Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jan 1.
Published in final edited form as: Cancer Res. 2009 Dec 22;70(1):129–138. doi: 10.1158/0008-5472.CAN-09-3170

Central Role of IFNγ-Indoleamine 2,3 Dioxygenase Axis in Regulation of Interleukin-12-mediated Antitumor Immunity

Tao Gu 1, Rachael B Rowswell-Turner 1, Mehmet O Kilinc 1, Nejat K Egilmez 1
PMCID: PMC2805056  NIHMSID: NIHMS157637  PMID: 20028855

Abstract

Sustained intratumoral delivery of IL-12 and GM-CSF induces tumor regression via restoration of tumor-resident CD8+ T-effector/memory cell cytotoxicity and subsequent re-priming of a secondary CD8+ T-effector cell response in tumor-draining lymph nodes (TDLN). However, treatment-induced T-effector activity is transient and is accompanied with a CD4+ CD25+ Foxp3+ T-suppressor cell rebound. Molecular and cellular changes in post-therapy tumor microenvironment and TDLN were monitored to elucidate the mechanism of counter-regulation. Real-time PCR analysis revealed a 5-fold enhancement of indoleamine 2, 3 dioxygenase (IDO) expression in the tumor and the TDLN after treatment. IDO induction required IFNγ and persisted for up to 7 days. Administration of the IDO inhibitor D-1-methyl tryptophan (D-1MT) concurrent with treatment resulted in a dramatic enhancement of tumor regression. Enhanced efficacy was associated with a diminished T-suppressor cell rebound, revealing a link between IDO activity and post-therapy regulation. Further analysis established that abrogation of the regulatory counter-response resulted in a 10-fold increase in the intratumoral CD8+ T-cell to CD4+ Foxp3+ T-cell ratio. The ratio of proliferating CD8+ T-effector to CD4+ Foxp3+ T-suppressor cells was prognostic for efficacy of tumor suppression in individual mice. IFNγ-dependent IDO induction and T-suppressor cell expansion were primarily driven by IL-12. These findings demonstrate a critical role for IDO in the regulation of IL-12-mediated antitumor immune responses.

Introduction

Cancer vaccines can break tolerance to tumor antigens and induce potent anti-tumor T-cell activity (1, 2). However, in the great majority of cases anti-tumor T-cells fail to induce effective tumor regression, independent of the intensity of the response (13). The immune suppressive nature of the tumor microenvironment has been identified as a major factor contributing to the inability of T-cells to mediate effective tumor regression (4, 5). Blocking of suppressive mechanisms during therapy results in improved tumor eradication however, durable regressions are rarely achieved (6, 7). Whether the lack of long-term efficacy is due to the inherent transient nature of effector T-cell responses (8) and the associated T-cell intrinsic regulatory pathways (9), to resurgence of tumor-mediated immune dysfunction (4) or a combination of these factors is not established.

Previous studies demonstrated that sustained intratumoral delivery of IL-12 and GM-CSF restored tumor-resident CD8+ T-effector/memory cell (Tem) cytotoxicity and induced de novo priming of antitumor CD8+ T-effector cells in the TDLN (10, 11). Treatment-induced immune activation however, subsided within a week and was followed by a T-suppressor cell rebound (12). Since IL-12 directly restored full effector function to pre-existing quiescent CD8+ Tem in the presence of T-suppressor cells (10), we hypothesized that repeated stimulation could revive and extend the window of CD8+ Tem cytotoxicity, and enhance long-term tumor suppression. Chronic treatment however, resulted in progressive loss of therapeutic efficacy culminating in tumor resurgence (12). Long-term monitoring of intratumoral T-cell populations demonstrated that multiple treatments led to the intensification of the T-suppressor cell rebound after each cycle of therapy and ultimately resulted in the loss of anti-tumor Tem (12). These findings suggested that feedback inhibition may represent an important tumor-independent mechanism that can limit long-term therapeutic efficacy.

Whereas the majority of tumor immune therapy protocols focus on the induction phase of the anti-tumor immune response, few studies have addressed the mechanism of post-activation regulatory rebound. Consistent with our findings, these studies demonstrated that repeated vaccination or immune stimulation can promote the expansion of the CD4+ CD25+ Foxp3+ T-suppressor cell subset, in murine models of autoimmunity and cancer (1315). On the other hand, the molecular basis of therapy-induced T-suppressor cell expansion has not been delineated. To this end, quantitative and qualitative monitoring of the tumor microenvironment and the TDLN was undertaken to investigate the mechanism of post-IL-12/GM-CSF T-suppressor cell amplification. The results demonstrate a central role for the IL-12-IFNγ-IDO axis in treatment-induced CD4+ Foxp3+ T-cell expansion and the associated loss of therapeutic efficacy in our model.

Materials and Methods

Mice and tumors

BALB/c mice were purchased from Taconic laboratories (Germantown, NY) or bred in our facility. IFNγ knockout mice in BALB/c background were purchased from Jackson Laboratories (Bar Harbor, ME). The BALB/c syngeneic mammary carcinoma cell line 4T1 (16) was used in all experiments.

Microspheres and treatments

Recombinant murine IL-12 was a gift from Wyeth Pharmaceuticals (Wyeth, Andover, MA). Recombinant murine GM-CSF was purchased from Peprotech Inc. (Rocky Hill, NJ). Cytokines were encapsulated into polylactic acid microspheres via phase inversion nanoencapsulation (10). Mice were treated with a single intratumoral injection of 2 mg of each preparation (1 μg of cytokine) as described previously (10).

Surgical metastasis model

Mice were injected subcutaneously with 3 × 105 tumor cells in 0.1 ml of PBS in the mammary tissue and tumors were allowed to grow to 6–7 mm in diameter (14 days) to establish metastases. Primary tumors were treated with a single intratumoral injection of microspheres and tumors were surgically resected one week after treatment (17). Mice were euthanized 3 weeks after surgery and lung tumor burden was quantified using a clonogenic metastasis assay (17).

Quantitative real-time PCR (qRT-PCR) analysis

qRT-PCR was performed essentially as described (17). Transcript levels were calculated by the comparative threshold cycle (Ct) method (17). The relative target quantity, normalized to an endogenous control (GADPH) and relative to the day zero calibrator, was expressed as 2−ΔΔCt (fold), where ΔCt = Ct of the target gene – Ct of endogenous control gene, and ΔΔCt = ΔCt of samples for the target gene – ΔCt of zero day calibrator for the target gene. CD8 and IFNγ primers were published previously (17). The primer sequences for Foxp3, IDO-1 and IDO-2 were: Foxp3, 5'-TCC CAC GCT CGG GTA CAC-3' (forward), 5'-TTG CCA GCA GTG GGT AGG AT-3' (reverse); IDO-1, 5'-CAG GCC AGA GCA GCA TCT TC-3' (forward), 5'-GCC AGC CTC GTG TTT TAT TCC-3' (reverse); IDO-2, 5'-TGTCCTGGTGCTTAGCAGTCATGT-3' (forward), 5'-TGCAGGATGTGAACCTCTAACGCT-3'(reverse).

In vivo cell depletions

CD8+ T-cells were depleted via in vivo antibody administration (17).

BrdU labeling

BrdU labeling was performed as described by us previously (11).

Preparation of single cell suspensions

Single cell suspensions from tumors and TDLN were prepared essentially as described (10).

Flow cytometry

Flow cytometric analysis of single cell suspensions prepared from tumors and TDLN was performed on a 4-color FACSCalibur Flow Cytometer (BD Pharmingen) using established protocols (10). All antibodies and staining protocols were as described previously (10, 17).

Administration of D/L-1MT

D- and L-1MT (Sigma-Aldrich) were prepared and administered to tumor-bearing mice in drinking water starting 2 days prior to microsphere treatments until day 14 post-microsphere injection as described (18).

Statistical analysis

Student's t-test was used to determine the significance of the differences between control and experimental groups. In experiments with multiple groups homogeneity of inter-group variance was analyzed by ANOVA. Log-Rank analysis was employed to determine significance in survival studies. In all analyses p ≤ 0.05 was considered significant.

Results

Post-treatment CD8+ T-cell cytotoxicity is transient

Previous studies demonstrated that intratumoral IL-12 and GM-CSF promoted rapid activation of tumor-resident CD8+ T-effector/memory cells followed by priming of an anti-tumor CD8+ T-effector cell response in the TDLN (10, 11). Although the secondary CD8+ T-effectors effectively homed to and infiltrated the tumors, whether they mediated effective long-term tumor killing was not determined (11). To this end, we monitored the cytotoxicity window for the secondary CD8+ T-effectors in a murine surgical tumor metastasis model that allows quantitative analysis of systemic tumor eradication (19, 20). In this model, subcutaneous primary tumors are established and are allowed to metastasize to the lungs (Figure 1A). Primary tumors are then treated with a single intratumoral injection of IL-12 and GM-CSF microspheres followed by resection of the primary tumors one week after treatment. Lung tumor burden in post-surgical mice is then quantified 3 weeks after surgery using a clonogenic metastasis assay (17). In the current study mice were depleted of CD8+ T-cells in a time-dependent manner to identify the interval of CD8+ T-effector cell activity required for eradication of systemic disease (Figure 1A). The results are shown in Figure 1B. The data establish that a single treatment resulted in effective elimination of established lung metastases, reducing tumor burden from an average of 560 ± 89 to 65 ± 48 colonies/lung. Importantly, 60% of the mice in the treatment group were found to be lung tumor-free whereas all mice in the control group had advanced lesions. Depletion of CD8+ T-cells starting one day prior to IL-12/GM-CSF microsphere injection resulted in the complete loss of anti-tumor efficacy demonstrating the central role of CD8+ T-cells in the eradication of systemic disease. Similar results were obtained when CD8+ T-cell depletion was initiated starting on day 4 post-treatment, revealing that CD8+ T-cell activity was required beyond 4 days to achieve effective tumor kill. In contrast, depletion of CD8+ T-cells starting on day 10 still resulted in significant tumor elimination, suggesting that peak effector activity occurred during the first 10 days.

Figure 1. Long-term CD8+ T-effector cell activity in post-therapy mice.

Figure 1

Panel A. CD8 depletion in the surgical metastasis model. The schematic outlines the surgical metastasis model and the different CD8+ T-cell depletion timelines. Tumor bearing mice (200 mm3) with established lung metastases were treated with a single intratumoral injection of IL-12/GM-CSF microspheres on day 0. Treated primary tumors were then resected on day 7. Mice were sacrificed on day 28, lungs were removed, processed into single cell suspensions and plated in culture. Tumor colonies growing in plates were quantified on day 42. Panel B. Results of the clonogenic metastasis assay. Lung tumor burden from mice in different groups are shown. In the untreated group tumors were resected upon reaching 200 mm3 in volume without treatment. Error bars = SE, n = 5–6 mice per group. Assumption of homogeneity of variance among groups was tested by ANOVA and satisfied (p < 0.05).

Intratumoral IL-12 + GM-CSF induces concurrent expansion of effector and suppressor T-cells

The above findings suggested that treatment-induced effector and/or memory CD8+ T-cells were not effective against tumors that persisted beyond 10 days. Previous studies had demonstrated that treatment was followed by a rebound in intratumoral CD4+ CD25+ Foxp3+ T-suppressor cells (12). We therefore hypothesized that CD8+ T-cell priming could be paralleled by CD4+ Foxp3+ T-cell expansion, potentially curbing long-term T-effector cell activity in the tumor. To this end, CD8 and Foxp3 mRNA kinetics were monitored in the TDLN and tumors of treated mice via qRT-PCR. The results are shown in Figure 2A. Consistent with previous findings (11), treatment was followed by a 4-fold enhancement of CD8 mRNA in the TDLN on day 3 and an 8-fold increase in the tumor on day 7. Importantly, a similar increase (3-fold) in Foxp3 mRNA in the TDLN on day 3 was followed by a 2-fold increase in intratumoral Foxp3 on day 7. The increase in Foxp3 expression in the TDLN was slightly delayed when compared to CD8 during the first 24 hours suggesting that CD8+ T-cell-priming preceded T-suppressor cell expansion. Collectively, these data demonstrate that treatment-induced CD8+ T-cell priming and proliferation was accompanied by a similar expansion of T-suppressor cells in the TDLN and that both populations eventually homed to tumors.

Figure 2. Effector and suppressor activity in the tumors and the TDLN of post-therapy mice.

Figure 2

Panel A. Quantitative analysis of CD8 and Foxp3 mRNA expression. Total RNA was isolated from the tumors and the TDLN of mice on day 0 (pre-treatment), at 6 hours post-treatment and on days 1, 3, 5 and 7. The mRNAs for CD8 and Foxp3 were then quantified using real-time PCR analysis. The fold-change in mRNA level as normalized to day 0 is shown. Error bars = SE, n = 4 per group. For CD8 the differences between day 0 and days 5 or 7 were significant in tumors (p ≤ 0.024), and between day 0 and days 3 or 7 in the TDLN (p ≤ 0.009). For Foxp3, the differences between day 0 and days 3 or 7 were significant in the TDLN (p ≤ 0.012) and between day 0 and day 7 in the tumor (p = 0.033). Panel B. Quantitative analysis of IFNγ and IDO mRNA expression. Fold-change in mRNA levels was normalized to day 0 (pre-therapy). For IDO the differences between day 0 and days 3 or 7 were significant in the TDLN (p ≤ 0.005), and between day 0 and 1 in the tumor (p = 0.02). For IFNγ the differences between day 0 and all other days were significant in the TDLN (p ≤ 0.04). In the tumor the differences between day 0 and days 1 or 3 were significant (p ≤ 0.05). Error bars = SE, n = 4 per time point. Panel C. IDO induction in BALB/c versus GKO mice. Fold-changes in mRNA levels one day after treatment are shown. The differences between wild-type and GKO mice were significant in both the tumor and the TDLN (p = 0.004 and 0.043, respectively). Error bars = SE, n = 4 per group. The data are representative of 2 separate studies.

Treatment promotes rapid IFNγ-dependent IDO expression

The overlapping nature of post-therapy CD8 and Foxp3 mRNA expression patterns in the TDLN and tumors raised the possibility that treatment itself was responsible for the T-suppressor cell rebound. IFNγ is the primary downstream cytokine induced by IL-12 and is central to its antitumor effects (21). Among more than 200 immunologically-relevant genes induced by IFNγ is IDO (2224), a tryptophan-catabolizing enzyme with immune suppressive function (25). Importantly, recent studies demonstrated a role for IDO in CD4+ Foxp3+ T-suppressor cell activation and generation (26, 27). These findings provided the rationale for the next series of experiments in which the putative link between the IL-12-IFNγ-IDO axis and post-therapy immune suppression was investigated. Initially, expression of IFNγ and IDO were monitored in the TDLN and the tumors of treated mice by qRT-PCR. Treatment induced a rapid increase in IFNγ mRNA both in the TDLN and the tumor (Figure 2B). This enhancement was more dramatic in the TDLN (5- and 34-fold increases at 6 and 24 hours over background, respectively) compared to the tumor (2- and 3-fold enhancement at 6 and 24 hours, respectively). Upregulation of IFNγ expression in tumors was transient and declined rapidly after day 1 returning to background levels on day 7. IFNγ mRNA levels also declined in the TDLN but remained above pre-therapy levels for at least 7 days. Importantly, treatment promoted a similar increase in the expression of IDO (referred to as IDO-1 from this point on) as well as the more recently described IDO-2 (28) in the TDLN except that induction of these enzymes was delayed by 24 hours in comparison to IFNγ. In contrast, treatment resulted in the upregulation of IDO-1 but not IDO-2 in tumors (Figure 2B). Whereas IDO-1 mRNA returned to background levels in the tumor on day 3, both IDO-1 and 2 remained high in the TDLN for at least 7 days.

The sequential nature of IFNγ and IDO-1/IDO-2 expression kinetics in the TDLN between days 0 and 2 suggested that their transcription were linked. To test this hypothesis, tumors were induced in either wild-type or IFNγ-knockout (GKO) mice, and IDO-1 mRNA was quantified in pre- and post-therapy tumors and TDLN. The results are shown in Figure 2C. In wild-type mice treatment promoted 5 and 2-fold increases in IDO-1 mRNA in the tumor and the TDLN on day 1, respectively (Figure 2C). In contrast, treatment failed to induce detectable IDO-1 mRNA in the GKO mice (Figure 2C). Analysis of day 3 TDLN revealed a similar loss of IDO-2 induction in GKO mice demonstrating that IDO-2 upregulation was also IFNγ-dependent (data not shown).

Blocking IDO activity during treatment results in complete tumor eradication

The above findings were consistent with the notion that the IL-12-IFNγ-IDO axis represented a critical feedback inhibitory mechanism in our model. D-1MT, an inhibitor of IDO-2 activity, enhances tumor suppression in immune-competent but not in immune-deficient mice (18). To this end, we tested whether co-administration of D-1MT with IL-12 and GM-CSF to mice could enhance tumor suppression by blocking post-therapy IDO activity. Tumor-bearing mice were treated either with IL-12/GM-CSF microspheres, D-1MT alone or a combination of IL-12/GM-CSF + D-1MT and tumor growth was monitored. The results are shown in Figure 3. In the control group (blank microspheres) all tumors grew rapidly and mice had to be sacrificed by day 21 or earlier. Administration of D-1MT alone resulted in delayed tumor growth during the first week but tumors grew rapidly thereafter. Administration of IL-12/GM-CSF microspheres suppressed tumor growth during the first 2 weeks but all tumors eventually resumed growth. In contrast, complete tumor regression was achieved in 5 of 11 mice in the combination group resulting in long-term cure (see Supplemental Figure 1 for survival analysis). Since both IDO-1 and 2 were upregulated in the TDLN in response to treatment, we also tested the antitumor efficacy of L1-MT, which specifically targets IDO-1. L1-MT also delayed tumor growth when administered in combination with IL-12/GM-CSF therapy however complete regressions were achieved only with D1-MT suggesting that IDO-2 was the primary mediator of post-therapy feedback inhibition in this model (Supplemental Figure 2).

Figure 3. Effect of D-1MT co-administration on antitumor efficacy of IL-12/GM-CSF microsphere therapy.

Figure 3

Tumors were induced and allowed to grow to ~40mm3 in size. They were then treated with a single intratumoral injection of IL-12/GM-CSF microspheres. D-1MT was administered once intratumorally on day 0 (2mg/ml in 0.05 ml with microspheres) and orally in drinking water (2 mg/ml) between days -2 and 14. Control mice received a single intratumoral injection of blank microspheres. Each line represents a single mouse. Control and D-1MT groups had 6 mice each whereas IL-12/GM-CSF alone and the combination groups had 11 mice each. Tumors regressed completely in 5 of 11 mice in the combination treatment group. The data shown are representative of 3 separate experiments. Log-Rank analysis of survival curves demonstrated that the difference between IL-12/GM-CSF-alone and IL-12/GM-CSF + D1-MT groups was significant (p = 0.022, Supplemental Figure 1).

D-1MT administration delays CD4+ Foxp3+ T-suppressor cell expansion and extends CD8+ T-effector activity window

To determine whether IDO-2 activity and CD4+ Foxp3+ T-cell expansion were linked, post-therapy T-suppressor cell numbers were monitored in the presence or absence of D-1MT. The results shown in Figure 4A reveal that IL-12/GM-CSF treatment resulted in a dramatic 6-fold expansion of T-suppressor cells in tumors and a more modest 1.4-fold increase in the TDLN between days 3 and 10. Importantly, co-administration of D-1MT resulted in a significant reduction (1.8 to 6-fold) in the numbers of CD4+ Foxp3+ T-cells in the TDLN and the tumors on days 3 and 7. However, this inhibition was transient and the CD4+ Foxp3+ T-cell numbers in the experimental mice rebounded to levels observed in control mice on day 10. These findings establish that D-1MT transiently inhibited post-therapy T-suppressor cell expansion.

Figure 4. Effect of D-1MT co-administration on post-treatment T-cell kinetics in mice receiving IL- 12/GM-CSF therapy.

Figure 4

Panel A. CD4+ Foxp3+ T-suppressor cell expansion in post-therapy mice. Mice bearing tumors were treated either with IL-12/GM-CSF alone (control) or IL-12/GM-CSF + D-1MT (D-1MT). Tumors and TDLN were harvested on days 3, 7 and 10 and T-suppressor cells were quantified. The differences between control and D-1MT groups were significant on days 3 and 7 in the TDLN (p ≤ 0.031) and on day 3 in the tumor (p = 0.036). The dot-plots represent typical results (CD45+ CD3+ cells were gated on and analyzed for CD4 and Foxp3 expression) on day 7. Panel B. Analysis of proliferating CD8+ T-cells. The above samples were also analyzed for proliferating CD8+ T-cell populations using the BrdU pulse-labeling strategy. CD45+ CD3+ cells were gated on and analyzed for CD8 and BrdU staining. The differences between the control and D-1MT samples for day 7 TDLN (p = 0.006) and for day 10 tumors (0.026) were significant. Error bars = SE, n = 4–5 mice per group. This experiment was repeated twice with similar results.

Since inhibition of T-suppressor cell expansion is expected to result in enhanced CD8+ T-cell activity, the effect of IDO-2 inhibition on CD8+ T-cell proliferation was monitored. To this end, CD8+ T-cell proliferation was quantified in both the TDLN and tumors of control and D-1MT-treated mice. The results are shown in Figure 4B. These data demonstrate that initially TDLN CD8+ T-cells proliferated equally well in both control and D-1MT-treated mice independent of Foxp3+ T-cell levels. On day 7 however, CD8+ T-cell proliferation declined in the TDLN of control mice but remained high in D-1MT-treated animals consistent with extended priming activity. On day 10, proliferation returned to background levels in both groups. In tumors CD8+ T-cell proliferation on day 3 was minimal in both groups. On both days 7 and 10 however, CD8+ T-cells incorporated BrdU more vigorously (>3-fold higher) in the D-1MT group demonstrating that CD8+ T-effectors continued to proliferate in the tumors for at least until day 10. These data establish that the diminished T-suppressor cell expansion correlated with extended CD8+ T-cell activity.

Ratio of actively dividing CD8+ T-cells to T-suppressor cells predicts therapeutic efficacy

Several studies showed a correlation between intratumoral CD8+ T-cell to CD4+ Foxp3+ T-cell ratio and tumor progression (29, 30). Since D-1MT administration resulted in reduced T-suppressor cell numbers and enhanced CD8+ T-cell proliferation, the BrdU+ CD8+ T-cell to CD4+ Foxp3+ T-cell ratio was evaluated as a potential prognostic marker for tumor regression. To this end, the ratios were determined between days 0 and 10 in the TDLN and the tumors of control (IL-12/GM-CSF alone) and D-1MT (IL-12/GM-CSF + D-1MT) groups. These data are presented in Figure 5A. The results show that treatment did not significantly change the ratio in control mice between days 0 and 7 in the TDLN or tumors. On day 10 however, the ratio declined by 2-fold suggestive of the development of a more immune suppressive microenvironment. In contrast, the ratio of actively-dividing CD8+ T-cells to T-suppressor cells increased dramatically in both the TDLN and the tumors on days 3 and 7 (2 to 3-fold in the TDLN and 6 to 15-fold in the tumor compared to day 0) in the D-1MT-treated group. Although the ratio declined in both microenvironments on day 10 it still remained above pre-therapy levels.

Figure 5. Ratio of proliferating CD8+ T-cells to CD4+ Foxp3+ T-suppressor cells as a prognostic marker of therapeutic efficacy.

Figure 5

Panel A. Kinetics of CD8+ T-cell to T-suppressor cell ratio in mice treated with IL-12/GM-CSF alone (control) or IL-12/GM-CSF + D-1MT (D1-MT). TDLN and tumors were harvested from mice on indicated days after treatment and the ratio of CD8+ BrdU+ T-cells to CD4+Foxp3+ T-suppressor cells was determined. Day 0 data are from untreated mice. The differences between day 0 and all other time points were significant (p ≤ 0.013) in the TDLN. The differences between day 0 and days 3 or 7 were significant (p ≤ 0.046) in the tumor. Error bars = SE, n = 4–5 mice per group. Panel B. Correlation of CD8+ BrdU+ to CD4+ Foxp3+ T-cell ratio with tumor suppression. Mice were treated with IL-12/GM-CSF alone (control) or with cytokines plus D-1MT. Tumors were harvested on day 7 after treatment, weighed and the ratio of CD8+ BrdU+ to CD4+ Foxp3+ T-cells was determined for each tumor. The ratio was then plotted against tumor weight for each individual. Correlation coefficients (r) for control and D-1MT-treated groups were 0.637 and 0.881, respectively (n = 8 and 9).

The absolute values for total or proliferating CD8+ T-cell to CD4+ Foxp3+ T-suppressor cell ratios in tumors were plotted against tumor weight to determine whether the ratio correlated with tumor size. More specifically, the correlation coefficients (r-values) were determined in control and D-1MT groups on days 3, 7 and 10 (Supplemental Figure 3). Of these, the r-values for proliferating CD8+ T-cell to CD4+ Foxp3+ T-cell ratio on days 3 and 7 were most predictive. Data for day 7 tumors with or without D-1MT are shown in Figure 5B. The correlation between BrdU+ CD8+ T-cell to CD4+ Foxp3+ ratio and tumor suppression was significant in both groups with the D-1MT group displaying stronger linkage. In contrast, total CD8+ T-cell to CD4+ Foxp3+ T-cell ratio was found to be less predictive (Table 1).

Table 1.

The ratio of proliferating CD8+ T-cells to CD4+ Foxp3+ T-cells is prognostic for therapeutic efficacy.

T-cell ratio (cells/gram tumor) IL-12/GM-CSF D1-MT correlation coefficient (r)
Total CD8+ / CD4+ Foxp3+ + 0.155
Total CD8+ / CD4+ Foxp3+ + + 0.635
BrdU+ CD8+/ CD4+ Foxp3+ + 0.64
BrdU+ CD8+ / CD4+ Foxp3+ + + 0.88

T-suppressor cell rebound is driven primarily by IL-12

Since IL-12 and GM-CSF were found to be synergistic in mediating tumor regression in previous studies (19) we wanted to determine their respective roles in the observed feedback inhibition. Combined administration of IL-12 + GM-CSF promoted 5.1 ± 0.45 and 5.2 ± 1.01-fold increases in intratumoral IFNγ and IDO-1 mRNA on day 1, respectively. IL-12 alone resulted in similar increases in both IFNγ (4.5 ± 1.27-fold) and IDO-1 (3.1 ± 0.2-fold) expression. In contrast, GM-CSF alone did not promote any significant increases in IFNγ (1.3 ± 0.04-fold) or IDO-1 (0.7 ± 0.17) expression (n = 3–4 mice per group). With regard to the T-suppressor cell rebound between days 3 and 7, combination, IL-12-alone, GM-CSF-alone and control (blank microsphere) groups demonstrated 13.9 ± 4.7, 6.0 ± 0.2, 2.5 ± 0.6 and 2.3 ± 0.4-fold increases in T-suppressor cell numbers, respectively. These data demonstrate that T-suppresssor cell expansion was driven by IL-12. GM-CSF appeared to contribute to the T-suppressor cell rebound when administered with IL-12, but had no effect when injected alone.

Discussion

The above studies demonstrate that the pro-inflammatory activity of IL-12 is regulated via a feedback inhibitory mechanism involving the IFNγ-IDO-T-suppressor cell axis. Our results also establish that blocking of IDO activity with D-1MT can be effectively employed to overcome feedback inhibition and enhance IL-12-mediated antitumor responses. Finally, the above data identify the proliferating CD8+ T-cell to CD4+ Foxp3+ T-cell ratio in tumors as a highly accurate prognostic marker of therapeutic efficacy.

Numerous clinical studies have demonstrated the tachyphylactic nature of IL-12 therapy (3134). However, the mechanism underlying the progressive loss of therapeutic efficacy that accompanies repeated IL-12 administration has not been defined. Our findings, for the first time, demonstrate that IDO activation is, at least in part, responsible for this effect. Whether suppression occurred primarily via T-suppressor cell activity (26, 27) or also involved direct inhibition by tryptophan catabolites (35, 36) was not investigated. In our model, induction of IDO-1 in the tumor microenvironment was rapid and transient with no change in intratumoral IDO-2. These data, combined with our previous finding that tumor-resident CD8+ Tem display full-effector function during this interval (10), are inconsistent with a direct role for tryptophan catabolites in T-effector inhibition. To this end, it was recently reported that IDO-1 overexpression does not inhibit the effector function of pre-existing Tem (36). In contrast, the kinetics of intratumoral T-suppressor cell expansion between days 3 and 10 closely paralleled the loss of post-therapy CD8+ T-effector cytotoxicity in tumors (10). This finding, combined with the well-known ability of T-suppressor cells to inhibit CD8+ T-cell cytotoxicity in vivo (37), suggest that the inhibitory activity of IDO was mediated indirectly via T-suppressor cells.

The sources of post-treatment IDO-1 and IDO-2 were not investigated in this study. Numerous reports have established that plasmacytoid Dendritic Cells (DC), conventional tolerogenic DC and Macrophages are the primary producers of IDO-1 (3840). IDO-2, on the other hand, is expressed in the kidney, testis and liver, but has also been detected in dendritic cells (28). Our preliminary findings are consistent with the hypothesis that DC are the primary source of IDO-1 and IDO-2 in the TDLN of post-IL-12 mice (Harden and Egilmez, unpublished data). The mechanistic basis of the differential IDO-1 and IDO-2 expression patterns in post-treatment tumors and TDLN, on the other hand, remain undefined. In the case of IDO-1, treatment-induced migration of DC from tumors to TDLN (41) can potentially account for the initial intratumoral spike and the subsequent upregulation and persistence of IDO-1 in the TDLN. The restriction of IDO-2 expression to the TDLN on the other hand, is more difficult to explain. One potential mechanism would involve selective infiltration of tumors and the TDLN by DC subsets with differential IFNγ responsiveness and/or IDO-1 or 2 expression profiles (42).

The finding that D-1MT was more efficient than L-1MT in achieving complete tumor eradication in IL-12/GM-CSF-treated mice is consistent with others' findings in different tumor models (18). The mechanistic basis of the in vivo superiority of D-1MT to L-1MT in promoting tumor suppression is controversial (18, 43, 44). In our study, treatment promoted both IDO-1 and IDO-2 expression in the TDLN demonstrating that the differential antitumor efficacy of the 1-MT isomers was not due to selective expression of the target enzymes in post-treatment TDLN. On the other hand, whether IDO-1 and IDO-2 differ in their abilities to induce T-suppressor cell activation/expansion, and whether they are expressed by different tolerogenic DC subsets is not known. Analysis of post-therapy TDLN DC subsets with regard to IDO-1 and 2 expression and defining their relative abilities to activate/expand T-suppressor cells may shed further light on the respective roles of each enzyme in the development of post-therapy regulatory rebound.

The above results suggest that selective blocking of IFNγ-mediated regulatory pathways can overcome IL-12 tachyphylaxis. Whereas repeated activation of tumor-resident T-effector/memory cells via this strategy represents a potentially effective therapeutic approach, its utility may still be limited by T-cell intrinsic regulatory mechanisms (9) and/or the finite clonal proliferative potential of cytotoxic CD8+ T-cells (45). We are currently investigating the limits of this strategy in a model that allows long-term quantitative monitoring of tumor-specific CD8+ T-effector cell activity.

Supplementary Material

1
NIHMS157637-supplement-1.eps (1,003.1KB, eps)
2
3

Acknowledgements

We thank Dr. Stanley Wolf of Wyeth, Inc. for providing the murine IL-12 and his continued support of our studies.

This work was supported by the NIH grant R01-CA100656-01A1 and the NY State Office of Science Technology and Academic Research (NYSTAR) faculty recruitment award C040070 to NKE

References

  • 1.Morse MA, Chui S, Hobeika A, et al. Recent developments in therapeutic cancer vaccines. Nature Clin Practice Oncol. 2005;2:108–13. doi: 10.1038/ncponc0098. [DOI] [PubMed] [Google Scholar]
  • 2.Finn OJ. Molecular origins of cancer. Cancer Immunology. New Engl J Med. 2008;358:2704–15. doi: 10.1056/NEJMra072739. [DOI] [PubMed] [Google Scholar]
  • 3.Rosenberg SA, Sherry RM, Morton KE, et al. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. J Immunol. 2005;175:6169–76. doi: 10.4049/jimmunol.175.9.6169. [DOI] [PubMed] [Google Scholar]
  • 4.Gajewski TF, Meng Y, Harlin H. Immune suppression in the tumor microenvironment. J Immunother. 2006;29:233–40. doi: 10.1097/01.cji.0000199193.29048.56. [DOI] [PubMed] [Google Scholar]
  • 5.Vieweg J, Su Z, Dahm P, Kusmartsev S, et al. Reversal of tumor-mediated immunosuppression. Clin Cancer Res. 2007;13:727s–32s. doi: 10.1158/1078-0432.CCR-06-1924. [DOI] [PubMed] [Google Scholar]
  • 6.Schlom J, Gulley JL, Arlen PM. Paradigm shifts in cancer vaccine therapy. Exp Biol Med. 2008;233:522–34. doi: 10.3181/0708-MR-226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol. 2009;27:83–117. doi: 10.1146/annurev.immunol.021908.132544. [DOI] [PubMed] [Google Scholar]
  • 8.Badovinac VP, Porter BB, Harty JT. Programmed contraction of CD8+ T cells after infection. Nature Immunol. 2002;3:619–26. doi: 10.1038/ni804. [DOI] [PubMed] [Google Scholar]
  • 9.Blackburn SD, Shin H, Haining WN, et al. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nature Immunol. 2009;10:29–37. doi: 10.1038/ni.1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kilinc MO, Aulakh KS, Nair RE, et al. Reversing tumor immune suppression with intratumoral IL-12: Activation of tumor-associated T effector/memory cells, induction of T suppressor apoptosis, and infiltration of CD8+ T effectors. J Immunol. 2006;177:6962–73. doi: 10.4049/jimmunol.177.10.6962. [DOI] [PubMed] [Google Scholar]
  • 11.Kilinc MO, Gu T, Harden JL, et al. Central role of tumor-associated CD8+ T-effector/memory cells in restoring systemic anti-tumor immunity. J Immunol. 2009;182(7):4217–25. doi: 10.4049/jimmunol.0802793. [DOI] [PubMed] [Google Scholar]
  • 12.Nair RE, Kilinc MO, Jones SA, et al. Chronic immune therapy induces a progressive increase in intratumoral T suppressor activity and a concurrent loss of tumor-specific CD8+ T effectors in her-2/neu transgenic mice bearing advanced spontaneous tumors. J Immunol. 2006;176:7325–34. doi: 10.4049/jimmunol.176.12.7325. [DOI] [PubMed] [Google Scholar]
  • 13.Bynoe MS, Evans JT, Viret C, et al. Epicutaneous immunization with autoantigenic peptides induces T suppressor cells that prevent experimental allergic encephalomyelitis. Immunity. 2003;19:317–28. doi: 10.1016/s1074-7613(03)00239-5. [DOI] [PubMed] [Google Scholar]
  • 14.Apostolou I, von Boehmer H. In vivo instruction of suppressor commitment in naïve T cells. J Exp Med. 2004;199:1401–8. doi: 10.1084/jem.20040249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhou G, Drake CG, Levitsky HI. Amplification of tumor-specific regulatory T cells following therapeutic cancer vaccines. Blood. 2006;107:628–36. doi: 10.1182/blood-2005-07-2737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wu RS, Kobie JJ, Besselsen CG, et al. Comparative analysis of IFN-γ B7.1 and antisense TGF-β gene transfer on the tumorigenicity of a poorly immunogenic metastatic mammary carcinoma. Cancer Immunol Immunother. 2001;50:229–40. doi: 10.1007/s002620100197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gu T, Kilinc MO, Egilmez NK. Transient activation of tumor-associated T-effector/memory cells promotes tumor eradication via NK-cell recruitment: minimal role for long-term T-cell immunity in cure of metastatic disease. Cancer Immunol Immunother. 2008;57:997–1005. doi: 10.1007/s00262-007-0430-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hou D-Y, Muller AJ, Sharma MD, et al. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 2007;67:792–801. doi: 10.1158/0008-5472.CAN-06-2925. [DOI] [PubMed] [Google Scholar]
  • 19.Hill HC, Conway TF, Sabel MS, et al. Cancer Immunotherapy with Interleukin-12 and Granulocyte-Macrophage Colony-Stimulating Factor-encapsulated microspheres: Coinduction of innate and adaptive immunity and cure of disseminated disease. Cancer Res. 2002;62:7254–63. [PubMed] [Google Scholar]
  • 20.McLean M, Wallace HL, Sharma A, et al. A murine surgical metastasis model for the evaluation of anticancer strategies. Clinical and Experimental Metastasis. 2004;21(4):363–9. doi: 10.1023/b:clin.0000046176.33867.c5. [DOI] [PubMed] [Google Scholar]
  • 21.Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nature Rev Immunol. 2003;3:133–46. doi: 10.1038/nri1001. [DOI] [PubMed] [Google Scholar]
  • 22.Taylor MW, Feng G. Relationship between interferon-γ, indoleamine 2,3-dioxygenase, and tryptophan catabolism. FASEB J. 1991;5:2516–22. [PubMed] [Google Scholar]
  • 23.Chon SY, Hassanain HH, Gupta SL. Cooperative role of interferon regulatory factor 1 and p91 (STAT1) response elements in interferon-γ-inducible expression of human indoleamine 2,3-dioxygenase gene. J Biol Chem. 1996;271:17247–52. doi: 10.1074/jbc.271.29.17247. [DOI] [PubMed] [Google Scholar]
  • 24.Boehm U, Klamp T, Groot M, et al. Cellular responses to interferon-γ. Annu Rev Immunol. 1997;15:749–95. doi: 10.1146/annurev.immunol.15.1.749. [DOI] [PubMed] [Google Scholar]
  • 25.Mellor AL, Munn DH. IDO expression by dendritic cells: Tolerance and tryptophan catabolism. Nature Rev Immunol. 2004;4:762–74. doi: 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
  • 26.Sharma MD, Baban B, Chandler P, et al. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest. 2007;117:2570–82. doi: 10.1172/JCI31911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen W, Liang X, Peterson AJ, et al. The indoleamine 2,3-dioxygenase pathway is essential for human plasmacytoid dendritic cell-induced adaptive T regulatory cell generation. J Immunol. 2008;181:5396–5404. doi: 10.4049/jimmunol.181.8.5396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ball HJ, Yuasa HJ, Austin CJD, et al. Indoleamine 2,3-dioxygenase-2; a new enzyme in the kynurenine pathway. Int J Biochem and Cell Biol. 2009;41:467–71. doi: 10.1016/j.biocel.2008.01.005. [DOI] [PubMed] [Google Scholar]
  • 29.Sato E, Olson SH, Ahn J, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci. 2005;102:18538–43. doi: 10.1073/pnas.0509182102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bui JD, Uppaluri R, Hsieh C-S, et al. Comparative analysis of regulatory and effector T cells in progressively growing versus rejecting tumors of similar origins. Cancer Res. 2006;66:7301–9. doi: 10.1158/0008-5472.CAN-06-0556. [DOI] [PubMed] [Google Scholar]
  • 31.Atkins MB, Robertson MJ, Gordon M, et al. Phase I evaluation of intravenous recombinant human interleukin 12 in patients with advanced malignancies. Clin Cancer Res. 1997;3:409–17. [PubMed] [Google Scholar]
  • 32.Motzer RJ, Rakhit A, Schwartz LH, et al. Phase I trial of subcutaneous recombinant human interleukin-12 in patients with advanced renal cell carcinoma. Clin Cancer Res. 1998;4:1183–91. [PubMed] [Google Scholar]
  • 33.Gollob JA, Mier JW, Veenstra K, et al. Phase I trial of twice-weekly intravenous interleukin 12 in patients with metastatic renal cell cancer or malignant melanoma: Ability to maintain IFN-γ induction is associated with clinical response. Clin Cancer Res. 2000;6:1678–92. [PubMed] [Google Scholar]
  • 34.Haicheur N, Escudier B, Dorval T, et al. Cytokines and soluble cytokine receptor induction after IL-12 administration in cancer patients. Clin Exp Immunol. 2000;119:28–37. doi: 10.1046/j.1365-2249.2000.01112.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Belladonna ML, Grohmann U, Guidetti P, et al. Kynurenine pathway enzymes in dendritic cells initiate tolerogenesis in the absence of functional IDO. J Immunol. 2006;177:130–7. doi: 10.4049/jimmunol.177.1.130. [DOI] [PubMed] [Google Scholar]
  • 36.Liu Z, Dai H, Wan N, et al. Suppression of memory CD8 T cell generation and function by tryptophan catabolism. J Immunol. 2007;178:4260–6. doi: 10.4049/jimmunol.178.7.4260. [DOI] [PubMed] [Google Scholar]
  • 37.Chen ML, Pittet MJ, Gorelik L, et al. Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc Natl Acad Sci. 005;102(2):419–24. doi: 10.1073/pnas.0408197102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hwu P, Du MX, Lapointe R, Do M, et al. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol. 2000;164:3596–9. doi: 10.4049/jimmunol.164.7.3596. [DOI] [PubMed] [Google Scholar]
  • 39.Munn DH, Sharma MD, Hou D, et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114:280–90. doi: 10.1172/JCI21583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Munn DH, Shafizadeh E, Attwood JT, et al. Inhibition of T cell proliferation by Macrophage tryptophan catabolism. J Exp Med. 1999;189(9):1363–72. doi: 10.1084/jem.189.9.1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Guiducci C, Vicari AP, Sangaletti S, et al. Redirecting in vivo elicited tumor infiltrating macrophages and dendritic cells towards tumor rejection. Cancer Res. 2005;65(8):3437–46. doi: 10.1158/0008-5472.CAN-04-4262. [DOI] [PubMed] [Google Scholar]
  • 42.Kim YH, Choi BL, Kang WJ, et al. IFN-γ-indoleamine 2,3 dioxygenase acts as a major suppressive factor in 4-1BB-mediated immune suppression in vivo. J Leukoc Biol. 2009;85:817–25. doi: 10.1189/jlb.0408246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lob S, Konigsgrainer A, Schafer R, et al. Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells. Blood. 2008;111(4):2152–54. doi: 10.1182/blood-2007-10-116111. [DOI] [PubMed] [Google Scholar]
  • 44.Lob S, Konigsgrainer A, Zieker D, et al. IDO1 and IDO2 are expressed in human tumors: levo- but not dextro-1-methyl tryptophan inhibits trytophan catabolism. Cancer Imm Immunother. 2009;58:153–7. doi: 10.1007/s00262-008-0513-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Masopust D, Ha S-J, Vezys V, Ahmed R. Stimulation history dictates memory CD8 T cell phenotype: Implications for prime-boost vaccination. J Immunol. 2006;177:831–9. doi: 10.4049/jimmunol.177.2.831. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
NIHMS157637-supplement-1.eps (1,003.1KB, eps)
2
3

RESOURCES