Although an immune response to tumors may be generated using vaccines, so far, this approach has only shown minimal clinical success. This is attributed to the tendency of cancer to escape immune surveillance via multiple immune suppressive mechanisms. Successful cancer immunotherapy requires targeting these inhibitory mechanisms along with enhancement of antigen-specific immune responses to promote sustained tumor-specific immunity. Here, we evaluated the effect of indoximod, an inhibitor of the immunosuppressive indoleamine-(2,3)-dioxygenase (IDO) pathway, on antitumor efficacy of anti-OX40 agonist in the context of vaccine in the IDO TC-1 tumor model. We demonstrate that although the addition of anti-OX40 to the vaccine moderately enhances therapeutic efficacy, incorporation of indoximod into this treatment leads to enhanced tumor regression and cure of established tumors in 60% of treated mice. We show that the mechanisms by which the IDO inhibitor leads to this therapeutic potency include (i) an increment of vaccine-induced tumor-infiltrating effector T cells that is facilitated by anti-OX40 and (ii) a decrease of IDO enzyme activity produced by nontumor cells within the tumor microenvironment that results in enhancement of the specificity and the functionality of vaccine-induced effector T cells. Our findings suggest a translatable strategy to enhance the overall efficacy of cancer immunotherapy. Cancer Immunol Res; 6(2); 201–8. ©2018 AACR.

Improved cancer immunotherapy strategies would both evoke an immune response and resist tumor-driven immunosuppression. Successful immunotherapy depends on breaking tumor-mediated immune tolerance such that the immune system can be modulated to deliver T cells that could recognize and eradicate tumors. Induction of immune response against tumors is an efficient method to educate T cells with tumor antigens; however, suppressive mechanisms within the tumor microenvironment weaken the effector immune response. Thus, mitigating the immunosuppression while concurrently augmenting vaccine-induced antigen-specific T cells could improve the overall potency of cancer immunotherapy.

Favorable outcomes follow increased number of tumor-specific effector T cells within the tumor environment (1–3). To generate strong and long-lasting anti-cancer immune response upon antigen recognition, an engagement of secondary costimulatory receptors is often employed in combinational immune therapeutic studies (4–8).

A member of the tumor necrosis factor receptor (TNFR) superfamily, OX40 (CD134) is a costimulatory molecule (9) that is currently in clinical trials (10). The OX40 receptor is inducible on activated T cells (11–15) and its engagement with cognate ligand or agonistic antibody enhances T-cell proliferation, cytokine production, survival, and memory development (16, 17). In animal models, anti-OX40 has shown favorable antitumor activities and in autoimmune models increases the severity of disease (18–23).

In addition to augmenting the effector arm of the immune system, extensive studies have been devoted to immune tolerance and tumor-mediated immunosuppressive mechanisms, which render effector T cells ineffective in clearing malignant cells (24–26). Tumor-mediated immune suppression includes inhibitory mechanisms elicited by tumor cells directly (coinhibitory receptor/ligand interactions, secretion of inhibitory cytokines and other molecules) or indirectly (mechanisms based on tumor-recruited suppressive immune cells). We believe that targeting the indirect mechanisms would have wide application and that such a strategy could be used for immune therapy of a broad spectrum of tumors, regardless of their expression of specific molecules.

One of the protective mechanisms is production of indoleamine-(2,3)-dioxygenase (IDO). IDO is produced not only by some tumors but also by inhibitory immune cells such as suppressive dendritic cells and monocyte-derived macrophages (27, 28). In fact, IDO expression within the tumor microenvironment and tumor draining lymph nodes correlates with poor cancer prognosis (29–32). IDO enzymatic activity potentiates tryptophan degradation, causing cell-cycle arrest and induction of anergy in effector T cells. Toxic tryptophan catabolites may lead to apoptosis of T cells and induction of regulatory T cells (Treg; refs. 33–38). Accordingly, inhibition of the IDO pathway increases antitumor responses in murine models (27, 33, 38–40). One of the relatively well-characterized IDO pathway inhibitors is indoximod (1-methyl-D-tryptophan; refs. 27, 39, 40).

We and others have previously demonstrated that successful cancer immunotherapy requires simultaneous targeting of both effector and suppressor arms of the immune system (41–45). Because OX40 leads to increased INFγ expression in the periphery and within the tumor microenvironment (16, 17) and INFγ promotes the induction of IDO, we hypothesized that OX40-based antitumor immunotherapy could be improved by reducing immune suppression through inhibition of IDO. We, therefore, evaluated the therapeutic efficacy and immune mechanisms of treatment combining anti-OX40 with indoximod in a vaccine setting in mice with established tumors. To test the effect of IDO inhibition on the immune component of the tumor microenvironment, we tested the combination in an IDO tumor model, TC-1. We demonstrate that this treatment is therapeutically potent and discuss immune mechanisms that explain the role of each component within the combination. Here, we provide evidence that simultaneous targeting of the effector arm with anti-OX40 and the suppressor arm of immune system with an IDO inhibitor, indoximod, produces a synergistic effect on the tumor immune response resulting in the enhancement of vaccine potency and in tumor eradication. Considering that, on average, more than half of human tumors are IDO (46), we believe that this treatment strategy is promising, can be applied to a wide range of tumors (including IDO tumors), and can improve the efficacy of cancer treatment.

Mice and cell lines

C57BL/6 female mice (6–8 weeks old, The Jackson Laboratory) were housed in pathogen-free environment. All procedures were carried out with approved Augusta University institutional animal protocols and NIH guidelines.

TC-1 cell line, derived by cotransfection of human papilloma-virus strain 16 (HPV16) early proteins 6 and 7 (E6 and E7) and activated h-ras oncogene into primary lung epithelial cells, was obtained from the ATCC and propagated in RPMI1640 supplemented with 10% FBS, penicillin and streptomycin (100 U/mL each), L-glutamine (2 mmol/L) at 37°C with 5% CO2. Cells were maintained at a confluence of 70% to 80% and were always used from a passage number less than 10. These cells were tested routinely for absence of mycoplasma by using PCR at Georgia Cancer Center, Augusta University (Augusta, GA). All tests were negative. During the last one year, these cells were not reauthenticated.

This cell line was deliberately selected, as these tumor cells do not express IDO, allowing the study of the effect of the IDO inhibitor on the tumor microenvironment without interference from the direct effect on tumor cells.

Vaccine and other reagents

The CTL epitope from HPV16 E749–57 (9aa peptide, RAHYNIVTF, 100 μg/mouse) mixed with 13 amino acid synthetic nonnatural pan DR CD4+ T helper epitope (PADRE; ref. 47; 13aa peptide, aK-Cha-VAAWTLKAAa, where a is D-alanine, and Cha is L-cyclohexylalanine, 20 μg/mouse; both from Celtek Bioscience) and QuilA adjuvant (20 μg/mouse; Brenntag) was used as the model vaccine (that contains both CD8 and CD4 epitopes) in all studies [subcutaneous (s.c.) injections].

Indoximod (1-MT, Sigma-Aldrich) was provided in drinking water (freshly prepared every 3 days) at 2 mg/mL concentration. Water was supplemented with low-fat sweetener to improve the water intake by mice. Once all the material was dissolved, the pH was adjusted to a range of 7 to 7.5 with 1N HCl and filtered through a 75-mm filter unit. Indoximod drinking solution was provided to mice in amber-colored or aluminum foil-wrapped drinking bottle to protect from exposure to light.

Agonist rat anti-mouse OX40 (clone OX86, rat IgG1) was provided by MedImmune and delivered by intraperitoneal (i.p.) injections at a dose of 1 mg/kg.

Tumor implantation and treatment

For both therapeutic and immunology experiments, 7 × 104 TC-1 cells were inoculated s.c. into the right flank of the mice on day 0. Vaccine was given weekly s.c. starting on days 10 to 12 after tumor implantation when tumor diameter reached 4 to 5 mm. For therapeutic experiments, vaccine was given weekly throughout the experiment. Anti-OX40 and indoximod treatments started on the same day with vaccination. Anti-OX40 was given twice a week for the entire duration of treatment. Similarly, indoximod was provided in drinking water until the end of the study. A total of eight groups of mice (n = 5) were utilized in these experiments, including nontreated, single and double agent-treated and vaccine/anti-OX40/indoximod treated groups. Tumor growth and survival was monitored. Tumors were measured every 3 to 4 days using digital calipers, and tumor volume was calculated using the formula V = (W2 × L)/2, whereby V is the volume, L is the length (longer diameter) and W is width (shorter diameter). Mice were sacrificed when moribund or if tumor volume reached 1.5 cm3. For immunology experiments, mice were treated similarly and were sacrificed 6 days after the second immunization. Tumors were harvested for analysis of tumor infiltrating cells and IDO activity.

Profiling tumor-infiltrating immune cells

Resected tumors were homogenized using GentleMACS Dissociator (Miltenyi Biotec), and strained through a 70-μm nylon filters (BD Biosciences). Red blood cells were lysed with ACK buffer according to manufacturer's suggestions (Life Technologies). Samples were fixed and permeabilized using BD Pharmigen Mouse FoxP3 buffer set (BD Biosciences). LSRII SORP flow cytometer (BD Biosciences) was used to analyze the following tumor-infiltrating cell subsets: CD8+, CD8+E7+, CD8+E7+GrzB+, CD4+Foxp3 (non-Treg), and CD4+Foxp3+ (Treg) cells within live CD3+CD45+ population.

The following fluorophore-conjugated antibodies purchased from BD Biosciences or eBioscience were used for flow cytometry assay: CD45-PE, CD3-PE-CF594 or V450, CD8-V450 or APC, CD4-FITC, FoxP3-APC or PE-CF594, Granzyme B-APC. The FITC-conjugated dextramers specific to E749–57 peptide was purchased from Immudex . Live cells were identified by fixable near infrared live/dead viability staining as suggested by manufacturer (Life Technologies). Acquired data were analyzed using FlowJo software, version 10 (TreeStar).

The absolute numbers of tumor-infiltrating cells were standardized per 106 of total tumor cells. The fold change of tumor-infiltrating immune cells was presented as fold change over the nontreated group.

Evaluation of IDO activity

The IDO enzymatic activity in tumor homogenates was assessed using high-performance liquid chromatography (HPLC) techniques as described earlier (48). Briefly, a concentration of kynurenine (tryptophan catabolite) was measured before and after 2 hours of addition of exogenous L-tryptophan substrate solution into tumor homogenates. Tumors were harvested from control and treated mice. The treatment schedule and tumor harvest were the same as for the assessment of immunology response.

Statistical analysis

All statistical parameters (average values, SD, significant differences between groups) were calculated using GraphPad Prism software. Statistical significance between groups was determined by one-way ANOVA with a Tukey multiple comparison post-test (P < 0.05 was considered statistically significant).

Indoximod enhances agonistic effect of anti-OX40 to eradicate tumors

It is known that engagement of OX40 on T cells with its ligand or agonist antibody leads to increased INFγ expression in the periphery and within the tumor microenvironment (16, 17). INFγ induces IDO, which in turn produces a range of immune inhibitory effects within the tumor microenvironment (49, 50). Accordingly, we tested whether inhibiting the IDO pathway would enhance the therapeutic effect of anti-OX40. We tested this hypothesis in the TC-1 tumor model that lacks IDO expression to identify the effects of the combination specifically on the immune component of the tumor microenvironment rather than on the tumor cells. We first evaluated tumor growth and survival of animals after treatment of TC-1 tumor-bearing mice with vaccine, anti-OX40 agonist Ab and indoximod. TC-1 tumor cells were implanted on day 0 and treatment was initiated when tumors reached 4 to 5 mm in diameter. Tumor-bearing mice were treated weekly with an E7 peptide vaccine to generate an effector T-cell response against the tumor. Anti-OX40 agonist was administered twice a week and indoximod was provided in drinking water throughout the experiment as depicted in Fig. 1A, and tumor growth and survival were monitored.

Figure 1.

Combination of vaccine, anti-OX40 Ab, and indoximod provides potent antitumor therapeutic efficacy. A, C57BL/6 mice (n = 5/group) were injected s.c. in the right flank with 7 × 104 TC-1 cells. Mice from appropriate groups were injected weekly with vaccine (s.c.) or PBS as a control into the right flank. Anti-OX40 was injected i.p. twice a week starting on day 10 after tumor implantation throughout the experiment. Indoximod was provided in drinking water starting day 10 throughout the experiment. Tumor sizes were measured every 3–4 days. B, Plots represent tumor volumes of individual mice for each treatment. Ratio on the plots shows number of mice with completely regressed tumors over total number of mice in the group. Average tumor volumes ± SD on days 26 (C) and 40 (D). E, Kaplan–Meier plot of the overall survival. Similar results were obtained from two independent experiments. *, P < 0.05; **, P < 0.01. Experiment was repeated three times with similar results.

Figure 1.

Combination of vaccine, anti-OX40 Ab, and indoximod provides potent antitumor therapeutic efficacy. A, C57BL/6 mice (n = 5/group) were injected s.c. in the right flank with 7 × 104 TC-1 cells. Mice from appropriate groups were injected weekly with vaccine (s.c.) or PBS as a control into the right flank. Anti-OX40 was injected i.p. twice a week starting on day 10 after tumor implantation throughout the experiment. Indoximod was provided in drinking water starting day 10 throughout the experiment. Tumor sizes were measured every 3–4 days. B, Plots represent tumor volumes of individual mice for each treatment. Ratio on the plots shows number of mice with completely regressed tumors over total number of mice in the group. Average tumor volumes ± SD on days 26 (C) and 40 (D). E, Kaplan–Meier plot of the overall survival. Similar results were obtained from two independent experiments. *, P < 0.05; **, P < 0.01. Experiment was repeated three times with similar results.

Close modal

We found that although none of the single-agent treatments affects the tumor growth, vaccine/anti-OX40 leads to slower tumor progression and complete tumor regression in 20% of the mice when added to the vaccine (Fig. 1B). The addition of indoximod to this treatment significantly enhanced therapeutic efficacy resulting in a complete eradication of established tumors in 60% of the mice. In addition, enhanced therapeutic efficacy after treatment with vaccine and anti-OX40 was seen early in the treatment (Fig. 1C; day 26 representing the last day when all mice are surviving in all control groups). However, incorporation of indoximod into this treatment improved the long-term therapeutic outcome as shown at day 40 (Fig. 1D). Moreover, compared with the single treatments, vaccine/anti-OX40 led to prolonged survival and addition of indoximod to this treatment resulted in a sustained response beyond 65 days (Fig. 1B and E). These data demonstrate that IDO inhibition with indoximod significantly enhances the effect of OX40 agonist antibody.

Combination of anti-OX40 with indoximod results in significant increase of CD8+ T cell to Treg ratio

To dissect the immune mechanisms leading to the enhanced therapeutic efficacy of vaccine/anti-OX40/indoximod treatment, we tested the effect of the combination on the tumor-infiltrating T cells. Animals were treated as mentioned above; 6 days after the second vaccination, mice were sacrificed and tumors were harvested for evaluation of tumor-infiltrating T-cell subsets by flow cytometry.

Neither the single agent treatments nor the vaccine/anti-OX40 combination significantly affected CD4+FoxP3 T-cell tumor infiltration compared with either agent alone. However, we found that the addition of indoximod to vaccine/anti-OX40 led to a significant increase in tumor-infiltrating CD4+FoxP3 (non-Treg CD4+ T cells; Fig. 2A). None of the treatments significantly affected the numbers of tumor-infiltrating Tregs (Fig. 2B). Although the percentage of tumor-infiltrating Tregs within the total CD4+ T-cell population was decreased in the vaccine/anti-OX40 group (due to increase of CD4+FoxP3 T cells), this reduction was more significant when indoximod was added to the treatment (Fig. 2C).

Figure 2.

Vaccine/anti-OX40/indoximod treatments lead to an increase of non-Treg CD4+ T-cell infiltration into the tumor without affecting Treg cell number. C57BL/6 mice (n = 4–5/group) were injected with 7 × 104 TC-1 cells and treated as for therapeutic studies, except on day 23 mice were sacrificed, and tumor-infiltrating CD4+ T cells were analyzed in tumor homogenates by flow cytometry. The absolute numbers of tumor-infiltrating cells were standardized per 106 of total tumor cells and presented as fold change over nontreated (NT) group for A. Non-Treg CD4+ T cells (CD4+ FoxP3), B. Tregs (CD4+ FoxP3+—no statistically significant differences were observed between any of the groups), or as a percentage of Tregs within CD4+ population (C). *, P < 0.05; ** P < 0.01. Combined data from three independent experiments are shown.

Figure 2.

Vaccine/anti-OX40/indoximod treatments lead to an increase of non-Treg CD4+ T-cell infiltration into the tumor without affecting Treg cell number. C57BL/6 mice (n = 4–5/group) were injected with 7 × 104 TC-1 cells and treated as for therapeutic studies, except on day 23 mice were sacrificed, and tumor-infiltrating CD4+ T cells were analyzed in tumor homogenates by flow cytometry. The absolute numbers of tumor-infiltrating cells were standardized per 106 of total tumor cells and presented as fold change over nontreated (NT) group for A. Non-Treg CD4+ T cells (CD4+ FoxP3), B. Tregs (CD4+ FoxP3+—no statistically significant differences were observed between any of the groups), or as a percentage of Tregs within CD4+ population (C). *, P < 0.05; ** P < 0.01. Combined data from three independent experiments are shown.

Close modal

Next, we explored the effect of the addition of indoximod to anti-OX40 on tumor-infiltrating CD8+ T cells. We found that treatment with vaccine/anti-OX40 significantly increased the number of CD8+ T cells infiltrating the tumor compared with mice from control groups (Fig. 3A). Although addition of indoximod to vaccine/anti-OX40 resulted in better antitumor response, no significant differences were found in the numbers of tumor-infiltrating CD8+ T cells over the vaccine/anti-OX40 group (Fig. 3A). Both groups showed a significant increase in CD8+/Treg ratio when compared with control groups (Fig. 3B).

Figure 3.

Both vaccine/anti-OX40 and vaccine/anti-OX40/indoximod treatments lead to an increase of CD8+ T-cell infiltration into the tumor and CD8+/Treg ratio. C57BL/6 mice (n = 4–5/group) were injected with 7 × 104 TC-1 cells and treated as for Fig. 2, and tumor-infiltrating CD8+ T cells were analyzed in tumor homogenates by flow cytometry. The absolute numbers of tumor-infiltrating CD8+ T cells were standardized per 106 of total tumor cells and presented as fold change over nontreated (NT) group (A). B, The CD8+/Treg ratio was calculated from normalized numbers of CD8+ T cells and Tregs per 106 of total tumor cells. *, P < 0.05; **, P < 0.01; ***; P < 0.001. Combined data from three independent experiments are shown.

Figure 3.

Both vaccine/anti-OX40 and vaccine/anti-OX40/indoximod treatments lead to an increase of CD8+ T-cell infiltration into the tumor and CD8+/Treg ratio. C57BL/6 mice (n = 4–5/group) were injected with 7 × 104 TC-1 cells and treated as for Fig. 2, and tumor-infiltrating CD8+ T cells were analyzed in tumor homogenates by flow cytometry. The absolute numbers of tumor-infiltrating CD8+ T cells were standardized per 106 of total tumor cells and presented as fold change over nontreated (NT) group (A). B, The CD8+/Treg ratio was calculated from normalized numbers of CD8+ T cells and Tregs per 106 of total tumor cells. *, P < 0.05; **, P < 0.01; ***; P < 0.001. Combined data from three independent experiments are shown.

Close modal

These data demonstrate that the enhanced antitumor efficacy caused by inhibition of IDO is not facilitated by the increase in CD8+ T-cell number, suggesting the existence of other mechanisms driving the therapeutic efficacy of the combination treatment.

Addition of indoximod to anti-OX40 treatment increases antigen-specific functional CD8+ T cells

We have demonstrated that the addition of indoximod to vaccine/anti-OX40 immunotherapy did not affect the total number of tumor-infiltrating CD8+ T cells or the CD8+/Treg ratio, despite the fact that addition of indoximod improved therapeutic efficacy of anti-OX40.

Therefore, to understand the reasons for improved therapeutic outcome when indoximod is added to vaccine/anti-OX40, we first confirmed the efficacy of indoximod for the IDO enzymatic activity within tumor homogenates by measuring the levels of kynurenine catabolites. As expected, we detected no differences of kynurenine catabolites produced by tumor homogenates after addition of exogenous tryptophan in untreated mice and animals treated with vaccine alone or vaccine/anti-OX40 compared with nontreated mice (Fig. 4A). Moreover, there was no difference in IDO activity in mice treated with anti-OX40/indoximod and vaccine/indoximod. However, mice treated with vaccine/anti-OX40/indoximod had significantly decreased IDO activity (P < 0.05) compared with nontreated mice (Fig. 4A).

Figure 4.

Addition of indoximod to vaccine/anti-OX40 treatment leads to decrease in kynurenine and an increase of tumor-infiltrating E7-specific and E7+GrzB+ CD8+ T cells. A, Enzymatic activity of IDO was evaluated in tumor homogenates from control and treated mice. IDO activity was calculated as pmol kynurenine/hour/mg tissue. B and C, Tumor-bearing C57BL/6 mice (n = 5/group) were treated with vaccine alone, vaccine/anti-OX40, vaccine/indoximod, or vaccine/anti-OX40/indoximod as described above. The absolute numbers of tumor-infiltrating E7 dextramer+ CD8+ T cells (B) and E7+GrzB+ CD8+ T cells (C) were standardized per 106 of total tumor cells and presented as mean values ± SD. *, P < 0.05 compared with all other groups. Similar results were obtained from two independent experiments.

Figure 4.

Addition of indoximod to vaccine/anti-OX40 treatment leads to decrease in kynurenine and an increase of tumor-infiltrating E7-specific and E7+GrzB+ CD8+ T cells. A, Enzymatic activity of IDO was evaluated in tumor homogenates from control and treated mice. IDO activity was calculated as pmol kynurenine/hour/mg tissue. B and C, Tumor-bearing C57BL/6 mice (n = 5/group) were treated with vaccine alone, vaccine/anti-OX40, vaccine/indoximod, or vaccine/anti-OX40/indoximod as described above. The absolute numbers of tumor-infiltrating E7 dextramer+ CD8+ T cells (B) and E7+GrzB+ CD8+ T cells (C) were standardized per 106 of total tumor cells and presented as mean values ± SD. *, P < 0.05 compared with all other groups. Similar results were obtained from two independent experiments.

Close modal

Next, we tested the antigen specificity and functionality of tumor-infiltrating CD8+ T cells, which could also be responsible for the differences in therapeutic efficacy between vaccine/anti-OX40 and vaccine/anti-OX40/indoximod treatments.

The specificity and functionality of tumor-infiltrating CD8+ T cells were evaluated from mice treated with vaccine, vaccine/anti-OX40, vaccine/indoximod, or vaccine/anti-OX40/indoximod as described above. Although the addition of indoximod to anti-OX40/vaccine was unable to increase the total number of CD8+ T cells in the tumor microenvironment compared with vaccine/OX40 group (Fig. 3A), incorporation of indoximod into the treatment resulted in a significantly higher number of tumor-infiltrating antigen (E7)-specific CD8+ T cells compared with all other groups (Fig. 4B). Granzyme B (Grz B) expression was used to characterize the functionality of these CD8+ T cells. We demonstrated that the addition of indoximod to vaccine/anti-OX40 also led to a significant increase in the number of tumor-infiltrating CD8+GrzB+ antigen-specific T cells compared with all other groups (Fig. 4C).

These data demonstrate that providing indoximod simultaneously with vaccine/anti-OX40 not only leads to decreased IDO activity in tumor but also increases numbers of functional antigen-specific CD8+ T cells, which might explain the differences in therapeutic efficacy observed between the treatment groups.

Thus, we show that although the addition of anti-OX40 to vaccine treatment enhances the numbers of CD8+ T cells in tumors (“quantity”), addition of indoximod improves the “quality” of these CD8+ T cells (enhanced antigen specificity and functionality).

Here, we tested the agonist anti-OX40 in combination with the IDO pathway inhibitor, indoximod in the context of cancer vaccine. We showed that anti-OX40 agonist can enhance the effect of vaccine treatment resulting in slower tumor progression and complete regression of established tumors in 20% of treated mice. We hypothesized that the addition of indoximod to anti-OX40 treatment can improve the overall therapeutic efficacy of vaccine treatment. We showed that combinational treatment with vaccine/anti-OX40/indoximod significantly enhanced antitumor efficacy and resulted in complete tumor eradication in 60% of mice. While evaluating immunologic mechanisms responsible for this potent therapeutic outcome, we found that an increase of tumor-infiltrating CD8+ T cells for the combinational treatments is mediated by anti-OX40, as both vaccine/anti-OX40 and vaccine/anti-OX40/indoximod treatments had similar numbers of CD8+ T cells. Likewise, anti-OX40 was responsible for the increase in non-Treg CD4+ T-cell infiltration into the tumor, thus resulting in decreased percentage of Tregs within the tumor-infiltrating CD4+ T-cell population.

We found no significant effect of anti-OX40 on the number of tumor infiltrating Tregs. This contrasts with results in the CT26 tumor model, where the same anti-OX40 clone resulted in Treg depletion within the tumor and led to enhanced therapeutic efficacy (51). This could be attributed to the different immune profiles of the two tumor models.

We found no differences in numbers of tumor-infiltrating CD8+ T cells or in CD8+/Treg ratio, a well-established criterion that correlates with cancer prognosis (45, 52), between vaccine/anti-OX40 and vaccine/anti-OX40/indoximod treatments, even though the therapeutic efficacy for these treatments was significantly different. To understand this phenomenon, we evaluated the specificity and functionality of tumor-infiltrating CD8+ T cells. We found that numbers of both E7-specific and Granzyme B+ functional antigen-specific CD8+ T cells were elevated within the tumors of mice treated with vaccine/anti-OX40/indoximod compared with other groups. This, at least partially, explains the differences in antitumor therapeutic efficacies between these two treatments. Considering that more than half of the human tumors are IDO (46), we selected the TC-1 tumor model for evaluation of indoximod-based immunotherapy. As these tumor cells (in culture and in vivo) do not express IDO, the observed effect elicited by indoximod would be due not to its direct effect on tumor cells but rather to its effect on immune suppressive cells within the tumor microenvironment. Under normal physiological conditions, IDO contributes to maintenance of immune tolerance (27). The catabolic effect of IDO on tryptophan depletion and kynurenine accumulation leads to T-cell arrest or apoptosis (34–37). In the tumor setting, this phenomenon enhances tumor growth by hindering immune effector cell function (28, 33, 38). We observed no differences in IDO activity between animals treated with vaccine or with vaccine/anti-OX40, whereas the addition of indoximod to vaccine/anti-OX40 decreased IDO activity compared with other groups. Thus, one possibility for increased specificity and functionality of tumor-infiltrating CD8+ T cells from mice treated with indoximod in addition to vaccine/anti-OX40 is the decrease in IDO enzymatic activity within the tumors of these mice.

In conclusion, here we demonstrate the synergistic antitumor therapeutic effect of vaccine/anti-OX40/indoximod combination that leads to tumor regression, and explore the immune mechanisms responsible for that synergy. Dissecting the mechanisms for each compound within the combination shows that anti-OX40 is responsible for the overall increase in numbers, and indoximod for specificity and functionality of vaccine-induced CD8+ T cells. We provide evidence that simultaneous targeting of the effector and suppressor arms of the immune system synergistically promotes tumor eradication. This translatable strategy may enhance the overall efficacy of cancer treatment.

M. Mkrtichyan is a scientist at FivePrime Therapeutics Inc. J.E. Janik is executive director at Incyte. S.N. Khleif reports receiving a commercial research grant from MedImmune and is a consultant/advisory board member for NewLink. No potential conflicts of interest were disclosed by the other authors.

Conception and design: Z. Berrong, M. Mkrtichyan, S. Hammond, J.E. Janik, S.N. Khleif

Development of methodology: Z. Berrong, S. Ahmad, S.N. Khleif

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Z. Berrong, S. Ahmad, M. Webb, E. Mohamed, G. Okoev, R. Shrimali, S.N. Khleif

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Z. Berrong, M. Mkrtichyan, S. Ahmad, G. Okoev, R. Abu Eid, J.E. Janik, S.N. Khleif

Writing, review, and/or revision of the manuscript: Z. Berrong, M. Mkrtichyan, S. Ahmad, E. Mohamed, G. Okoev, R. Shrimali, R. Abu Eid, S. Hammond, J.E. Janik, S.N. Khleif

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Z. Berrong, G. Okoev, A. Matevosyan, S.N. Khleif

Study supervision: Z. Berrong, S.N. Khleif

The authors thank Rhea-Beth Markowitz, Charles C. Berrong, and Seema Gupta for reviewing the manuscript.

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.
Naito
Y
,
Saito
K
,
Shiiba
K
,
Ohuchi
A
,
Saigenji
K
,
Nagura
H
,
Ohtani
H
. 
CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer
.
Cancer Res
1998
;
58
:
3491
4
.
2.
Carretero
R
,
Sektioglu
IM
,
Garbi
N
,
Salgado
OC
,
Beckhove
P
,
Hämmerling
GJ
, et al
Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8 T cells
.
Nat Immunol
2015
;
16
:
609
17
.
3.
Romano
E
,
Kusio-Kobialka
M
,
Foukas
PG
,
Baumgaertner
P
,
Meyer
C
,
Ballabeni
P
, et al
Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients
.
Proc Natl Acad Sci U S A
2015
;
112
:
6140
5
.
4.
Burns
WR
,
Zhao
Y
,
Frankel
TL
,
Hinrichs
CS
,
Zheng
Z
,
Xu
H
, et al
A high molecular weight melanoma-associated antigen-specific chimeric antigen receptor redirects lymphocytes to target human melanomas
.
Cancer Res
2010
;
70
:
3027
33
.
5.
Almasbak
H
,
Walseng
E
,
Kristian
A
,
Myhre
MR
,
Suso
EM
,
Munthe
LA
, et al
Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model
.
Gene Ther
2015
;
22
:
391
403
.
6.
Hombach
AA
,
Abken
H
. 
Of chimeric antigen receptors and antibodies: OX40 and 41BB costimulation sharpen up T cell-based immunotherapy of cancer
.
Immunotherapy
2013
;
5
:
677
81
.
7.
Hombach
AA
,
Heiders
J
,
Foppe
M
,
Chmielewski
M
,
Abken
H
. 
OX40 costimulation by a chimeric antigen receptor abrogates CD28 and IL-2 induced IL-10 secretion by redirected CD4(+) T cells
.
Oncoimmunology
2012
;
1
:
458
466
.
8.
Chacon
JA
,
Wu
RC
,
Sukhumalchandra
P
,
Molldrem
JJ
,
Sarnaik
A
,
Pilon-Thomas
S
, et al
Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy
.
PLoS One
2013
;
8
:
e60031
.
9.
Mallett
S
,
Barclay
AN
. 
A new superfamily of cell surface proteins related to the nerve growth factor receptor
.
Immunol Today
1991
;
12
:
220
3
.
11.
Sleckman
BP
,
Peterson
A
,
Jones
WK
,
Foran
JA
,
Greenstein
JL
,
Seed
B
, et al
Expression and function of CD4 in a murine T-cell hybridoma
.
Nature
1987
;
328
:
351
3
.
12.
Mallett
S
,
Fossum
S
,
Barclay
AN
. 
Characterization of the MRC OX40 antigen of activated CD4 positive T lymphocytes–a molecule related to nerve growth factor receptor
.
EMBO J
1990
;
9
:
1063
8
.
13.
Croft
M
Control of immunity by the TNFR-related molecule OX40 (CD134)
.
Annu Rev Immunol
2010
;
28
:
57
78
.
14.
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
.
15.
Gramaglia
I
,
Jember
A
,
Pippig
SD
,
Weinberg
AD
,
Killeen
N
,
Croft
M
. 
The OX40 costimulatory receptor determines the development of CD4 memory by regulating primary clonal expansion
.
J Immunol
2000
;
165
:
3043
50
.
16.
Rogers
PR
,
Song
J
,
Gramaglia
I
,
Killeen
N
,
Croft
M
. 
OX40 promotes Bcl-xL and Bcl-2 expression and is essential for long-term survival of CD4 T cells
.
Immunity
2001
;
15
:
445
55
.
17.
Sugamura
K
,
Ishii
N
,
Weinberg
AD
. 
Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40
.
Nat Rev Immunol
2004
;
4
:
420
31
.
18.
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
.
19.
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
.
20.
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
.
21.
Piconese
S
,
Valzasina
B
,
Colombo
MP
. 
OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection
.
J Exp Med
2008
;
205
:
825
39
.
22.
Humphreys
IR
,
Loewendorf
A
,
de Trez
C
,
Schneider
K
,
Benedict
CA
,
Munks
MW
, et al
OX40 costimulation promotes persistence of cytomegalovirus-specific CD8 T Cells: A CD4-dependent mechanism
.
J Immunol
2007
;
179
:
2195
202
.
23.
Biagi
E
,
Dotti
G
,
Yvon
E
,
Lee
E
,
Pule
M
,
Vigouroux
S
, et al
Molecular transfer of CD40 and OX40 ligands to leukemic human B cells induces expansion of autologous tumor-reactive cytotoxic T lymphocytes
.
Blood
2005
;
105
:
2436
42
.
24.
Draghiciu
O
,
Nijman
HW
,
Daemen
T
. 
From tumor immunosuppression to eradication: targeting homing and activity of immune effector cells to tumors
.
Clin Dev Immunol
2011
;
2011
:
439053
.
25.
Mellman
I
,
Coukos
G
,
Dranoff
G
. 
Cancer immunotherapy comes of age
.
Nature
2011
;
480
:
480
9
.
26.
Töpfer
K
,
Kempe
S
,
Müller
N
,
Schmitz
M
,
Bachmann
M
,
Cartellieri
M
, et al
Tumor evasion from T cell surveillance
.
J Biomed Biotechnol
2011
;
2011
:
918471
.
27.
Uyttenhove
C
,
Pilotte
L
,
Théate
I
,
Stroobant
V
,
Colau
D
,
Parmentier
N
, et al
Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase
.
Nat Med
2003
;
9
:
1269
74
.
28.
Munn
DH
,
Mellor
AL
. 
IDO and tolerance to tumors
.
Trends Mol Med
2004
;
10
:
15
8
.
29.
Harlin
H
,
Kuna
TV
,
Peterson
AC
,
Meng
Y
,
Gajewski
TF
. 
Tumor progression despite massive influx of activated CD8(+) T cells in a patient with malignant melanoma ascites
.
Cancer Immunol Immunother
2006
;
55
:
1185
97
.
30.
Polak
ME
,
Borthwick
NJ
,
Gabriel
FG
,
Johnson
P
,
Higgins
B
,
Hurren
J
, et al
Mechanisms of local immunosuppression in cutaneous melanoma
.
Br J Cancer
2007
;
96
:
1879
87
.
31.
Weinlich
G
,
Murr
C
,
Richardsen
L
,
Winkler
C
,
Fuchs
D
. 
Decreased serum tryptophan concentration predicts poor prognosis in malignant melanoma patients
.
Dermatology
2007
;
214
:
8
14
.
32.
Thaker
AI
,
Rao
MS
,
Bishnupuri
KS
,
Kerr
TA
,
Foster
L
,
Marinshaw
JM
, et al
IDO1 metabolites activate beta-catenin signaling to promote cancer cell proliferation and colon tumorigenesis in mice
.
Gastroenterology
2013
;
145
:
416
25
e1–4
.
33.
Munn
DH
,
Sharma
MD
,
Hou
D
,
Baban
B
,
Lee
JR
,
Antonia
SJ
, et al
Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes
.
J Clin Invest
2004
;
114
:
280
90
.
34.
Fallarino
F
,
Grohmann
U
,
Vacca
C
,
Bianchi
R
,
Orabona
C
,
Spreca
A
, et al
T cell apoptosis by tryptophan catabolism
.
Cell Death Differ
2002
;
9
:
1069
77
.
35.
Frumento
G
,
Rotondo
R
,
Tonetti
M
,
Damonte
G
,
Benatti
U
,
Ferrara
GB
. 
Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase
.
J Exp Med
2002
;
196
:
459
68
.
36.
Terness
P
,
Bauer
TM
,
Röse
L
,
Dufter
C
,
Watzlik
A
,
Simon
H
, et al
Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites
.
J Exp Med
2002
;
196
:
447
57
.
37.
Munn
DH
,
Sharma
MD
,
Lee
JR
,
Jhaver
KG
,
Johnson
TS
,
Keskin
DB
, et al
Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase
.
Science
2002
;
297
:
1867
70
.
38.
Munn
DH
,
Mellor
AL
. 
Indoleamine 2,3-dioxygenase and tumor-induced tolerance
.
J Clin Invest
2007
;
117
:
1147
54
.
39.
Friberg
M
,
Jennings
R
,
Alsarraj
M
,
Dessureault
S
,
Cantor
A
,
Extermann
M
, et al
Indoleamine 2,3-dioxygenase contributes to tumor cell evasion of T cell-mediated rejection
.
Int J Cancer
2002
;
101
:
151
5
.
40.
Muller
AJ
,
DuHadaway
JB
,
Donover
PS
,
Sutanto-Ward
E
,
Prendergast
GC
. 
Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy
.
Nat Med
2005
;
11
:
312
9
.
41.
Mkrtichyan
M
,
Najjar
YG
,
Raulfs
EC
,
Abdalla
MY
,
Samara
R
,
Rotem-Yehudar
R
, et al
Anti-PD-1 synergizes with cyclophosphamide to induce potent anti-tumor vaccine effects through novel mechanisms
.
Eur J Immunol
2011
;
41
:
2977
86
.
42.
Sharma
P
,
Wagner
K
,
Wolchok
JD
,
Allison
JP
. 
Novel cancer immunotherapy agents with survival benefit: recent successes and next steps
.
Nat Rev Cancer
2011
;
11
:
805
12
.
43.
Mkrtichyan
M
,
Najjar
YG
,
Raulfs
EC
,
Liu
L
,
Langerman
S
,
Guittard
G
, et al
B7-DC-Ig enhances vaccine effect by a novel mechanism dependent on PD-1 expression level on T cell subsets
.
J Immunol
2012
;
189
:
2338
47
.
44.
Mkrtichyan
M
,
Chong
N
,
Eid
RA
,
Wallecha
A
,
Singh
R
,
Rothman
J
, et al
Anti-PD-1 antibody significantly increases therapeutic efficacy of Listeria monocytogenes (Lm)-LLO immunotherapy
.
J Immunother Cancer
2013
;
1
:
15
.
45.
Curran
MA
,
Montalvo
W
,
Yagita
H
,
Allison
JP
. 
PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors
.
Proc Natl Acad Sci U S A
2010
;
107
:
4275
80
.
46.
Godin-Ethier
J
,
Hanafi
LA
,
Piccirillo
CA
,
Lapointe
R
. 
Indoleamine 2,3-dioxygenase expression in human cancers: clinical and immunologic perspectives
.
Clin Cancer Res
2011
;
17
:
6985
91
.
47.
Alexander
J
,
del Guercio
MF
,
Maewal
A
,
Qiao
L
,
Fikes
J
,
Chesnut
RW
, et al
Linear PADRE T helper epitope and carbohydrate B cell epitope conjugates induce specific high titer IgG antibody responses
.
J Immunol
2000
;
164
:
1625
33
.
48.
Hoshi
M
,
Saito
K
,
Hara
A
,
Taguchi
A
,
Ohtaki
H
,
Tanaka
R
, et al
The absence of IDO upregulates type I IFN production, resulting in suppression of viral replication in the retrovirus-infected mouse
.
J Immunol
2010
;
185
:
3305
12
.
49.
Spranger
S
,
Spaapen
RM
,
Zha
Y
,
Williams
J
,
Meng
Y
,
Ha
TT
, et al
Up-regulation of PD-L1, IDO, and T(regs) in the melanoma tumor microenvironment is driven by CD8(+) T cells
.
Sci Transl Med
2013
;
5
:
200ra116
.
50.
Munn
DH
,
Shafizadeh
E
,
Attwood
JT
,
Bondarev
I
,
Pashine
A
,
Mellor
AL
. 
Inhibition of T cell proliferation by macrophage tryptophan catabolism
.
J Exp Med
1999
;
189
:
1363
72
.
51.
Bulliard
Y
,
Jolicoeur
R
,
Zhang
J
,
Dranoff
G
,
Wilson
NS
,
Brogdon
JL
. 
OX40 engagement depletes intratumoral Tregs via activating FcgammaRs, leading to antitumor efficacy
.
Immunol Cell Biol
2014
;
92
:
475
80
.
52.
Shen
Z
,
Zhou
S
,
Wang
Y
,
Li
RL
,
Zhong
C
,
Liang
C
, et al
Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer
.
J Cancer Res Clin Oncol
2010
;
136
:
1585
95
.