENCA tumors were eliminated following CD4 depletion.

Early depletion using anti-CD25, anti-CD4, and anti-FR4 mAbs was also effective in preventing MCA-induced fibrosarcoma. Further analysis of anti-FR4 treatment revealed that Treg depletion remained largely effective in preventing tumor formation in single effector molecule (pfp, granzyme B, IL-4, DR5, TNF-α)–deficient mice or those mice deficient in CD8+ T cells, type I NKT cells, or NK cells alone. By contrast, all Treg depletion strategies were completely ineffective in mice depleted of both CD8+ T cells and NK cells, anti-FR4 and anti-CD25 were less effective in IFN-γ–deficient mice, and anti-FR4 was not effective in mice doubly deficient for pfp and IFN-γ. Collectively, this work highlights the redundancy and diversity of effector responses that must be considered when removing key checkpoints in immune regulatory control.

The finding that pfp was not essential for RENCA tumor rejection following Treg depletion was interesting in the light of the fact that all methods of depletion enhanced the proportion of CD8+ tumor-infiltrating lymphocytes and increased the cytotoxic capacity of CTL in the tumor-draining lymph nodes. Furthermore, two previous studies suggested that Tregs control antitumor CD8+ T-cell responses by inhibiting cytotoxicity (7, 25). However, whereas RENCA cells are susceptible to pfp-mediated lysis in vitro, RENCA-specific CD8+ T cells can protect mice from growth of RENCA tumors in vivo independently of pfp (24). Furthermore, the cytokines IFN-γ, IL-4, and IL-13 were more crucial for tumor rejection in the RENCA model, as mice lacking these cytokines were unable to clear tumors following anti-CD25 treatment. Our finding that tumor rejection in the RENCA model is more dependent on cytokines indicates that Treg may affect the antitumor immune response at a number of levels, depending on the model system under study. In concert, another recent study has suggested that Tregs act to inhibit the accumulation of IFN-γ–producing CD8+ T cells within tumor tissues rather than by inhibiting CTL cytotoxic activity (26). Notably, in our study, whereas anti-FR4 was equally effective in WT and pfp-deficient mice, a role for pfp in the carcinogen-induced fibrosarcoma model was revealed by the reduced effectiveness of anti-FR4 in pfp−/− IFN-γ−/− mice compared with IFN-γ–deficient mice.

Unexpectedly, rejection of RENCA tumors following anti-CD25 treatment required both Th1 and Th2 type cytokines. Generally Th1 and Th2 responses are thought to represent opposite poles of an immune response (27), and in some cases, they directly antagonize each other's functions. A role for IFN-γ in rejection of tumors following Treg depletion has been previously documented (5); however, a role of IL-4 and IL-13 has not previously been described. It is clear that under some circumstances Th2-type responses to tumors can be beneficial (2832), including in the RENCA model (33). Furthermore, evidence that Th1 and Th2 responses can act in concert to eliminate tumors has been reported (34), including in the RENCA model (35). Collectively, these observations support that notion that in some cases Th1 and Th2 cytokines can act in concert to generate effective antitumor responses. The tumor cytokine microenvironment resulting in mice that had been depleted with anti-CD4, anti-CD25, or anti-FR4 may regulate the type of effector response mounted; however, precisely determining Treg factors that suppress effector responses in these tumors will require the use of conditional knockouts of candidate suppressor molecules expressed by Treg and/or adoptive transfer techniques.

Models that use carcinogens, like MCA, may be considered more relevant for deconstructing the relationship between the immune system and developing tumors. It was revealing that such early and transient Treg depletion was sufficient to confer protection in the MCA model, and these data would suggest that the FR4hiFoxP3+ cell subset of Tregs very effectively depleted by this protocol is particularly important in suppressing the immune response to carcinogen inoculation. Certainly our preliminary analyses indicate that anti-FR4 is not effective in significantly suppressing established tumor growth of de novo MCA-induced sarcomas (data not shown). A key question arising from the studies of Betts and colleagues (21) and Nishikawa and colleagues (22) using MCA-induced sarcomas was this: what type of effector cells promoted tumor suppression after depletion of Tregs? Tregs induced by immunization inhibited NK and NKT cells capable of preventing the development of MCA-induced tumors (22). Here we have shown for the first time that CD8+ T cells, NK cells, and type I NKT cells alone are not essential, rather a combination of CD8+ T cells and NK cells seemed to account for the immune effector function stimulated by Treg depletion by anti-FR4 or anti-CD25. Extending the work of Betts and colleagues (21), we assessed a variety of effector pathways potentially downstream of Treg depletion, including IFN-γ, pfp, IL-4, granzyme B, TNF-α, and TRAIL. No pathway alone was absolutely essential, although in agreement with Betts and colleagues (21) IFN-γ was important. Our data support recent findings in K-ras–driven tumorigenesis, which indicate the central role for FoxP3+ Tregs in preventing lung adenocarcinomas (36) and a previous report indicating the role of CD25+ Tregs in promoting erbB2-driven mammary carcinoma (37).

Overall, the study presented here shows that a number of redundant effector mechanisms can potentially eradicate tumors when Treg suppression mechanisms are prophylactically abrogated. Clearly the finding that the method of Treg depletion can significantly influence the downstream immune responses raises some significant issues for the application of various Treg depletion strategies in priming immunity in the context of prophylactic vaccines or minimal residual disease.

No potential conflicts of interest were disclosed.

We thank Mark Shannon for reagent acquisition; Ben Martin for genotyping; Rachel Cameron, Shannon Griffiths, and Michelle Stirling for maintaining the gene-targeted mice; and Yoshihiro Hayakawa for helpful suggestions.

Grant Support: National Health and Medical Research Council of Australia (NH&MRC) Peter Doherty Fellowship (M.W.L. Teng), Australian Postgraduate Award (J.B. Swann), NH&MRC Career Development Award (P.K. Darcy), and NH&MRC Australia Fellowship and Program Grant and Parker Family and Peter Mac Foundation kidney cancer research (M.J. Smyth).

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
Onizuka
S
,
Tawara
I
,
Shimizu
J
,
Sakaguchi
S
,
Fujita
T
,
Nakayama
E
. 
Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor α) monoclonal antibody
.
Cancer Res
1999
;
59
:
3128
33
.
2
Golgher
D
,
Jones
E
,
Powrie
F
,
Elliott
T
,
Gallimore
A
. 
Depletion of CD25+ regulatory cells uncovers immune responses to shared murine tumor rejection antigens
.
Eur J Immunol
2002
;
32
:
3267
75
.
3
Tawara
I
,
Take
Y
,
Uenaka
A
,
Noguchi
Y
,
Nakayama
E
. 
Sequential involvement of two distinct CD4+ regulatory T cells during the course of transplantable tumor growth and protection from 3-methylcholanthrene-induced tumorigenesis by CD25-depletion
.
Jpn J Cancer Res
2002
;
93
:
911
6
.
4
Yu
P
,
Lee
Y
,
Liu
W
, et al
. 
Intratumor depletion of CD4+ cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors
.
J Exp Med
2005
;
201
:
779
91
.
5
Casares
N
,
Arribillaga
L
,
Sarobe
P
, et al
. 
CD4+/CD25+ regulatory cells inhibit activation of tumor-primed CD4+ T cells with IFN-γ-dependent antiangiogenic activity, as well as long-lasting tumor immunity elicited by peptide vaccination
.
J Immunol
2003
;
171
:
5931
9
.
6
Ko
K
,
Yamazaki
S
,
Nakamura
K
, et al
. 
Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells
.
J Exp Med
2005
;
202
:
885
91
.
7
Chen
ML
,
Pittet
MJ
,
Gorelik
L
, et al
. 
Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-β signals in vivo
.
Proc Natl Acad Sci U S A
2005
;
102
:
419
24
.
8
den Boer
AT
,
van Mierlo
GJ
,
Fransen
MF
,
Melief
CJ
,
Offringa
R
,
Toes
RE
. 
CD4+ T cells are able to promote tumor growth through inhibition of tumor-specific CD8+ T-cell responses in tumor-bearing hosts
.
Cancer Res
2005
;
65
:
6984
9
.
9
Ghiringhelli
F
,
Menard
C
,
Martin
F
,
Zitvogel
L
. 
The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression
.
Immunol Rev
2006
;
214
:
229
38
.
10
Ghiringhelli
F
,
Menard
C
,
Terme
M
, et al
. 
CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-β-dependent manner
.
J Exp Med
2005
;
202
:
1075
85
.
11
Smyth
MJ
,
Teng
MW
,
Swann
J
,
Kyparissoudis
K
,
Godfrey
DI
,
Hayakawa
Y
. 
CD4+CD25+ T regulatory cells suppress NK cell-mediated immunotherapy of cancer
.
J Immunol
2006
;
176
:
1582
7
.
12
Terme
M
,
Chaput
N
,
Combadiere
B
,
Ma
A
,
Ohteki
T
,
Zitvogel
L
. 
Regulatory T cells control dendritic cell/NK cell cross-talk in lymph nodes at the steady state by inhibiting CD4+ self-reactive T cells
.
J Immunol
2008
;
180
:
4679
86
.
13
Yamaguchi
T
,
Hirota
K
,
Nagahama
K
, et al
. 
Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor
.
Immunity
2007
;
27
:
145
59
.
14
Quezada
SA
,
Peggs
KS
,
Simpson
TR
,
Shen
Y
,
Littman
DR
,
Allison
JP
. 
Limited tumor infiltration by activated T effector cells restricts the therapeutic activity of regulatory T cell depletion against established melanoma
.
J Exp Med
2008
;
205
:
2125
38
.
15
Seki
N
,
Brooks
AD
,
Carter
CR
, et al
. 
Tumor-specific CTL kill murine renal cancer cells using both perforin and Fas ligand-mediated lysis in vitro, but cause tumor regression in vivo in the absence of perforin
.
J Immunol
2002
;
168
:
3484
92
.
16
Smyth
MJ
,
Crowe
NY
,
Godfrey
DI
. 
NK cells and NKT cells collaborate in host protection from methylcholanthrene-induced fibrosarcoma
.
Int Immunol
2001
;
13
:
459
63
.
17
Smyth
MJ
,
Thia
KY
,
Street
SE
, et al
. 
Differential tumor surveillance by natural killer (NK) and NKT cells
.
J Exp Med
2000
;
191
:
661
8
.
18
Crowe
NY
,
Smyth
MJ
,
Godfrey
DI
. 
A critical role for natural killer T cells in immunosurveillance of methylcholanthrene-induced sarcomas
.
J Exp Med
2002
;
196
:
119
27
.
19
Crowe
NY
,
Coquet
JM
,
Berzins
SP
, et al
. 
Differential antitumor immunity mediated by NKT cell subsets in vivo
.
J Exp Med
2005
;
202
:
1279
88
.
20
Bui
JD
,
Uppaluri
R
,
Hsieh
CS
,
Schreiber
RD
. 
Comparative analysis of regulatory and effector T cells in progressively growing versus rejecting tumors of similar origins
.
Cancer Res
2006
;
66
:
7301
9
.
21
Betts
G
,
Twohig
J
,
Van den Broek
M
,
Sierro
S
,
Godkin
A
,
Gallimore
A
. 
The impact of regulatory T cells on carcinogen-induced sarcogenesis
.
Br J Cancer
2007
;
96
:
1849
54
.
22
Nishikawa
H
,
Kato
T
,
Tawara
I
, et al
. 
Accelerated chemically induced tumor development mediated by CD4+CD25+ regulatory T cells in wild-type hosts
.
Proc Natl Acad Sci U S A
2005
;
102
:
9253
7
.
23
Terabe
M
,
Matsui
S
,
Noben-Trauth
N
, et al
. 
NKT cell-mediated repression of tumor immunosurveillance by IL-13 and the IL-4R-STAT6 pathway
.
Nat Immunol
2000
;
1
:
515
20
.
24
Seki
N
,
Hayakawa
Y
,
Brooks
AD
, et al
. 
Tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis is an important endogenous mechanism for resistance to liver metastases in murine renal cancer
.
Cancer Res
2003
;
63
:
207
13
.
25
Mempel
TR
,
Pittet
MJ
,
Khazaie
K
, et al
. 
Regulatory T cells reversibly suppress cytotoxic T cell function independent of effector differentiation
.
Immunity
2006
;
25
:
129
41
.
26
Rudge
G
,
Barrett
SP
,
Scott
B
,
van Driel
IR
. 
Infiltration of a mesothelioma by IFN-γ-producing cells and tumor rejection after depletion of regulatory T cells
.
J Immunol
2007
;
178
:
4089
96
.
27
Mosmann
TR
,
Sad
S
. 
The expanding universe of T-cell subsets: Th1, Th2 and more
.
Immunol Today
1996
;
17
:
138
46
.
28
Pericle
F
,
Giovarelli
M
,
Colombo
MP
, et al
. 
An efficient Th2-type memory follows CD8+ lymphocyte-driven and eosinophil-mediated rejection of a spontaneous mouse mammary adenocarcinoma engineered to release IL-4
.
J Immunol
1994
;
153
:
5659
73
.
29
Shen
Y
,
Fujimoto
S
. 
A tumor-specific Th2 clone initiating tumor rejection via primed CD8+ cytotoxic T-lymphocyte activation in mice
.
Cancer Res
1996
;
56
:
5005
11
.
30
Rodolfo
M
,
Zilocchi
C
,
Accornero
P
,
Cappetti
B
,
Arioli
I
,
Colombo
MP
. 
IL-4-transduced tumor cell vaccine induces immunoregulatory type 2 CD8 T lymphocytes that cure lung metastases upon adoptive transfer
.
J Immunol
1999
;
163
:
1923
8
.
31
Dobrzanski
MJ
,
Reome
JB
,
Dutton
RW
. 
Therapeutic effects of tumor-reactive type 1 and type 2 CD8+ T cell subpopulations in established pulmonary metastases
.
J Immunol
1999
;
162
:
6671
80
.
32
Yamaguchi
N
,
Hiraoka
S
,
Mukai
T
, et al
. 
Induction of tumor regression by administration of B7-Ig fusion proteins: mediation by type 2 CD8+ T cells and dependence on IL-4 production
.
J Immunol
2004
;
172
:
1347
54
.
33
Golumbek
PT
,
Lazenby
AJ
,
Levitsky
HI
, et al
. 
Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4
.
Science
1991
;
254
:
713
6
.
34
Schuler
T
,
Qin
Z
,
Ibe
S
,
Noben-Trauth
N
,
Blankenstein
T
. 
T helper cell type 1-associated and cytotoxic T lymphocyte-mediated tumor immunity is impaired in interleukin 4-deficient mice
.
J Exp Med
1999
;
189
:
803
10
.
35
Hillman
GG
,
Younes
E
,
Visscher
D
, et al
. 
Inhibition of murine renal carcinoma pulmonary metastases by systemic administration of interferon γ: mechanism of action and potential for combination with interleukin 4
.
Clin Cancer Res
1997
;
3
:
1799
806
.
36
Granville
CA
,
Memmott
RM
,
Balogh
A
, et al
. 
A central role for Foxp3+ regulatory T cells in K-Ras-driven lung tumorigenesis
.
PLoS ONE
2009
;
4
:
e5061
.
37
Ambrosino
E
,
Spadaro
M
,
Iezzi
M
, et al
. 
Immunosurveillance of Erbb2 carcinogenesis in transgenic mice is concealed by a dominant regulatory T-cell self-tolerance
.
Cancer Res
2006
;
66
:
7734
40
.