In preclinical tumor models, αOX40 therapy is often successful at treating small tumors, but is less effective once the tumors become large. For a tumor immunotherapy to be successful to cure large tumors, it will most likely require not only an agonist to boost effector T-cell function but also inhibitors of T-cell suppression. In this study, we show that combining αOX40 antibodies with an inhibitor of the TGFβ receptor (SM16) synergizes to elicit complete regression of large established MCA205 and CT26 tumors. Evaluation of tumor-infiltrating T cells showed that SM16/αOX40 dual therapy resulted in an increase in proliferating granzyme B+ CD8 T cells, which produced higher levels of IFNγ, compared with treatment with either agent alone. We also found that the dual treatment increased pSTAT3 expression in both CD4 and CD8 T cells isolated from tumors. Because others have published that STAT3 signaling is detrimental to T-cell function within the tumor microenvironment, we explored whether deletion of STAT3 in OX40-expressing cells would affect this potent combination therapy. Surprisingly, we found that deletion of STAT3 in OX40-expressing cells decreased the efficacy of this combination therapy, showing that the full therapeutic potential of this treatment depends on STAT3 signaling, most likely in the T cells of tumor-bearing mice. Cancer Immunol Res; 3(5); 526–35. ©2015 AACR.

OX40 (CD134, TNFRSF4) is a member of the TNF receptor (TNFR) superfamily expressed on activated CD4+ and CD8+ T cells (1–3). Engagement of OX40 with cognate ligand or soluble agonist enhances T-cell proliferation, survival, and cytokine production by activated T cells (4). In addition to OX40 being an activation antigen on T cells, regulatory T cells (Treg) constitutively express OX40 in mice. Under certain conditions, treatment with OX40 agonist antibodies (αOX40) mitigates Treg development and function while at the same time enhancing effector T-cell function (4, 5). Importantly, a murine anti-human OX40 antibody has recently completed a phase I clinical trial for the treatment of patients with advanced cancer with encouraging results (6). Currently, humanized OX40 agonists are being developed to conduct future trials in patients with cancer.

Although αOX40 therapy has been effective at controlling small tumors (0–50 mm2) in a variety of mouse tumor models (3, 7, 8), it has been less effective at eradicating large established tumors (>50 mm2; refs. 8–11). A potential explanation for this deficiency is that the microenvironment of large tumors becomes immune suppressive, which most likely reflects tumors observed in patients with late-stage cancer (10, 12–14). Therefore, it is conceivable that a strategy that blocks immunosuppression will improve the therapeutic efficacy of anti-OX40 immunotherapy, especially in mice with large established tumors.

TGFβ is an immunosuppressive cytokine that is produced by a variety of tumor types (15–17) as well as by immune cells found within the tumor microenvironment (TME; refs. 18–20), and it has been shown that elevated levels of TGFβ are associated with tumor progression (15, 16). TGFβ protumor functions are mediated through direct action on tumor cells (20) as well as inhibition of immune cells, including monocytes, dendritic cells, natural killer cells, and T cells (19, 20). These include inhibition of antigen presentation, T-cell differentiation, effector/cytotoxic activity of CD8+ T cells, and Th1 cytokine production by CD4+ T cells (19–22). In addition to suppression of effector T cells, TGFβ signaling in peripheral Tregs has also been shown to be important for their expansion, survival, and suppressive function in vivo (23, 24).

Collectively, the previously discussed studies provide a rationale for blocking TGFβ to enhance an antitumor immunity. This can be achieved using neutralizing antibodies (25), fusion proteins (26), or small-molecule TGFβ receptor signaling inhibitors that have demonstrated some efficacy in suppressing primary tumor growth and metastasis in several mouse tumor models (27–30). Unlike antibodies and fusion proteins, whose ability to effectively restore immune function depends on complete neutralization of circulating TGFβ, small-molecule TGFβ receptor inhibitors, which interfere with TGFβ receptor signaling, allow immune cells to retain normal function even in a TGFβ-rich environment. SM16, which is used in this study, is an orally bioavailable small molecule that has demonstrated antitumor efficacy when administered in the diet (27). SM16 binds to the kinase active site of TGFβ type I receptor (ALK5), thus inhibiting phosphorylation of SMAD proteins and downstream signal transduction (28). We hypothesized that using an agonist (αOX40) to boost effector T-cell function in conjunction with an inhibitor of T-cell suppression (SM16) would endow the immune system with the ability to eradicate large established tumors. Indeed, this combination has recently been shown to be effective in eradicating smaller tumors via an immune-dependent mechanism in the poorly immunogenic 4T1 breast carcinoma model (31). Therefore, we wanted to determine if this combination would be effective in curing mice of large established tumors.

Our data demonstrate that SM16 plus αOX40 stimulates a vigorous antitumor response that results in complete and durable tumor regression in mice bearing large (50–120 mm2) well-established MCA205 and CT26 tumors that resulted in tumor-specific immunity. Examination of the tumors in mice receiving αOX40 + SM16 therapy revealed an increase in tumor-infiltrating T cells (TIL) that were phenotypically and functionally distinct from T cells found in tumors from mice treated with the single agents. The combination therapy increased the frequency of OX40-expressing, Ki67+/granzyme B+ CD8+ TILs, pSTAT3-expressing CD4+ and CD8+ TILs, and IFNγ-producing TILs. Although the increased effector function in T cells was to be expected, the increase in pSTAT3 was somewhat unexpected as others have shown that STAT3 signaling in T cells is detrimental to their function, especially in regard to tumor eradication (32). Surprisingly, we found that when STAT3 was deleted in OX40-expressing T cells, the efficacy of this potent combination therapy was significantly reduced. Collectively, these results demonstrate that combining OX40 agonists with an orally bioavailable inhibitor of TGFβ signaling can overcome the immunosuppressive environment in large established tumors, resulting in tumor regression. We also show that this potent immunotherapy combination is mediated in part by STAT3 signaling in OX40-expressing cells.

Mice

Six- to eight-week-old C57BL/6 female mice were purchased from The Jackson Laboratory. C57BL/6 OX40-Cre mice were kindly provided by Dr. Nigel Killeen (UCSF, San Francisco, CA) and were crossed with mice carrying the Rosa26-loxP-STOP-loxP-YFP allele (33) obtained from The Jackson Laboratory (JAX). In the double transgenic OX40-Cre/Rosa-YFP mice, cells that express OX40-driven cre protein will knock out a floxed stop cassette that is juxtaposed between a promoter that is constitutively active in lymphocytes and a gene encoding YFP (34). STAT3flox/flox mice were supplied by Dr. Hua Yu (City of Hope Research Institute, Duarte, CA), and the OX40cre/wt mice were supplied by Dr. Nigel Killeen (UCSF). STAT3-deficient mice were generated by crossing OX40cre/wt × STAT3flox/flox (STAT3−/−) with OX40cre/wt × STAT3wt/wt (STAT3+/+) mice. It should be noted that the OX40cre gene is knocked into the OX40 locus, which only allows for hemizygous expression of OX40 in the crossed progeny described above. We have found that the OX40 agonist therapy is less effective in mice that express hemizygous levels of OX40 (data not shown). All mice were maintained under specific pathogen-free conditions in the Providence Portland Medical Center animal facility in accordance with the Principles of Animal Care (NIH publication no. 85-23, revised 1985). All studies were reviewed and approved by the Institutional Animal Care and Use Committee of the Earle A. Chiles Research Institute.

Tumor cell lines

Murine MCA205 sarcoma cells, 3LL Lewis lung carcinoma cells, and CT26 colon carcinoma cells were propagated in vitro using (complete media) RPMI (Lonza) containing 0.292 ng/mL glutamine (Hyclone Laboratories), 100 U/mL streptomycin (Hyclone), 100 U/mL penicillin (Hyclone), 1× nonessential amino acids (Lonza BioWhittaker), 1 mmol/L sodium pyruvate (Lonza BioWhittaker), and 10 mmol/L HEPES (Sigma). Both tumor cell lines were routinely tested and shown to be free of Mycoplasma contamination.

Reagents

The following antibodies were used for analysis by flow cytometry: CD4 Pacific Blue or PerCP-Cy5.5 (eBioscience; GK1.5), CD8 PerCP-Cy5.5 (ebioscience; 53-6.7), OX40 PE (Biolegend; OX-86), CD25 APC (eBioscience; PC61.5), Ki67 FITC (BD; 35), Foxp3 Pacific Blue (eBioscience; FJK-16s), Granzyme B PE (Invitrogen; GB12), and IFNγ APC (ebioscience; 4S.B3). Live cells were identified using the eFluor 780 viability dye (eBioscience). The OX40 agonist is a rat anti-mouse OX40 antibody (clone OX86). Rat Ig (Sigma) was used as a control for OX86. SM16 was synthesized by Biogen Idec and was incorporated into standard Purina rodent chow (#5001) by Research Diets at a concentration of 0.3 g SM16 per kg of chow (0.03%). A calorie and nutrient-matched diet without SM16 (Purina) was used as the control diet.

Flow cytometry

Cells (1–5 million) in 100 μL of flow wash buffer (FWB, PBS with 0.5% FBS) were incubated with the appropriate antibodies at 4°C for 30 minutes. For intracellular staining, cells were fixed and permeabilized with the Foxp3 Staining Buffer Set (eBioscience) according to the manufacturer's protocol. Intracellular staining was performed at 4°C for 30 minutes with the appropriate antibodies to detect specific intracellular proteins. Cells were analyzed using an LSR II flow cytometer (BectonDickinson) and FlowJo4 software.

In vivo tumor growth and survival experiments

For primary tumor challenge, mice were injected s.c. in the right flank with 4 × 105 MCA205 or CT26 tumor cells. Eleven days (∼50–120 mm2) after tumor inoculation, mice were randomized into the following treatment groups: control diet + rat IgG1 (isotype control for αOX40), control diet + αOX40 (OX86), SM16 diet (0.03% SM16) + rat IgG1, and SM16 + αOX40. Mice were then given i.p. injections of 250 μg of either αOX40 or rat IgG1 on days 2 and 6 after starting the diet. Mice were taken off the control and SM16 diets after 2 weeks and monitored for tumor growth two to three times a week by measuring tumor length (L) and width (W) to determine tumor size [L × W (mm2)]. All mice were sacrificed when the tumors either ulcerated or reached >150 mm2. For rechallenge experiments, mice were injected s.c. with 4 × 105 MCA205 tumor cells in the right flank and 4 × 105 3LL cells in the left flank and were examined for tumor growth in the absence of any additional therapy.

Phenotyping of TILs and splenocytes

Mice were injected s.c. with 4 × 105 MCA205 cells. Once tumors were established for 11 days (∼50–120 mm2), mice were put on SM16 or control diet and administered 250 μg of either αOX40 or Rat IgG1 on days 2 and 6 after starting diet. Seven days after starting feed, tumors and spleens were harvested. Resected tumors were minced with scissors and disaggregated using a triple-enzyme digest cocktail containing 10 mg/mL of collagenase type IV (Worthington Biochemical Corp.), 1 mg/mL hyaluronidase (Sigma-Aldrich), and 200 μg/mL DNAse I (Roche Applied Sciences) in complete media for 30 minutes with agitation at 37°C using a shaker. The tumor digest was then passed through a 70-μm nylon filter, subjected to red blood cell lysis using ACK lysing buffer (Lonza), and then washed and used for analysis. Spleen cells were minced between slides; subjected to red blood cell lysis; and passed through 70-μm nylon filter, washed, and used for analysis. Cells were then incubated with fluorescently labeled antibodies and analyzed by flow cytometry.

In vitro stimulation of isolated immune cells

Half of tumor digest or 1 × 107 spleen cells were resuspended in 1 mL of complete media and stimulated for 1 hour at 37°C with 1× phorbol myristate acetate (PMA)/ionomycin cocktail (eBioscience) in 48-well plates. Brefeldin A (BioLegend) was added to a final concentration of 5 μg/mL and incubated with the cells for an additional 4 hours at 37°C. Cells were then stained for surface markers, fixed and permeabilized, and then evaluated for intracellular IFNγ production as described above.

Statistical analysis

Statistical significance was determined by one-way ANOVA (for comparison among three or more groups), unpaired Student t tests (for comparison between two groups), or Kaplan–Meier survival and log-rank (Mantel–Cox) test (for tumor survival studies) using GraphPad Prism software (GraphPad); P value is either stated in the figure or indicated by *, P < 0.05.

SM16 plus anti-OX40 therapy stimulates regression of large established tumors and prolongs overall survival

αOX40 therapy has been shown to cure mice in several different mouse tumor models if administered shortly after tumor challenge when tumors are either not yet palpable or small (0–50 mm2; refs. 3, 7, 8). However, αOX40 has been less effective at curing large established tumors (>50 mm2), either having no impact or only delaying tumor growth, with very little extension of long-term survival (Fig. 1A and B; refs. 8–11). Examination of the TME of large tumors after OX40 agonist treatment revealed that the αOX40 antibody was able to bind to tumor-infiltrating T cells (data not shown). Yet, despite being able to penetrate large tumors and bind to infiltrating T lymphocytes, anti-OX40 was unable to initiate a therapeutic immune response. The inability of αOX40 to activate an effective antitumor T-cell response may be due to immunosuppressive signals present within the tumor as it grows in size (19, 35–38). Therefore, we hypothesized that using an OX40 agonist antibody in conjunction with a small-molecule inhibitor of the TGFβ type 1 receptor will initiate a potent antitumor immune response with the potential to cure mice harboring large tumors. To test this hypothesis, MCA205 sarcoma cells and CT26 colon carcinoma cells were implanted into mice and allowed to grow until the tumors were approximately 40 to 120 mm2 in size (11 days after implant). Tumor-bearing mice were fed SM16 or control feed for 14 days, during which they were treated with either rat IgG or an αOX40 antibody on days 13 and 17 as depicted in Fig. 1A. The SM16 + anti-OX40 combination therapy caused complete tumor regression in 75% and 45% of mice harboring large preestablished MCA205 and CT26 tumors, respectively (Fig. 1). Tumor-free survival was sustained over 100 days and was significantly lower with single-agent treatment of anti-OX40 or SM16. Tumor regression was not observed in mice that received control IgG. In summary, treatment with an OX40 agonist antibody in conjunction with a TGFβ signaling inhibitor (SM16) provided a therapeutic immune-boosting combination that caused regression of large established tumors in two separate tumor models tested in two different mouse strains.

Figure 1.

Therapeutic benefit in large established tumors of combined agonistic antibody to OX40 and a small-molecule blockade of the TGFβ receptor. A, MCA205 sarcoma cells were injected s.c. on the flank of 6- to 8-week-old C57Bl/6J female mice and allowed to grow for 11 days. Eleven days after tumor inoculation, SM16 feed (0.3 g SM16/kg of feed) or control feed was administered and then stopped 14 days later. On days 13 and 17 after tumor inoculation, 250 μg of anti-OX40 (clone OX86) or isotype control antibody was injected i.p. Tumor growth and survival were monitored for 100 days. B, CT26 colon cancer cells were injected s.c. into 6- to 8-week-old Balb/C mice that were treated as described in A. For both tumor types, survival and tumor growth curves are shown, and statistics were performed on the Kaplan–Meier survival analyses.

Figure 1.

Therapeutic benefit in large established tumors of combined agonistic antibody to OX40 and a small-molecule blockade of the TGFβ receptor. A, MCA205 sarcoma cells were injected s.c. on the flank of 6- to 8-week-old C57Bl/6J female mice and allowed to grow for 11 days. Eleven days after tumor inoculation, SM16 feed (0.3 g SM16/kg of feed) or control feed was administered and then stopped 14 days later. On days 13 and 17 after tumor inoculation, 250 μg of anti-OX40 (clone OX86) or isotype control antibody was injected i.p. Tumor growth and survival were monitored for 100 days. B, CT26 colon cancer cells were injected s.c. into 6- to 8-week-old Balb/C mice that were treated as described in A. For both tumor types, survival and tumor growth curves are shown, and statistics were performed on the Kaplan–Meier survival analyses.

Close modal

SM16/αOX40 therapy increases the frequency of CD4+ and CD8+ T cells in tumors

To understand how SM16/αOX40 mediates tumor regression, TILs were evaluated and compared with peripheral lymphocytes. Lymphocytes from the TME were analyzed from large tumors in mice that were put on the SM16 or control diet, administered two injections of αOX40 or control antibodies (as in the scheme in Fig 1A), and then sacrificed 1 day after the second antibody injection. Excised tumors were subjected to triple-enzyme digest without density gradient centrifugation so as not to enrich for immune cells and thus evaluate the proportion of total cells within the tumor digest that were CD4+ or CD8+ T cells. This analysis revealed an increase in the proportion of cells that were CD4+ and CD8+ in the SM16- and SM16/αOX40-treated mice compared with that of the control and αOX40-treated mice (Fig. 2A and B). No increase in proportion of CD4+ and CD8+ T cells in the spleens was found with any of the treatments. The increase of T cells within the tumors of SM16- and SM16/αOX40-treated mice suggested that the mechanism of tumor regression was immune mediated. However, TGFβ is also known to directly affect tumor growth, independent of immune suppression (20). Therefore, to confirm a role for lymphocytes in SM16-mediated antitumor responses, we performed the SM16 therapy experiment using RAG−/− mice. As shown in Fig. 2C, SM16 was unable to alter the outgrowth of large tumors in mice devoid of lymphocytes in the MCA205 model (log-rank test: P < 0.0001).

Figure 2.

Increased T-cell infiltration in tumor-bearing mice treated with combination therapy. A and B, MCA205 sarcoma cells were injected s.c. on the flank of 6- to 8-week-old C57Bl/6J female mice and allowed to grow for 11 days. Eleven days after tumor inoculation, SM16 feed or control feed was initiated. On days 13 and 17, 250 μg anti-OX40 or isotype control antibody was administered. Tumors and spleens were harvested on day 18, and T cells were assessed for (A) proportion of singlet gate that was CD4+ or CD8+ and (B) proportion of splenocytes that were CD4+ or CD8+. Each dot on all graphs represents results from individual mice from at least three independent experiments. C, RAGKO mice or C57Bl/6 female mice were inoculated with MCA205. On day 11, mice were fed SM16 and taken off the SM16 feed 14 days later (n = 13/group). Both tumor growth and survival curves are shown, and these mice were monitored for 80 days after tumor inoculation. Statistical significance for all graphs was determined by one-way ANOVA analysis. *, P < 0.05.

Figure 2.

Increased T-cell infiltration in tumor-bearing mice treated with combination therapy. A and B, MCA205 sarcoma cells were injected s.c. on the flank of 6- to 8-week-old C57Bl/6J female mice and allowed to grow for 11 days. Eleven days after tumor inoculation, SM16 feed or control feed was initiated. On days 13 and 17, 250 μg anti-OX40 or isotype control antibody was administered. Tumors and spleens were harvested on day 18, and T cells were assessed for (A) proportion of singlet gate that was CD4+ or CD8+ and (B) proportion of splenocytes that were CD4+ or CD8+. Each dot on all graphs represents results from individual mice from at least three independent experiments. C, RAGKO mice or C57Bl/6 female mice were inoculated with MCA205. On day 11, mice were fed SM16 and taken off the SM16 feed 14 days later (n = 13/group). Both tumor growth and survival curves are shown, and these mice were monitored for 80 days after tumor inoculation. Statistical significance for all graphs was determined by one-way ANOVA analysis. *, P < 0.05.

Close modal

SM16/αOX40 combination therapy affects the phenotype and effector function of TILs

We further evaluated the TILs to determine if there was simply an increase of T cells in the tumor or if these T cells were phenotypically and functionally distinct between treatment groups. Because IFNγ is a known beneficial antitumor cytokine (39–43), we evaluated the ability of TILs from the different treatment groups to produce IFNγ, directly ex vivo. When the cells were stimulated with PMA/ionomycin, we found an increase in the frequency of CD4+ and CD8+ TILs capable of producing IFNγ from mice treated with SM16/αOX40 compared with that from mice in the other treatment groups (Fig. 3A). A significant increase in IFNγ production was also observed in splenic CD4+ T cells from the SM16/αOX40 treatment group (Fig. 3A).

Figure 3.

Effector function and OX40 expression are increased with SM16/anti-OX40 combination therapy. Tumor-bearing mice were treated as in Fig. 2. A, TILs were stimulated with PMA/ionomycin, after which the cells were treated with brefeldin A and evaluated for intracellular IFNγ. Graphs show proportion of CD4+ and CD8+ cells that were IFNγ+. B, representative dot plots of CD8+ cells isolated from spleens and tumors for expression of Ki67 and granzyme B and Ki67/granzyme B double-positive CD8+ T cells (right) for an individual experiment (each dot represents an individual mouse). C, OX40cre-YFP reporter tumor-bearing mice were treated as in A and B, and representative flow cytometry plots of TILs for expression of YFP in CD4+ and CD8+ T-cell populations are shown in the left plots. A summary of the proportion of CD8+ and CD4+ cells from the tumor that were YFP+ is shown in the right plots, which represent a compilation of three independent experiments; the wide horizontal bar indicates mean ± SD. Each dot on all graphs represents results from an individual mouse. Statistical significance for all graphs was determined by one-way ANOVA analysis. *, P < 0.05.

Figure 3.

Effector function and OX40 expression are increased with SM16/anti-OX40 combination therapy. Tumor-bearing mice were treated as in Fig. 2. A, TILs were stimulated with PMA/ionomycin, after which the cells were treated with brefeldin A and evaluated for intracellular IFNγ. Graphs show proportion of CD4+ and CD8+ cells that were IFNγ+. B, representative dot plots of CD8+ cells isolated from spleens and tumors for expression of Ki67 and granzyme B and Ki67/granzyme B double-positive CD8+ T cells (right) for an individual experiment (each dot represents an individual mouse). C, OX40cre-YFP reporter tumor-bearing mice were treated as in A and B, and representative flow cytometry plots of TILs for expression of YFP in CD4+ and CD8+ T-cell populations are shown in the left plots. A summary of the proportion of CD8+ and CD4+ cells from the tumor that were YFP+ is shown in the right plots, which represent a compilation of three independent experiments; the wide horizontal bar indicates mean ± SD. Each dot on all graphs represents results from an individual mouse. Statistical significance for all graphs was determined by one-way ANOVA analysis. *, P < 0.05.

Close modal

To further characterize these lymphoctyes, we determined the proportion of CD8+ cells that were actively undergoing proliferation as assessed by Ki67 expression. Although we found no difference in the proportion of CD8+ cells proliferating in tumors of SM16/αOX40-treated mice, there was a significant increase in proliferating (Ki67+)/granzyme B+ CD8+ T cells in both the tumors and the spleens (Fig. 3B). The increase in proliferating granzyme B+ CD8+ T cells suggested that there might be an increase in CD8+ T-cell–mediated killing of tumors in mice injected with the combination therapy. To determine whether the SM16/αOX40 dual treatment was reliant on granule-mediated killing of tumors, we tested the dual treatment in tumor-bearing perforin knockout (KO) mice. We found that although 15 of 18 wild-type (WT) tumor-bearing mice receiving the dual treatment survived, none of the perforin KO tumor-bearing mice treated with SM16/αOX40 survived (0/17; Fig. 4C). We next explored the contribution of CD4+ and/or CD8+ T cells to the therapeutic efficacy of the anti-OX40/SM16 combination therapy. Previous studies have shown that both CD4+ and CD8+ T cells are required for the therapeutic efficacy of OX40 agonists when delivered in mice harboring smaller tumors (5). However, Fig. 4A and B show that only the CD8+ T cells are necessary for the dual therapy to be effective. Depletion of CD4+ T cells had no effect on the therapeutic efficacy of the combination therapy; however, there were some autoimmune symptoms associated with depleting the CD4+ T cells (data not shown), which was most likely due to depletion of Tregs in mice receiving this potent combination immune therapy. Together, these data suggest that the dual therapy is highly dependent on CD8+ T cells and that increased IFNγ production and cytolytic granule activity in the CD8+ T cells likely contribute to the potent therapeutic effect.

Figure 4.

Tumor regression observed via the combination therapy is dependent on CD8+ T cells. MCA205 tumor–bearing mice were treated as in Fig. 1 and were administered CD8- or CD4-depleting antibodies or an isotype antibody on days −1, +4, +7, +14, and 1 week after SM16 and anti-OX40 administration. Tumor growth (A) and survival (B) were monitored for 40 days. C, perforin KO C57/Bl/6 or C57Bl/6 WT mice were treated as in Fig. 1, and tumor growth and survival were monitored for 80 days.

Figure 4.

Tumor regression observed via the combination therapy is dependent on CD8+ T cells. MCA205 tumor–bearing mice were treated as in Fig. 1 and were administered CD8- or CD4-depleting antibodies or an isotype antibody on days −1, +4, +7, +14, and 1 week after SM16 and anti-OX40 administration. Tumor growth (A) and survival (B) were monitored for 40 days. C, perforin KO C57/Bl/6 or C57Bl/6 WT mice were treated as in Fig. 1, and tumor growth and survival were monitored for 80 days.

Close modal

Previous studies have shown that TGFβ signaling can promote Treg proliferation and survival (23, 24). Therefore, one would expect to observe a decrease in the frequency of Tregs when TGFβ signaling is blocked. We found that in mice from the SM16-treated groups, there was an increase in the proportion of CD4+ cells that were FOXP3+ in the spleens; however, no statistical difference was observed in the tumor (data not shown). We also found an increase in the proportion of the Tregs undergoing proliferation in the spleens of SM16/αOX40-treated mice. The expansion of peripheral Tregs in the absence of TGFβ signaling has also been observed in other tumor models (18, 31), and in our model, the increase in peripheral Tregs may help quell off-target inflammation while the mice are receiving this potent dual therapy.

SM16/αOX40 combination therapy increases the proportion of OX40-expressing TILs

OX40-expressing tumor-infiltrating T cells are the most likely target for αOX40 therapy. Therefore, we wanted to determine if there was an increase in the proportion of T cells that express or previously expressed OX40 in the SM16- and SM16/αOX40-treated mice. Because our anti-OX40 detection antibody is the same as the therapeutic antibody and OX40 expression on CD8+ TILs is low and hard to detect (data not shown), we took advantage of the OX40Cre/ROSAYFP reporter mouse system that produces the YFP protein in cells that express or had previously expressed OX40 (33). Using these mice, we found a significant increase in the proportion of CD8+ TILs that were YFP+ in both the SM16 and SM16/αOX40 groups (Fig. 3C). There was no statistically significant difference in the proportion of tumor-infiltrating CD4+ that were YFP+ because the majority (∼90%) of them were YFP+ in all groups. This increase in CD8+ T cells that express OX40 in the tumor is most likely relevant to the mechanism of tumor destruction because previous work has shown that direct engagement of OX40 by CD8+ T cells is necessary to help drive a potent antitumor immune response by OX40 agonists (10, 11, 44). We also found that the YFP+ T cells within the tumor were significantly increased for both IFNγ and granzyme B, and this observation was consistent for all treatment groups, although the trend showed a greater increase in mice treated with the SM16/αOX40 dual therapy.

SM16/αOX40-cured mice develop long-term antitumor immunity

We wanted to determine if mice cured of their tumors following SM16 or SM16/αOX40 therapy developed long-term immunity. To achieve this goal, tumor-free mice in both treatment groups were challenged >60 days after primary tumor injection with viable MCA205 tumor cells. None of the 7 mice from the SM16/αOX40 treatment group that were cured developed tumors, whereas a fraction (2/7) of the SM16-treated mice developed tumors and, as expected, all (7/7) of the naïve mice developed tumors (data not shown). In contrast, neither the SM16- nor SM16/αOX40-cured mice were protected from challenge with 3LL tumor cells (data not shown). These results indicate that SM16/αOX40-mediated tumor regression resulted in a tumor-specific immunologic memory that protects against tumor rechallenge.

Conditional ablation of STAT3 in OX40-expressing cells reduces overall survival of mice receiving SM16/anti-OX40 combination therapy

In trying to understand the mechanism(s) underlying the efficient therapeutic activity of the SM16/anti-OX40 therapy, we unexpectedly found that STAT3 phosphorylation was increased in tumor-infiltrating CD8+ and CD4+ T cells in SM16/anti-OX40–treated animals compared with that in the other treatment groups (Fig. 5A). Previously, it had been published that pSTAT3-expressing tumor-specific T cells were associated with a dysfunctional T-cell response leading to decreased antitumor efficacy (32). Hence, to better understand the role that STAT3 plays in this combination therapy, we crossed STAT3flox/flox mice with OX40cre mice and assessed the antitumor efficacy of the combination treatment. We found that conditional deletion of the STAT3 gene reduced the efficacy of the combination therapy as we observed a significant decrease in overall survival and tumor shrinkage in the STAT3 KO mice (Fig. 5B). We also observed that the STAT3-deficient, tumor-bearing mice receiving the dual treatment had decreased frequencies of proliferating, granzyme B+ CD8+ T cells in the tumors as well as decreased IFNγ production by CD8+ TILs (Fig. 5C and D). We also found a significant increase in the percentage and proliferation of Tregs in the tumors of the STAT3-deficient mice. These data show for the first time that STAT3 function in T cells plays a positive role in T-cell–mediated rejection of tumors, a concept that opposes the current paradigm that STAT3 function is detrimental to T-cell function in tumor-bearing mice. This observation is most likely context dependent, and in this study, in which mice were treated with a potent immune-stimulatory therapy, STAT3 function plays a positive role in immune-mediated clearance of tumors.

Figure 5.

STAT3 signaling is essential for full therapeutic potency of the anti-OX40/SM16 combination immunotherapy. A, mice with established tumors were treated with SM16 and anti-OX40, after which tumors were resected and tumor-infiltrating cells were assessed ex vivo for phosphorylated STAT3 (pSTAT3) expression by intracellular staining as in Fig. 2. Mean fluorescence intensity of pSTAT3 was quantified in CD8+ and CD4+ TILs. B, OX40cre/wt × STAT3flox/flox (STAT3−/−) and OX40cre/wt × STAT3wt/wt mice were inoculated with MCA205 tumor cells and treated as in Fig. 1 and monitored for tumor growth and overall survival. C, frequency of Ki67+GrzB+CD8+ T cells and CD4+ T cells. On day 18, splenocytes and TILs were isolated and assessed for Ki67 and GrzB by flow cytometry. D, CD4+ and CD8+ T cells were sorted from TILs of SM16- and anti-OX40–treated OX40cre/wt × STAT3flx.flx and OX40cre/wt × STAT3wt.wt and restimulated for 5 hours with PMA/ionomycin, from which the supernatant was collected and analyzed by ELISA for IFNγ production. E, TILs from treated mice were analyzed for FOXP3+/CD25+CD4+ T-regulatory cell percentages and percent proliferating based on anti-Ki67 staining. All statistics were performed by the Student t test analysis.

Figure 5.

STAT3 signaling is essential for full therapeutic potency of the anti-OX40/SM16 combination immunotherapy. A, mice with established tumors were treated with SM16 and anti-OX40, after which tumors were resected and tumor-infiltrating cells were assessed ex vivo for phosphorylated STAT3 (pSTAT3) expression by intracellular staining as in Fig. 2. Mean fluorescence intensity of pSTAT3 was quantified in CD8+ and CD4+ TILs. B, OX40cre/wt × STAT3flox/flox (STAT3−/−) and OX40cre/wt × STAT3wt/wt mice were inoculated with MCA205 tumor cells and treated as in Fig. 1 and monitored for tumor growth and overall survival. C, frequency of Ki67+GrzB+CD8+ T cells and CD4+ T cells. On day 18, splenocytes and TILs were isolated and assessed for Ki67 and GrzB by flow cytometry. D, CD4+ and CD8+ T cells were sorted from TILs of SM16- and anti-OX40–treated OX40cre/wt × STAT3flx.flx and OX40cre/wt × STAT3wt.wt and restimulated for 5 hours with PMA/ionomycin, from which the supernatant was collected and analyzed by ELISA for IFNγ production. E, TILs from treated mice were analyzed for FOXP3+/CD25+CD4+ T-regulatory cell percentages and percent proliferating based on anti-Ki67 staining. All statistics were performed by the Student t test analysis.

Close modal

It has been reported that the microenvironment of large tumors is immunosuppressive due to the presence of inhibitory cytokines and other cellular and soluble factors (19, 35–38). Therefore, a successful tumor immunotherapy may require both an agent to boost effector T-cell function and an inhibitor of tumor-mediated T-cell suppression (8–11). In this study, we found that αOX40 therapy alone was ineffective at treating large established tumors, but was capable of causing complete regression of large tumors if given in conjunction with a TGFβ receptor inhibitor (SM16). The dual treatment resulted in long-term survival in approximately 85% of mice compared with approximately 20% of mice treated with SM16 alone in the MCA-205 model (Fig. 1) and 45% survival of the αOX40/SM16-treated tumor-bearing mice compared with 5% of mice treated with αOX40 alone in the CT26 model. Furthermore, complete tumor regression was accompanied by the host's ability to resist tumor rechallenge in a tumor-specific fashion in mice cured by the dual therapy. Examination of the TME showed that the T-cell composition was significantly different in tumors obtained from the αOX40/SM16-treated mice compared with that with either treatment alone. Cytokine analyses revealed that a greater percentage of both CD4+ and CD8+ T cells from αOX40/SM16-treated mice were capable of producing IFNγ ex vivo (Fig. 3A). The finding that the efficacy of the anti-OX40/SM16 combination therapy is reliant on CD8+ T cells and not CD4+ T cells led us to focus our efforts in understanding the differences in CD8+ T cells in mice treated with the combination versus the monotherapies. Phenotypic analyses revealed a significant increase in the proportion of CD8+ T cells expressing granzyme B (GrzB) in the tumors of αOX40/SM16-treated mice (∼60%) when compared with the SM16- (∼30%) and αOX40-treated mice (∼30%; data not shown). Interestingly, a large proportion of these CD8+GrzB+ cells coexpressed the proliferation marker Ki67 in the αOX40/SM16-treated mice (Fig. 3B). To our knowledge, these CD8+ GrzB+ Ki67+ T cells represent a novel subset of terminally differentiated proliferating T cells within the tumors. Others have described granzyme B+ CD8 T cells as end-stage nonproliferating cells involved with tumor-cell killing (45); however, the CD8+ T cells in the dual therapy–treated group seem to have a novel phenotype potentially capable of multiple functions. Previous studies have shown that OX40 signaling increases CD8+ T-cell proliferation, IFNγ production, and granzyme B expression (46). Conversely, TGFβ signaling in CD8+ T cells can inhibit all of these functions (22). However, it is clear that single treatment with either SM16 or αOX40 was unable to significantly increase the proportion of proliferating/effector CD8+ T cells within the tumor. These data show that promoting OX40 signaling while simultaneously blocking TGFβ signaling has an additive/synergistic effect on tumor-infiltrating CD8+ T-cell differentiation and proliferation, which most likely led to the enhancement of tumor-cell killing. The enhanced efficacy of the dual therapy may in part be explained by the ability of SM16 to increase the proportion of CD8+ T cells that express(ed) OX40, hence increasing the likelihood of increased OX40 signaling within the TME (Fig. 4C).

Because OX40 and TGFβ signaling have been shown to modulate Treg proliferation, survival, and function (4, 5, 23, 24), we expected to find differences in the frequency of tumor-infiltrating Tregs. Surprisingly, we found no statistical change in the proportion of CD4+ T cells that expressed FOXP3+ within tumors of the different treatment groups in WT mice. However, we did observe a statistically significant increase in the percentage of tumor-isolated Tregs (FoxP3+CD25+ CD4 cells) in the STAT3 KO mice compared with that in WT mice treated with the dual therapy. There was also an increase in Treg proliferation in the STAT3 KO mice compared with WT mice. Hence, there was most likely a STAT3-dependent cytokine signaling event that was increased during dual therapy, which can influence Treg percentages within the TME. It is interesting that despite the high frequency of CD4+ cells that are FoxP3+ (sometimes >30% of CD4 cells) within the tumors of αOX40/SM16-treated WT mice, CD8+ T cells appear capable of proliferating and differentiating into effector CD8+GrzB+ cells. It currently remains unknown if αOX40/SM16 treatment tempers Treg function and/or alters CD8+ T-cell susceptibility to Treg suppression, but these types of questions will be addressed in future studies.

We found that combining αOX40 antibodies with a TGFβ receptor signaling inhibitor (SM16) resulted in complete and durable regression of large established tumors. These responses were accompanied by changes in the T-cell frequencies and phenotype within these tumors and the ability to resist tumor rechallenge. Interestingly, we observed an increase in STAT3 phosphorylation in both CD8+ and CD4+ T cells infiltrating tumors of mice receiving the combination therapy compared with tumor-bearing mice receiving either agent alone. Genetic ablation of the STAT3 gene in OX40-expressing T cells significantly reduced tumor cures in the dual therapy–treated mice. Characterization of TILs revealed an increase of proliferating effector CD8+ T cells expressing granzyme B in tumors of WT mice that received combination treatment, and these populations were significantly reduced in STAT3 KO mice. These findings are opposite to the prevailing paradigm that STAT3 activation in T cells is deleterious to their function within tumor-bearing hosts (32). Instead, STAT3 activates a beneficial pathway(s) in T cells leading to tumor regression during anti-OX40 immunotherapy when the TGFβ signaling pathway is blocked. These findings justify rethinking the current dogma that STAT3 expressed in T cells decreases their function leading to increased tumor progression and suggest that blocking STAT3 signaling could have detrimental effects, especially when potent immune-based cancer therapies are delivered to cancer-bearing hosts.

Recent articles about STAT3 signaling in T cells have shown a critical/positive role in regulating survival of CD8 and CD4 T cells (47, 48). Hence, recent evidence suggests that STAT3 is important for proinflammatory functions of T cells, especially in the setting of viral infections (48). One report showed that STAT3-deficient T cells had an increased proliferative capacity, but were more prone to activation-induced cell death (AICD). This increase in proliferation and sensitivity to AICD in STAT3 KO T cells correlated with a decrease in antiapoptotic proteins, including OX40, and an increase in proapoptotic proteins (48). In the context of Herpes simplex virus infection, STAT3 deficiency led to a decrease in the percentage of CD8 T cells producing viral-specific IFNγ. In humans, a dominant-negative mutation in STAT3 leads to a significant decrease in central memory CD4 and CD8 T cells, and patients carrying this defect are more susceptible to bacterial and fungal infections (47). Therefore, STAT3 is important for the survival of T cells, and the survival could be mediated through gamma-chain cytokines, which are known to signal through STAT3 and STAT5. We hypothesize that in the context of increased inflammation, such as viral Toll-like receptor signaling or SM16/αOX40 stimulation, STAT3 signaling increases T-cell survival and enhances antitumor immunity, and these results are quite opposite to previously published reports (32). Based on these differences, we favor a model in which STAT3 signaling could have opposing roles in T-cell function, depending on the environment the T cell encounters upon T-cell receptor engagement.

A.D. Weinberg is president/CSO at Agonox, Inc., reports receiving a commercial research grant from MedImmune, and has ownership interest in OX40 patents. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T.A. Triplett, C.G. Tucker, L.E. Ling, E.T. Akporiaye, A.D. Weinberg

Development of methodology: T.A. Triplett, C.G. Tucker, L.E. Ling, E.T. Akporiaye

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T.A. Triplett, C.G. Tucker, Z. Alderman, L. Sun, E.T. Akporiaye

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T.A. Triplett, C.G. Tucker, Z. Alderman, E.T. Akporiaye

Writing, review, and/or revision of the manuscript: T.A. Triplett, C.G. Tucker, E.T. Akporiaye, A.D. Weinberg

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L. Sun

Study supervision: E.T. Akporiaye, A.D. Weinberg

1.
Fujita
T
,
Ukyo
N
,
Hori
T
,
Uchiyama
T
. 
Functional characterization of OX40 expressed on human CD8+ T cells
.
Immunol Lett
2006
;
106
:
27
33
.
2.
Gramaglia
I
,
Weinberg
AD
,
Lemon
M
,
Croft
M
. 
Ox-40 ligand: a potent costimulatory molecule for sustaining primary CD4 T cell responses
.
J Immunol
1998
;
161
:
6510
7
.
3.
Weinberg
AD
,
Rivera
MM
,
Prell
R
,
Morris
A
,
Ramstad
T
,
Vetto
JT
, et al
Engagement of the OX-40 receptor in vivo enhances antitumor immunity
.
J Immunol
2000
;
164
:
2160
9
.
4.
Croft
M
. 
Control of immunity by the TNFR-related molecule OX40 (CD134)
.
Annu Rev Immunol
2010
;
28
:
57
78
.
5.
Redmond
WL
,
Ruby
CE
,
Weinberg
AD
. 
The role of OX40-mediated co-stimulation in T-cell activation and survival
.
Crit Rev Immunol
2009
;
29
:
187
201
.
6.
Curti
BD
,
Kovacsovics-Bankowski
M
,
Morris
N
,
Walker
E
,
Chisholm
L
,
Floyd
K
, et al
OX40 is a potent immune-stimulating target in late-stage cancer patients
.
Cancer Res
2013
;
73
:
7189
98
.
7.
Morris
A
,
Vetto
JT
,
Ramstad
T
,
Funatake
CJ
,
Choolun
E
,
Entwisle
C
, et al
Induction of anti-mammary cancer immunity by engaging the OX-40 receptor in vivo
.
Breast Cancer Res Treat
2001
;
67
:
71
80
.
8.
Song
A
,
Song
J
,
Tang
X
,
Croft
M
. 
Cooperation between CD4 and CD8 T cells for anti-tumor activity is enhanced by OX40 signals
.
Eur J Immunol
2007
;
37
:
1224
32
.
9.
Gough
MJ
,
Crittenden
MR
,
Sarff
M
,
Pang
P
,
Seung
SK
,
Vetto
JT
, et al
Adjuvant therapy with agonistic antibodies to CD134 (OX40) increases local control after surgical or radiation therapy of cancer in mice
.
J Immunother
2010
;
33
:
798
809
.
10.
Gough
MJ
,
Ruby
CE
,
Redmond
WL
,
Dhungel
B
,
Brown
A
,
Weinberg
AD
. 
OX40 agonist therapy enhances CD8 infiltration and decreases immune suppression in the tumor
.
Cancer Res
2008
;
68
:
5206
15
.
11.
Redmond
WL
,
Gough
MJ
,
Weinberg
AD
. 
Ligation of the OX40 co-stimulatory receptor reverses self-Ag and tumor-induced CD8 T-cell anergy in vivo
.
Eur J Immunol
2009
;
39
:
2184
94
.
12.
Berendt
MJ
,
North
RJ
: 
T-cell-mediated suppression of anti-tumor immunity. An explanation for progressive growth of an immunogenic tumor
.
J Exp Med
1980
;
151
:
69
80
.
13.
Bursuker
I
,
North
RJ
. 
Generation and decay of the immune response to a progressive fibrosarcoma. II. Failure to demonstrate postexcision immunity after the onset of T cell-mediated suppression of immunity
.
J Exp Med
1984
;
159
:
1312
21
.
14.
North
RJ
,
Bursuker
I
. 
Generation and decay of the immune response to a progressive fibrosarcoma. I. Ly-1+2- suppressor T cells down-regulate the generation of Ly-1-2+ effector T cells
.
J Exp Med
1984
;
159
:
1295
311
.
15.
Gorsch
SM
,
Memoli
VA
,
Stukel
TA
,
Gold
LI
,
Arrick
BA
. 
Immunohistochemical staining for transforming growth factor beta 1 associates with disease progression in human breast cancer
.
Cancer Res
1992
;
52
:
6949
52
.
16.
Robson
H
,
Anderson
E
,
James
RD
,
Schofield
PF
: 
Transforming growth factor beta 1 expression in human colorectal tumours: an independent prognostic marker in a subgroup of poor prognosis patients
.
Br J Cancer
1996
;
74
:
753
8
.
17.
Rodeck
U
,
Bossler
A
,
Graeven
U
,
Fox
FE
,
Nowell
PC
,
Knabbe
C
, et al
Transforming growth factor beta production and responsiveness in normal human melanocytes and melanoma cells
.
Cancer Res
1994
;
54
:
575
81
.
18.
Donkor
MK
,
Sarkar
A
,
Savage
PA
,
Franklin
RA
,
Johnson
LK
,
Jungbluth
AA
, et al
T cell surveillance of oncogene-induced prostate cancer is impeded by T cell-derived TGF-beta1 cytokine
.
Immunity
2011;
35
:
123
34
.
19.
Flavell
RA
,
Sanjabi
S
,
Wrzesinski
SH
,
Licona-Limon
P
: 
The polarization of immune cells in the tumour environment by TGFbeta
.
Nat Rev Immunol
2010
;
10
:
554
67
.
20.
Massague
J
: 
TGFbeta in cancer
.
Cell
2008
;
134
:
215
30
.
21.
Gorelik
L
,
Constant
S
,
Flavell
RA
. 
Mechanism of transforming growth factor beta-induced inhibition of T helper type 1 differentiation
.
J Exp Med
2002
;
195
:
1499
505
.
22.
Thomas
DA
,
Massague
J
. 
TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance
.
Cancer Cell
2005
;
8
:
369
80
.
23.
Huber
S
,
Schramm
C
,
Lehr
HA
,
Mann
A
,
Schmitt
S
,
Becker
C
, et al
Cutting edge: TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells
.
J Immunol
2004
;
173
:
6526
31
.
24.
Li
MO
,
Sanjabi
S
,
Flavell
RA
. 
Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms
.
Immunity
2006
;
25
:
455
71
.
25.
Nam
JS
,
Terabe
M
,
Mamura
M
,
Kang
MJ
,
Chae
H
,
Stuelten
C
, et al
An anti-transforming growth factor beta antibody suppresses metastasis via cooperative effects on multiple cell compartments
.
Cancer Res
2008
;
68
:
3835
43
.
26.
Muraoka
RS
,
Dumont
N
,
Ritter
CA
,
Dugger
TC
,
Brantley
DM
,
Chen
J
, et al
Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases
.
J Clin Invest
2002
;
109
:
1551
9
.
27.
Rausch
MP
,
Hahn
T
,
Ramanathapuram
L
,
Bradley-Dunlop
D
,
Mahadevan
D
,
Mercado-Pimentel
ME
, et al
An orally active small molecule TGF-beta receptor I antagonist inhibits the growth of metastatic murine breast cancer
.
Anticancer Res
2009
;
29
:
2099
109
.
28.
Suzuki
E
,
Kim
S
,
Cheung
HK
,
Corbley
MJ
,
Zhang
X
,
Sun
L
, et al
A novel small-molecule inhibitor of transforming growth factor beta type I receptor kinase (SM16) inhibits murine mesothelioma tumor growth in vivo and prevents tumor recurrence after surgical resection
.
Cancer Res
2007
;
67
:
2351
9
.
29.
Uhl
M
,
Aulwurm
S
,
Wischhusen
J
,
Weiler
M
,
Ma
JY
,
Almirez
R
, et al
SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo
.
Cancer Res
2004
;
64
:
7954
61
.
30.
Wallace
A
,
Kapoor
V
,
Sun
J
,
Mrass
P
,
Weninger
W
,
Heitjan
DF
, et al
Transforming growth factor-beta receptor blockade augments the effectiveness of adoptive T-cell therapy of established solid cancers
.
Clin Cancer Res
2008
;
14
:
3966
74
.
31.
Garrison
K
,
Hahn
T
,
Lee
WC
,
Ling
LE
,
Weinberg
AD
,
Akporiaye
ET
. 
The small molecule TGF-beta signaling inhibitor SM16 synergizes with agonistic OX40 antibody to suppress established mammary tumors and reduce spontaneous metastasis
.
Cancer Immunol Immunother
2012
;
61
:
511
21
.
32.
Yu
H
,
Pardoll
D
,
Jove
R
: 
STATs in cancer inflammation and immunity: a leading role for STAT3
.
Nat Rev Cancer
2009
;
9
:
798
809
.
33.
Klinger
M
,
Kim
JK
,
Chmura
SA
,
Barczak
A
,
Erle
DJ
,
Killeen
N
. 
Thymic OX40 expression discriminates cells undergoing strong responses to selection ligands
.
J Immunol
2009
;
182
:
4581
9
.
34.
Srinivas
S
,
Watanabe
T
,
Lin
CS
,
William
CM
,
Tanabe
Y
,
Jessell
TM
, et al
Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus
.
BMC Dev Biol
2001
;
1
:
4
.
35.
Coussens
LM
,
Werb
Z
. 
Inflammation and cancer
.
Nature
2002
;
420
:
860
7
.
36.
Frey
AB
,
Monu
N
. 
Signaling defects in anti-tumor T cells
.
Immunol Rev
2008
;
222
:
192
205
.
37.
Luscher
U
,
Filgueira
L
,
Juretic
A
,
Zuber
M
,
Luscher
NJ
,
Heberer
M
, et al
The pattern of cytokine gene expression in freshly excised human metastatic melanoma suggests a state of reversible anergy of tumor-infiltrating lymphocytes
.
Int J Cancer
1994
;
57
:
612
9
.
38.
Ohta
A
,
Gorelik
E
,
Prasad
SJ
,
Ronchese
F
,
Lukashev
D
,
Wong
MK
, et al
A2A adenosine receptor protects tumors from antitumor T cells
.
Proc Natl Acad Sci U S A
2006
;
103
:
13132
7
.
39.
Dighe
AS
,
Richards
E
,
Old
LJ
,
Schreiber
RD
: 
Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors
.
Immunity
1994
;
1
:
447
56
.
40.
Kaplan
DH
,
Shankaran
V
,
Dighe
AS
,
Stockert
E
,
Aguet
M
,
Old
LJ
, et al
Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice
.
Proc Natl Acad Sci U S A
1998
;
95
:
7556
61
.
41.
Shankaran
V
,
Ikeda
H
,
Bruce
AT
,
White
JM
,
Swanson
PE
,
Old
LJ
, et al
IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity
.
Nature
2001
;
410
:
1107
11
.
42.
Street
SE
,
Cretney
E
,
Smyth
MJ
. 
Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis
.
Blood
2001
;
97
:
192
7
.
43.
Street
SE
,
Trapani
JA
,
MacGregor
D
,
Smyth
MJ
. 
Suppression of lymphoma and epithelial malignancies effected by interferon gamma
.
J Exp Med
2002
;
196
:
129
34
.
44.
Redmond
WL
,
Gough
MJ
,
Charbonneau
B
,
Ratliff
TL
,
Weinberg
AD
. 
Defects in the acquisition of CD8 T cell effector function after priming with tumor or soluble antigen can be overcome by the addition of an OX40 agonist
.
J Immunol
2007
;
179
:
7244
53
.
45.
Gattinoni
L
,
Klebanoff
CA
,
Palmer
DC
,
Wrzesinski
C
,
Kerstann
K
,
Yu
Z
, et al
Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells
.
J Clin Invest
2005
;
115
:
1616
26
.
46.
Lee
SW
,
Park
Y
,
Song
A
,
Cheroutre
H
,
Kwon
BS
,
Croft
M
. 
Functional dichotomy between OX40 and 4-1BB in modulating effector CD8 T cell responses
.
J Immunol
2006
;
177
:
4464
72
.
47.
Siegel
AM
,
Heimall
J
,
Freeman
AF
,
Hsu
AP
,
Brittain
E
,
Brenchley
JM
, et al
A critical role for STAT3 transcription factor signaling in the development and maintenance of human T cell memory
.
Immunity
2011
;
35
:
806
18
.
48.
Yu
CR
,
Dambuza
IM
,
Lee
YJ
,
Frank
GM
,
Egwuagu
CE
. 
STAT3 regulates proliferation and survival of CD8+ T cells: enhances effector responses to HSV-1 infection, and inhibits IL-10+ regulatory CD8+ T cells in autoimmune uveitis
.
Mediators Inflamm
2013
;
2013
:
359674
.