Indoleamine 2, 3-dioxygenase 1 (IDO1), IDO2, and tryptophan 2, 3-dioxygenase (TDO) comprise a family of enzymes that catalyze the first- and rate-limiting step associated with the catabolic conversion of tryptophan (Trp) into kynurenine (Kyn). Through subsequent enzymatic and spontaneous reactions, Kyn is further converted into the energetic substrates, NAD+ and ATP, to fuel cellular metabolic functions. Coincidently, the depletion of Trp and accumulation of Kyn has been demonstrated to induce effector T-cell apoptosis/dysfunction and immunosuppressive regulatory T-cell induction, respectively. Similar to other immune checkpoints, IDO1 and TDO are suggested to be important targets for immunotherapeutic intervention. This is represented by the recent growth of efforts to inhibit the Trp-to-Kyn pathway as a means to control immunosuppression. Inhibitors currently in clinical trials, INCB024360, GDC-0919, indoximod, and an IDO1 peptide-based vaccine, are being evaluated for their efficacy against a wide range of cancers including melanoma, glioblastoma, non–small cell lung, pancreatic, and/or breast cancer, as well as metastatic disease. Despite the rapid development of potent clinical grade inhibitors, strategic questions remain. Here, we review the state of the literature with respect to current therapeutic inhibitors of tryptophan catabolism, evaluation of those efforts preclinically and clinically, compensatory changes that occur with therapeutic targeting, as well as newly recognized signaling features that raise critical questions to the field. Given the rapidly evolving interest in determining how IDO1/TDO, and to an unknown extent, IDO2, can be targeted for increasing cancer immunotherapeutic efficacy, we present a brief but comprehensive analysis that addresses critical questions, while highlighting the mechanics that remain to be explored. Clin Cancer Res; 21(24); 5427–33. ©2015 AACR.

Cancer immunology and immunotherapy

The immune system is composed of an immediate-acting innate arm comprised principally of granulocyte- and myeloid-lineage cells that quickly respond to cues of inflammation and/or injury, in addition to an adaptive arm, principally comprised of B and T cells that provide specificity and memory. Under normal circumstances, these immunologic arms are mutually dependent on one another for providing defense against infection, injury, and/or malignancy. T cells, which primarily mature following immunologic challenge(s), include CD4+ and CD8+ T lymphocytes that express a wide variety of cytokines based on the context of priming stimuli. Included in the CD4+ T-cell compartment are highly immunosuppressive regulatory T cells (Treg; CD4+CD25+FoxP3+CTLA-4+) that mature naturally in the thymus (nTreg) or are post-thymically induced from naïve CD4+Foxp3 cells into Foxp3-expressing cells (iTreg; refs. 1–3). With respect to solid cancer(s), immunosuppressive mechanisms utilized to evade antitumor immunity include Treg accumulation (4, 5) effector T-cell expression of the PD-1 receptor (6), as well as high PD-L1 levels that localize to multiple types of cells in the tumor microenvironment (7, 8). Therefore, an active effort both clinically and preclinically is needed to develop strategies that reactivate a productive antitumor effector T-cell response, while simultaneously inhibiting immunosuppressive mechanisms.

Recent studies have demonstrated great promise at targeting immunosuppression in cancer, including clinical trials aimed at inhibiting PD-1, PD-L1, and/or CTLA-4 in patients diagnosed with late-stage melanoma, non–small cell lung cancer, and/or renal cell cancer (9–12). Follow-up studies have also shown that the benefit of combined PD-1/CTLA-4 inhibition is not restricted to those patients previously treated with systemic therapy (13). Preclinical work using multiple tumor models in immunocompetent mice further confirms that these immune checkpoint–targeted therapies require effector T cells for antitumor activity, with several studies reporting a coincident neutralization of tumor-infiltrating Treg (14–16). These clinical studies, combined with extensive preclinical validation of combinatorial approaches confirm that immunotherapy is a high-value strategy for treating patients with aggressive and immunosuppressive malignancies.

IDO1, TDO, and the Trp→Kyn catabolic pathway

L-Tryptophan (L-Trp) is used in a variety of anabolic/catabolic processes and metabolized into serotonin, melatonin, protein, and Kyn. IDO1 and TDO are the primary enzymes that catalyze the rate-limiting cleavage of the Trp indole ring 2,3-double bond and incorporation of molecular oxygen. The product of this reaction is N-formylkynurenine, which is rapidly and spontaneously converted into L-Kyn. The latter catabolite is further converted into downstream intermediates, including 3-hydroxy-L-kynurenine (3-HK), 3-hydroxyanthranilate (3-HAA) and quinolinic acid (Quin), which also impact immune responses (17).

Although IDO1 and TDO both catabolize Trp, their quaternary structures (18, 19), expression in normal versus transformed tissue (20, 21) and regulation (22, 23) are quite distinct. While monomeric IDO1 acts on a broad range of substrates and is capable of cleaving both D- and L-Trp, homotetrameric TDO is enantiomer-specific and only catabolizes L-Trp (24). IDO1 expression in adults is relatively limited to lymphoid tissues and placenta (20), whereas TDO is constitutively expressed in liver and brain (25, 26), likely reflecting their primarily immunomodulatory or energy regulating roles, respectively. Until 2007, IDO1 was the only known indoleamine dioxygenase acting at the 2, 3 double bond. Three independent groups then identified the novel paralog, IDO2 (27–29). While the IDO1 and IDO2 genes are 43% homologous and found directly adjacent to one another on chromosome 8, the Km of human IDO1 and IDO2 for L-Trp is 20.90 ± 3.95 μmol/L and 6,809 ± 917 μmol/L, respectively, indicating a substantial decrease in activity for the latter enzyme (30). This is particularly interesting given that the residues required for tryptophan catalytic activity are present in both gene products (27). Also notable is that mouse IDO2 has been shown to possess higher enzymatic activity than the human homolog, although the genetic depletion of mouse IDO2 has no impact on systemic Kyn levels (31), a dramatic contrast to the impact of IDO1 deficiency (32).

IDO1 and the stress response

Because of IDO1 expression induced in response to infection, it was originally thought that it serves as an innate immune effector to restrict the amount of Trp required for microbial growth (33). This initial hypothesis was revised by Munn and colleagues, who demonstrated that the in vivo administration of an IDO1 inhibitor, 1-methyl tryptophan (1-MT), led to T-cell–dependent fetal allograft rejection (34). Subsequent work demonstrated that IDO1-expressing macrophages, dendritic cells (DC), and tumor cells mediate the inhibition of T-cell proliferation (35–38). IDO1 responses were found to be mediated by downstream stress–response pathways including general control non-depressible 2 (GCN2) and mTOR; both important regulators that sense amino acid sufficiency (Fig. 1). The GCN2 pathway is activated when amino acid deficiency increases overall uncharged tRNA levels, resulting in GCN2 kinase phosphorylation of the alpha subunit of translation initiation factor 2 α (eIF2α) and subsequent inhibition of translation. It was first discovered that GCN2-activated plasmacytoid DC could suppress T-cell proliferation in vivo by an IDO1-dependent mechanism (39). It was later discovered that the genetic deletion of IDO1, but not GCN2, prevented skin carcinogenesis in a mouse papilloma model, suggesting that additional critical pathways were downstream of IDO1 activity (40). In support of these findings, Metz and colleagues identified that IDO1-mediated Trp depletion suppressed mTOR, a critically important immunoregulatory kinase (40) that could be reactivated by treatment with D-1-MT, a Trp mimetic, in vitro.

Figure 1.

Signaling pathways associated with tryptophan (Trp) dioxygenases and cancer. The high expression of active IDO1 leads to a commensurately high rate of tryptophan conversion and depletion. This induces cell-cycle arrest and/or anergy in the effector cytotoxic lymphocyte (CTL) compartment via the eIF2a kinase-dependent GCN2 pathway. Simultaneously, this mechanism also contributes to the activation/maturation of Treg in association with CTLA4-mediated CD80/CD86 coinhibition. Kynurenine (Kyn) directly induces the apoptosis of CTL by an uncharacterized mechanism, while interacting with the aryl hydrocarbon receptor (AhR) in naïve CD4+ T cells, resulting in the induction of FoxP3+ iTreg. AhR interacts with the aryl hydrocarbon receptor nuclear translocator (ARNT) to mediate the specific transcriptional programming. Coincidently, IDO1 nonenzymatically enforces immunosuppression through two intrinsic immunoreceptor tyrosine-based inhibitory motifs (ITIM) in antigen-presenting cells (APC). TGFβ signaling results in the phosphorylation of the IDO1 ITIM, triggering noncanonical NF-κB activation and phosphorylation of IKKα, followed by nuclear translocation of the NF-κB subunits, p52 and RelB and autocrine reinforcement of IDO1 and TGFβ expression. The high-affinity Trp transporter is expressed by APC and tumor cells, with the majority of agonists leading to IDO1 activity demonstrated in APC. Similarly, the oncogene, c-KIT, and tumor-suppressor gene, Bin1, as well as the IL6/AhR/STAT3 signaling loop, have also been shown to impact the regulation of IDO1 in tumor cells. (39, 69–76). Notably, in the presence of IDO1/TDO, Kyn accumulation simultaneously contributes to Treg activation, promoting the disabling and/or apoptosis of CTL, thereby supporting tumor outgrowth by virtue of an unproductive antitumor response. Bin1, Myc box–dependent-interacting protein 1; EP2, prostaglandin receptor E2; Etv4, ETS translocation variant 4; GCN2, general control non-derepressible 2; GITRL, glucocorticoid-induced TNFR-related protein ligand; IRF1, IFN regulatory factor 1; IFNαR, IFNα receptor; IL6R, IL6 receptor; LPS, lipopolysaccharide; PGE2, prostaglandin E2; PKA, protein kinase A; PKCq, protein kinase C theta; SHP, SH2 domain containing protein tyrosine phosphatase; SOCS3, suppressor of cytokine signaling 3; TLR4, toll-like receptor 4; TNFαR, TNFα receptor.

Figure 1.

Signaling pathways associated with tryptophan (Trp) dioxygenases and cancer. The high expression of active IDO1 leads to a commensurately high rate of tryptophan conversion and depletion. This induces cell-cycle arrest and/or anergy in the effector cytotoxic lymphocyte (CTL) compartment via the eIF2a kinase-dependent GCN2 pathway. Simultaneously, this mechanism also contributes to the activation/maturation of Treg in association with CTLA4-mediated CD80/CD86 coinhibition. Kynurenine (Kyn) directly induces the apoptosis of CTL by an uncharacterized mechanism, while interacting with the aryl hydrocarbon receptor (AhR) in naïve CD4+ T cells, resulting in the induction of FoxP3+ iTreg. AhR interacts with the aryl hydrocarbon receptor nuclear translocator (ARNT) to mediate the specific transcriptional programming. Coincidently, IDO1 nonenzymatically enforces immunosuppression through two intrinsic immunoreceptor tyrosine-based inhibitory motifs (ITIM) in antigen-presenting cells (APC). TGFβ signaling results in the phosphorylation of the IDO1 ITIM, triggering noncanonical NF-κB activation and phosphorylation of IKKα, followed by nuclear translocation of the NF-κB subunits, p52 and RelB and autocrine reinforcement of IDO1 and TGFβ expression. The high-affinity Trp transporter is expressed by APC and tumor cells, with the majority of agonists leading to IDO1 activity demonstrated in APC. Similarly, the oncogene, c-KIT, and tumor-suppressor gene, Bin1, as well as the IL6/AhR/STAT3 signaling loop, have also been shown to impact the regulation of IDO1 in tumor cells. (39, 69–76). Notably, in the presence of IDO1/TDO, Kyn accumulation simultaneously contributes to Treg activation, promoting the disabling and/or apoptosis of CTL, thereby supporting tumor outgrowth by virtue of an unproductive antitumor response. Bin1, Myc box–dependent-interacting protein 1; EP2, prostaglandin receptor E2; Etv4, ETS translocation variant 4; GCN2, general control non-derepressible 2; GITRL, glucocorticoid-induced TNFR-related protein ligand; IRF1, IFN regulatory factor 1; IFNαR, IFNα receptor; IL6R, IL6 receptor; LPS, lipopolysaccharide; PGE2, prostaglandin E2; PKA, protein kinase A; PKCq, protein kinase C theta; SHP, SH2 domain containing protein tyrosine phosphatase; SOCS3, suppressor of cytokine signaling 3; TLR4, toll-like receptor 4; TNFαR, TNFα receptor.

Close modal

IDO1-mediated suppression of T-cell activity is hypothesized to rely on the depletion of free Trp. This premise requires cell-specific transport mechanisms that include both the transporter System L, which shuttles Trp and other large hydrophobic amino acids through a low-affinity (Km = 20–30 μmol/L; ref. 41) interaction, as well as through an independent high-affinity (Km = 200–300 nmol/L) interaction. Interestingly, the high-affinity transporter is upregulated in differentiated myeloid-derived macrophages (MDM) but not in T cells. In support of the requirement for transport, both Trp and the competitive inhibitor, L-1-MT, inhibit Trp uptake into cells, collectively suggesting that competitive IDO1 inhibitors target the transporter and enzyme, simultaneously.

Regulation of IDO1/IDO2/TDO

The literature is replete with redundant pathways that lead to IDO1 expression and activity. Proinflammatory signals including IFNγ, CpG DNA, and LPS are potent inducers of IDO1 expression (33, 42–44). Cytokines, including TNFα, IL6, and IL1β, synergize with each other to dramatically increase IDO1 expression. Other IDO1 modulators include soluble GITR, prostaglandin E2, the oncogene, c-Kit, as well as the tumor suppressor, Bin1 (45). Interesting new data suggests that Wnt5α also mediates IDO1 activity through β-catenin signaling in DC (46), while maintaining continuous expression through an AhR–IL6–STAT3 signaling loop in some cancer cell lines (47). Thus, based on the large number of pathways that modulate and/or sustain IDO1 expression/activity, the direct targeting of IDO1, rather than pathways that are up- or downstream, will likely be the most effective modality for controlling the overall impact mediated by this Trp dioxygenase.

Similar to IDO1, TDO mRNA expression has also been found in human tumors (21). Dominant factors that affect TDO expression and/or activity include sex steroid hormones (48) and glucocorticoids (22). New preclinical data also suggest that tumor-infiltrating T cells may regulate TDO expression based on findings from intracranially injected syngeneic murine brain tumors grown in Rag1−/− mice (15). Notably, intraperitoneal injection of mastocytoma cells overexpressing TDO induces potent immunosuppression that can be reversed with a pharmacologic inhibitor of enzymatic activity, leading to immune-mediated tumor rejection (P < 0.001; ref. 21).

In contrast, the newest member of the tryptophan catabolic family, IDO2, has yet to be confirmed as a critical contributor to Kyn accumulation and tumor immunity. Notably, while mouse IDO2 possesses some capacity for Trp→Kyn conversion, the human ortholog is devoid of the same enzymatic capacity at physiologic Trp levels (30). Furthermore, transcriptome analysis of 129 human tumor samples and 25 human tumor cell lines has demonstrated limited IDO2 expression (49). As IDO2 was originally cloned from the liver (27), it is still unknown whether there are IDO2 splice variants specific to subtypes of differentiated and/or transformed tissues.

IDO1 and inflammation in tumors

The interactions among inflammation, IDO1, and cancer (50, 51) are noteworthy and raise critical questions regarding how and when to optimally target tryptophan catabolism for therapeutic purposes. Furthermore, despite the presence of antigen-specific T cells within the microenvironment, tumors often escape, immunologically, without loss of antigen expression or presentation (MHC molecule) capacity. This effect is mediated, in-part, through the induction, upregulation, and/or enhanced participation of immunosuppressive T-cell–impairing ligands, CTLA-4 and PD-L1 (52). Similar to PD-L1, IDO1 expression also increases through a response to IFNγ released in the tumor microenvironment (53) as a potent compensatory mechanism contributing to the resistance of productive antitumor immunity (54). Interestingly, only a subset of patients have a T-cell–infiltrating presence within the tumor microenvironment, an observation reported for head and neck and bladder cancer, as well as melanoma, lung adenoma, and glioblastoma (55). A notable observation from those patients treated with the immune checkpoint inhibitor PD-1, correlates a high degree of clinical response to the pre-existence of tumor-infiltrating T cells (56). This observation, paired with the association of IDO1 induction by T-cell–derived IFNγ, leads to the hypothesis that IDO1 inhibitors will be most effective against T-cell–inflamed tumors, either de novo or caused by immunotherapeutic intervention. Preclinical studies support this hypothesis, establishing evidence that combinatorial immune checkpoint blockade and IDO1 pathway inhibition provide potent reactivation of tumor-infiltrating T cells and/or decreased tumor-resident immunosuppressive regulatory T cells (P < 0.01; refs. 15, 57).

No IDO1 inhibitor is currently approved by the FDA. However, results of recent phase I–II studies suggest that indoximod (D-1-MT), INCB024360, and/or IDO1-targeting vaccines are well tolerated by cancer patients, with clinical anticancer effects in a subset of patients (58, 59). Notably, the number of clinical trials focused on IDO1 has recently grown in size, with many coupling multiple modalities to test the combinatorial benefit (Table 1). These recent reports, in addition to preclinical data suggest that combining tryptophan enzyme targeting with chemotherapy, radiotherapy, and/or immunotherapy may be an effective tool against a wide range of malignancies.

Table 1.

Ongoing and historical clinical trials that target tryptophan catabolism in cancer

AgentIndication(s)PhaseStatusNotesIdentifier
Indoximod (D-1-MT) Metastatic solid tumor Completed Combined with docetaxel NCT01191216 
 Solid tumor Completed As single agent NCT00567931 
  Terminated As single agent NCT00739609 
 Malignant glioma I/II Recruiting For recurrent glioma patients NCT02052648 
 Metastatic breast cancer I/II Active, not recruiting Combined with vaccine NCT01042535 
  II Recruiting Combined with docetaxel NCT01792050 
 Melanoma I/II Recruiting Combined with ipilimumab NCT02073123 
 Metastatic adenoma of pancreas I/II Recruiting Combined with gemcitabine and nab-paclitaxel NCT02077881 
 Prostate carcinoma II Recruiting Combined with sipuleucel-T NCT01560923 
 NSCLC II Not yet recruiting Combined with docetaxel and tergenpumatucel-L NCT02460367 
INCB024360 Advanced neoplasms Completed As single agent NCT01195311 
 Myelodysplastic syndromes II Active, not recruiting As single agent NCT01822691 
 Melanoma I/II Recruiting Combined with ipilimumab NCT01604889 
  II Recruiting Combined with a multipeptide-based vaccine NCT01961115 
 Reproductive tract tumors II Completed Compared to tamoxifen NCT01685255 
  Recruiting As single agent NCT02042430 
  I/II Withdrawn Combined with vaccine therapy NCT01982487 
  Recruiting Combined with adoptive transfer of NK cells and IL2 NCT02118285 
  I/II Recruiting Combined with DC-targeted NY-ESO-1 and poly-ICLC NCT02166905 
 Solid tumors I/II Recruiting Combined with a PDCD1 mAb NCT02178722 
  I/II Recruiting Combined with MEDI4736 (PD-L1 mAb) NCT02318277 
 Previously treated NSCLC Recruiting Combined with MPDL3280A (PD-L1 mAb) NCT02298153 
GDC-0919 (formerly NLG-919) Solid tumors Recruiting As single agent NCT02048709 
 Locally advanced or metastatic solid tumors Not yet open Combined with MPDL3280A (PD-L1 mAb) NCT02471846 
IDO1 peptide NSCLC Completed As single agent NCT01219348 
 Melanoma Recruiting Combined with ipilimumab or vemurafenib NCT02077114 
 Melanoma II Recruiting Combined with temozolomide, imiquimod, GM-CSF and survivin peptide NCT01543464 
AgentIndication(s)PhaseStatusNotesIdentifier
Indoximod (D-1-MT) Metastatic solid tumor Completed Combined with docetaxel NCT01191216 
 Solid tumor Completed As single agent NCT00567931 
  Terminated As single agent NCT00739609 
 Malignant glioma I/II Recruiting For recurrent glioma patients NCT02052648 
 Metastatic breast cancer I/II Active, not recruiting Combined with vaccine NCT01042535 
  II Recruiting Combined with docetaxel NCT01792050 
 Melanoma I/II Recruiting Combined with ipilimumab NCT02073123 
 Metastatic adenoma of pancreas I/II Recruiting Combined with gemcitabine and nab-paclitaxel NCT02077881 
 Prostate carcinoma II Recruiting Combined with sipuleucel-T NCT01560923 
 NSCLC II Not yet recruiting Combined with docetaxel and tergenpumatucel-L NCT02460367 
INCB024360 Advanced neoplasms Completed As single agent NCT01195311 
 Myelodysplastic syndromes II Active, not recruiting As single agent NCT01822691 
 Melanoma I/II Recruiting Combined with ipilimumab NCT01604889 
  II Recruiting Combined with a multipeptide-based vaccine NCT01961115 
 Reproductive tract tumors II Completed Compared to tamoxifen NCT01685255 
  Recruiting As single agent NCT02042430 
  I/II Withdrawn Combined with vaccine therapy NCT01982487 
  Recruiting Combined with adoptive transfer of NK cells and IL2 NCT02118285 
  I/II Recruiting Combined with DC-targeted NY-ESO-1 and poly-ICLC NCT02166905 
 Solid tumors I/II Recruiting Combined with a PDCD1 mAb NCT02178722 
  I/II Recruiting Combined with MEDI4736 (PD-L1 mAb) NCT02318277 
 Previously treated NSCLC Recruiting Combined with MPDL3280A (PD-L1 mAb) NCT02298153 
GDC-0919 (formerly NLG-919) Solid tumors Recruiting As single agent NCT02048709 
 Locally advanced or metastatic solid tumors Not yet open Combined with MPDL3280A (PD-L1 mAb) NCT02471846 
IDO1 peptide NSCLC Completed As single agent NCT01219348 
 Melanoma Recruiting Combined with ipilimumab or vemurafenib NCT02077114 
 Melanoma II Recruiting Combined with temozolomide, imiquimod, GM-CSF and survivin peptide NCT01543464 

NOTE: Clinical trials were identified on the website clinicaltrials.gov as of 15 July, 2015.

The seminal observation associating IDO1, immunosuppression, and cancer utilized a polyclonal antibody to identify the immunohistochemical frequency of expression among different human malignancies (60). Unexpectedly, recent analyses utilizing a novel monoclonal anti-human IDO1 antibody have demonstrated distinct differences compared with those original observations (20). While it was initially reported that 90% to 100% of human prostate and pancreatic tumors, as well as glioblastoma, were IDO1 positive, the latter study found only 42%, 38%, and 8% of those malignancies positive, respectively. As the antibodies were well vetted in both investigations, these conclusions present a cautionary tale that likely reflects more than simple differences in antibody specificity, but more broadly, the potential for alternative splice variants and/or posttranslational modifications resulting in antigenic variation. Thus, immunohistochemical studies associating IDO1 expression and survival should be interpreted carefully (61). Furthermore, these conflicting findings complicate strategies that would ideally use IDO1 IHC as a prognostic tool for selecting patients who would benefit most from IDO1 inhibition.

Recent work studying the Kyn/Trp ratio in patients with glioblastoma has suggested that analyzing a time point well after surgical tumor resection of 10+ weeks following the procedure, may be prognostically valuable to clinicians planning to enroll patients in immunotherapy trials (62). While this finding requires further validation in a larger patient cohort, it suggests the possibility that IDO1 activity increases well after glioblastoma patients are operated on, as well as highlighting the potential relevance of using a clinical inhibitor against IDO1 systemically. Similarly, the Kyn/Trp ratio was recently validated as a prognostic tool in cervical cancer patients whereby low Trp levels indicated a tumor size greater than 4 cm and metastatic spread to the lymph node (63). Accordingly, high Kyn/Trp ratios in patient sera were associated with lymph node metastasis, FIGO stage, tumor size, parametrial invasion, and poor disease-specific survival, further suggesting the relevance of IDO1 targeting based on a tryptophan catabolic signature. Similar work was recently shown in a clinical study that identified higher Kyn/Trp ratios in T-cell lymphotropic virus type-1 asymptomatic carriers when compared with healthy controls (64). Importantly, the serum Kyn/Trp ratio was a significantly independent detrimental prognostic factor in patients with adult T-cell leukemia/lymphoma. These collective analyses have begun to elucidate the relevance of determining an IDO1 enzymatic “signature” in patient sera, which preliminarily appears to be both prognostically valuable and clinically informative.

Given that the majority of clinical studies aimed at IDO1 inhibition that are currently ongoing have yet to report results, we can gain insight into preclinical analyses that have shown great potential in targeting this immunosuppressive mediator. However, these models possess limited usefulness when considering the potential effects that standard-of-care treatment have on IDO1 activity and/or expression, as well as the potential change of expression between primary and recurrent tumors. Given that inflammation is a primary driver of IDO1 expression, it may be relevant to prognostically stratify tumors that possess a wide range of T-cell–infiltrating heterogeneity, when compared with the primary, versus relapsed malignancy (65, 66).

Our substantial knowledge of the role and expression of IDO1 in cancer has continued to expand over the past two decades, yet critical questions regarding alternative functions regulated by posttranslational modifications, the role that IDO2/TDO plays in the absence or inhibition of IDO1, as well as the impact of tissue-specific alternative splicing, still remain. Most inhibitory strategies against IDO1 focus on disabling enzymatic activity. However, preclinical mouse tumor models suggest that this tactic alone will not lead to effective antitumor immunity, further suggesting that IDO1 inhibition is best suited for combinatorial therapeutic strategies. However, these findings also raise the intriguing, yet unproven possibility that IDO1 subsumes a new/alternative immunosuppressive role when Trp catabolism is abrogated in vivo. In support of this hypothesis, it is notable that indoximod (D-1-MT), currently cast as an IDO1 pathway inhibitor, does not inhibit Trp to Kyn catabolism (refs. 67, 68; Supplementary Table S1). This combination of reported observations and untested hypotheses paints a blurry picture of a highly immunosuppressive player in tumor immunity. Unmistakably, IDO1 is a critical mediator that, given the normal limited expression throughout the body, makes it an ideal target for cancer immunotherapy. The central question going forward, thus becomes, how can we best inhibit the activity of this pleiotropic target?

No potential conflicts of interest were disclosed.

Conception and design: L. Zhai, G. Gritsina, F.J. Giles, D.A. Wainwright

Development of methodology: L. Zhai, D.A. Wainwright

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L. Zhai, D.A. Wainwright

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L. Zhai, F.J. Giles, D.A. Wainwright

Writing, review, and/or revision of the manuscript: L. Zhai, S. Spranger, D.C. Binder, G. Gritsina, K.L. Lauing, F.J. Giles, D.A. Wainwright

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

Study supervision: D.A. Wainwright

The authors thank Michael Gallagher for his expertise in medical illustration and contribution toward creation of Fig. 1.

S. Spranger is supported by a Cancer Research Institute Postdoctoral Fellowship. D.A. Wainwright is supported by PHS grant number R00NS082381, awarded by the NINDS, U.S. Department of Health and Human Services; a Robert H. Lurie Comprehensive Cancer Center—Zell Scholar Program of the Zell Family Foundation Gift; and the Northwestern Brain Tumor Institute.

1.
Fontenot
JD
,
Gavin
MA
,
Rudensky
AY
. 
Foxp3 programs the development and function of CD4+CD25+ regulatory T cells
.
Nat Immunol
2003
;
4
:
330
6
.
2.
Annunziato
F
,
Cosmi
L
,
Liotta
F
,
Lazzeri
E
,
Manetti
R
,
Vanini
V
, et al
Phenotype, localization, and mechanism of suppression of CD4(+)CD25(+) human thymocytes
.
J Exp Med
2002
;
196
:
379
87
.
3.
Bluestone
JA
,
Abbas
AK
. 
Natural versus adaptive regulatory T cells
.
Nat Rev Immunol
2003
;
3
:
253
7
.
4.
Curiel
TJ
,
Coukos
G
,
Zou
L
,
Alvarez
X
,
Cheng
P
,
Mottram
P
, et al
Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival
.
Nat Med
2004
;
10
:
942
9
.
5.
El Andaloussi
A
,
Lesniak
MS
. 
An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme
.
Neuro Oncol
2006
;
8
:
234
43
.
6.
Konishi
J
,
Yamazaki
K
,
Azuma
M
,
Kinoshita
I
,
Dosaka-Akita
H
,
Nishimura
M
. 
B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression
.
Clin Cancer Res
2004
;
10
:
5094
100
.
7.
Dong
H
,
Strome
SE
,
Salomao
DR
,
Tamura
H
,
Hirano
F
,
Flies
DB
, et al
Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion
.
Nat Med
2002
;
8
:
793
800
.
8.
Bloch
O
,
Crane
CA
,
Kaur
R
,
Safaee
M
,
Rutkowski
MJ
,
Parsa
AT
. 
Gliomas promote immunosuppression through induction of B7-H1 expression in tumor-associated macrophages
.
Clin Cancer Res
2013
;
19
:
3165
75
.
9.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
10.
Brahmer
JR
,
Tykodi
SS
,
Chow
LQ
,
Hwu
WJ
,
Topalian
SL
,
Hwu
P
, et al
Safety and activity of anti-PD-L1 antibody in patients with advanced cancer
.
N Engl J Med
2012
;
366
:
2455
65
.
11.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
12.
Wolchok
JD
,
Kluger
H
,
Callahan
MK
,
Postow
MA
,
Rizvi
NA
,
Lesokhin
AM
, et al
Nivolumab plus ipilimumab in advanced melanoma
.
N Engl J Med
2013
;
369
:
122
33
.
13.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
14.
Duraiswamy
J
,
Kaluza
KM
,
Freeman
GJ
,
Coukos
G
. 
Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors
.
Cancer Res
2013
;
73
:
3591
603
.
15.
Wainwright
DA
,
Chang
AL
,
Dey
M
,
Balyasnikova
IV
,
Kim
C
,
Tobias
AL
, et al
Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4 and PD-L1 in mice with brain tumors
.
Clin Cancer Res
2014
;
20
:
5290
301
.
16.
Weber
J
. 
Immune checkpoint proteins: a new therapeutic paradigm for cancer–preclinical background: CTLA-4 and PD-1 blockade
.
Semin Oncol
2010
;
37
:
430
9
.
17.
Grohmann
U
,
Fallarino
F
,
Puccetti
P
. 
Tolerance, DCs and tryptophan: much ado about IDO
.
Trends Immunol
2003
;
24
:
242
8
.
18.
Zhang
Y
,
Kang
SA
,
Mukherjee
T
,
Bale
S
,
Crane
BR
,
Begley
TP
, et al
Crystal structure and mechanism of tryptophan 2,3-dioxygenase, a heme enzyme involved in tryptophan catabolism and in quinolinate biosynthesis
.
Biochemistry
2007
;
46
:
145
55
.
19.
Sugimoto
H
,
Oda
S
,
Otsuki
T
,
Hino
T
,
Yoshida
T
,
Shiro
Y
. 
Crystal structure of human indoleamine 2,3-dioxygenase: catalytic mechanism of O2 incorporation by a heme-containing dioxygenase
.
Proc Natl Acad Sci U S A
2006
;
103
:
2611
6
.
20.
Theate
I
,
van Baren
N
,
Pilotte
L
,
Moulin
P
,
Larrieu
P
,
Renauld
JC
, et al
Extensive profiling of the expression of the indoleamine 2,3-dioxygenase 1 protein in normal and tumoral human tissues
.
Cancer Immunol Res
2015
;
3
:
161
72
.
21.
Pilotte
L
,
Larrieu
P
,
Stroobant
V
,
Colau
D
,
Dolusic
E
,
Frederick
R
, et al
Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase
.
Proc Natl Acad Sci U S A
2012
;
109
:
2497
502
.
22.
Comings
DE
,
Muhleman
D
,
Dietz
G
,
Sherman
M
,
Forest
GL
. 
Sequence of human tryptophan 2,3-dioxygenase (TDO2): presence of a glucocorticoid response-like element composed of a GTT repeat and an intronic CCCCT repeat
.
Genomics
1995
;
29
:
390
6
.
23.
Dai
W
,
Gupta
SL
. 
Regulation of indoleamine 2,3-dioxygenase gene expression in human fibroblasts by interferon-gamma. Upstream control region discriminates between interferon-gamma and interferon-alpha
.
J Biol Chem
1990
;
265
:
19871
7
.
24.
Shimizu
T
,
Nomiyama
S
,
Hirata
F
,
Hayaishi
O
. 
Indoleamine 2,3-dioxygenase. Purification and some properties
.
J Biol Chem
1978
;
253
:
4700
6
.
25.
Haber
R
,
Bessette
D
,
Hulihan-Giblin
B
,
Durcan
MJ
,
Goldman
D
. 
Identification of tryptophan 2,3-dioxygenase RNA in rodent brain
.
J Neurochem
1993
;
60
:
1159
62
.
26.
Knox
WE
,
Auerbach
VH
. 
The hormonal control of tryptophan peroxidase in the rat
.
J Biol Chem
1955
;
214
:
307
13
.
27.
Ball
HJ
,
Sanchez-Perez
A
,
Weiser
S
,
Austin
CJD
,
Astelbauer
F
,
Miu
J
, et al
Characterization of an indoleamine 2,3-dioxygenase-like protein found in humans and mice
.
Gene
2007
;
396
:
203
13
.
28.
Metz
R
,
DuHadaway
JB
,
Kamasani
U
,
Laury-Kleintop
L
,
Muller
AJ
,
Prendergast
GC
. 
Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target of the antitumor indoleamine 2,3-dioxygenase inhibitory compound d-1-Methyl-Tryptophan
.
Cancer Res
2007
;
67
:
7082
7
.
29.
Yuasa
H
,
Takubo
M
,
Takahashi
A
,
Hasegawa
T
,
Noma
H
,
Suzuki
T
. 
Evolution of vertebrate indoleamine 2,3-dioxygenases
.
J Mol Evol
2007
;
65
:
705
14
.
30.
Pantouris
G
,
Serys
M
,
Yuasa
HJ
,
Ball
HJ
,
Mowat
CG
. 
Human indoleamine 2,3-dioxygenase-2 has substrate specificity and inhibition characteristics distinct from those of indoleamine 2,3-dioxygenase-1
.
Amino Acids
2014
;
46
:
2155
63
.
31.
Metz
R
,
Smith
C
,
DuHadaway
JB
,
Chandler
P
,
Baban
B
,
Merlo
LMF
, et al
IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation
.
Int Immunol
2014
;
26
:
357
67
.
32.
Nagano
J
,
Shimizu
M
,
Hara
T
,
Shirakami
Y
,
Kochi
T
,
Nakamura
N
, et al
Effects of indoleamine 2,3-dioxygenase deficiency on high-fat diet-induced hepatic inflammation
.
PLoS One
2013
;
8
:
e73404
.
33.
Taylor
MW
,
Feng
GS
. 
Relationship between interferon-gamma, indoleamine 2,3-dioxygenase, and tryptophan catabolism
.
FASEB J
1991
;
5
:
2516
22
.
34.
Munn
DH
,
Zhou
M
,
Attwood
JT
,
Bondarev
I
,
Conway
SJ
,
Marshall
B
, et al
Prevention of allogeneic fetal rejection by tryptophan catabolism
.
Science
1998
;
281
:
1191
3
.
35.
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
.
36.
Hwu
P
,
Du
MX
,
Lapointe
R
,
Do
M
,
Taylor
MW
,
Young
HA
. 
Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation
.
J Immunol
2000
;
164
:
3596
9
.
37.
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
.
38.
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
.
39.
Munn
DH
,
Sharma
MD
,
Baban
B
,
Harding
HP
,
Zhang
Y
,
Ron
D
, et al
GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase
.
Immunity
2005
;
22
:
633
42
.
40.
Metz
R
,
Rust
S
,
Duhadaway
JB
,
Mautino
MR
,
Munn
DH
,
Vahanian
NN
, et al
IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: a novel IDO effector pathway targeted by D-1-methyl-tryptophan
.
Oncoimmunology
2012
;
1
:
1460
8
.
41.
Segawa
H
,
Fukasawa
Y
,
Miyamoto
K-i
,
Takeda
E
,
Endou
H
,
Kanai
Y
. 
Identification and functional characterization of a Na+-independent neutral amino acid transporter with broad substrate selectivity
.
J Biol Chem
1999
;
274
:
19745
51
.
42.
Mellor
AL
,
Baban
B
,
Chandler
PR
,
Manlapat
A
,
Kahler
DJ
,
Munn
DH
. 
Cutting edge: CpG oligonucleotides induce splenic CD19+ dendritic cells to acquire potent indoleamine 2,3-dioxygenase-dependent T cell regulatory functions via IFN Type 1 signaling
.
J Immunol
2005
;
175
:
5601
5
.
43.
Fallarino
F
,
Volpi
C
,
Zelante
T
,
Vacca
C
,
Calvitti
M
,
Fioretti
MC
, et al
IDO mediates TLR9-driven protection from experimental autoimmune diabetes
.
J Immunol
2009
;
183
:
6303
12
.
44.
Fujigaki
H
,
Saito
K
,
Fujigaki
S
,
Takemura
M
,
Sudo
K
,
Ishiguro
H
, et al
The Signal transducer and activator of transcription 1α and interferon regulatory factor 1 are not essential for the induction of indoleamine 2,3-dioxygenase by lipopolysaccharide: involvement of p38 mitogen-activated protein kinase and nuclear factor-κB pathways, and synergistic effect of several proinflammatory cytokines
.
J Biochem
2006
;
139
:
655
62
.
45.
Balachandran
VP
,
Cavnar
MJ
,
Zeng
S
,
Bamboat
ZM
,
Ocuin
LM
,
Obaid
H
, et al
Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido
.
Nat Med
2011
;
17
:
1094
100
.
46.
Holtzhausen
A
,
Zhao
F
,
Evans
K
,
Tsutsui
M
,
Orabona
C
,
Tyler
DS
, et al
Melanoma-derived Wnt5a promotes local dendritic-cell expression of IDO and immunotolerance: opportunities for pharmacologic enhancement of immunotherapy
.
Cancer Immunol Res
2015
;
3
:
1082
95
.
47.
Litzenburger
UM
,
Opitz
CA
,
Sahm
F
,
Rauschenbach
KJ
,
Trump
S
,
Winter
M
, et al
Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, STAT3 and the AHR
.
Oncotarget
2014
;
5
:
1038
51
.
48.
Li
DD
,
Gao
YJ
,
Tian
XC
,
Yang
ZQ
,
Cao
H
,
Zhang
QL
, et al
Differential expression and regulation of Tdo2 during mouse decidualization
.
J Endocrinol
2014
;
220
:
73
83
.
49.
van Baren
N
,
Van den Eynde
BJ
. 
Tryptophan-degrading enzymes in tumoral immune resistance. Frontiers in Immunology
.
Front Immunol
2015
;
6
:
34
.
50.
Muller
AJ
,
Sharma
MD
,
Chandler
PR
,
Duhadaway
JB
,
Everhart
ME
,
Johnson
BA
 III
, et al
Chronic inflammation that facilitates tumor progression creates local immune suppression by inducing indoleamine 2,3 dioxygenase
.
Proc Natl Acad Sci U S A
2008
;
105
:
17073
8
.
51.
Muller
AJ
,
DuHadaway
JB
,
Chang
MY
,
Ramalingam
A
,
Sutanto-Ward
E
,
Boulden
J
, et al
Non-hematopoietic expression of IDO is integrally required for inflammatory tumor promotion
.
Cancer Immunol Immunother
2010
;
59
:
1655
63
.
52.
Gajewski
TF
,
Schreiber
H
,
Fu
YX
. 
Innate and adaptive immune cells in the tumor microenvironment
.
Nat Immunol
2013
;
14
:
1014
22
.
53.
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
.
54.
Holmgaard
RB
,
Zamarin
D
,
Munn
DH
,
Wolchok
JD
,
Allison
JP
. 
Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4
.
J Exp Med
2013
;
210
:
1389
402
.
55.
Spranger
S
,
Bao
R
,
Gajewski
TF
. 
Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity
.
Nature
2015
;
523
:
231
5
56.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
57.
Spranger
S
,
Koblish
HK
,
Horton
B
,
Scherle
PA
,
Newton
R
,
Gajewski
TF
. 
Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8(+) T cells directly within the tumor microenvironment
.
J Immunother Cancer
2014
;
2
:
3
.
58.
Iversen
TZ
,
Engell-Noerregaard
L
,
Ellebaek
E
,
Andersen
R
,
Larsen
SK
,
Bjoern
J
, et al
Long-lasting disease stabilization in the absence of toxicity in metastatic lung cancer patients vaccinated with an epitope derived from indoleamine 2,3 dioxygenase
.
Clin Cancer Res
2014
;
20
:
221
32
.
59.
Soliman
HH
,
Jackson
E
,
Neuger
T
,
Dees
EC
,
Harvey
RD
,
Han
H
, et al
A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors
.
Oncotarget
2014
;
5
:
8136
46
.
60.
Uyttenhove
C
,
Pilotte
L
,
Theate
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
.
61.
Mitsuka
K
,
Kawataki
T
,
Satoh
E
,
Asahara
T
,
Horikoshi
T
,
Kinouchi
H
. 
Expression of indoleamine 2,3-dioxygenase and correlation with pathological malignancy in gliomas
.
Neurosurgery
2013
;
72
:
1031
8
.
62.
Zhai
L
,
Dey
M
,
Lauing
KL
,
Gritsina
G
,
Kaur
R
,
Lukas
RV
, et al
The kynurenine to tryptophan ratio as a prognostic tool for glioblastoma patients enrolling in immunotherapy
.
J Clin Neurosci.
2015
;
22
:
1964
8
.
63.
Ferns
DM
,
Kema
IP
,
Buist
MR
,
Nijman
HW
,
Kenter
GG
,
Jordanova
ES
. 
Indoleamine-2,3-dioxygenase (IDO) metabolic activity is detrimental for cervical cancer patient survival
.
Oncoimmunology
2015
;
4
:
e981457
.
64.
Masaki
A
,
Ishida
T
,
Maeda
Y
,
Suzuki
S
,
Ito
A
,
Takino
H
, et al
Prognostic significance of tryptophan catabolism in adult T-cell leukemia/lymphoma
.
Clin Cancer Res
2015
;
21
:
2830
9
.
65.
Prins
RM
,
Soto
H
,
Konkankit
V
,
Odesa
SK
,
Eskin
A
,
Yong
WH
, et al
Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy
.
Clin Cancer Res
2011
;
17
:
1603
15
.
66.
Gill
BJ
,
Pisapia
DJ
,
Malone
HR
,
Goldstein
H
,
Lei
L
,
Sonabend
A
, et al
MRI-localized biopsies reveal subtype-specific differences in molecular and cellular composition at the margins of glioblastoma
.
Proc Natl Acad Sci U S A
2014
;
111
:
12550
5
.
67.
Löb
S
,
Königsrainer
A
,
Zieker
D
,
Brücher
BDM
,
Rammensee
H-G
,
Opelz
G
, et al
IDO1 and IDO2 are expressed in human tumors: levo- but not dextro-1-methyl tryptophan inhibits tryptophan catabolism
.
Cancer Immunol Immunother
2009
;
58
:
153
7
.
68.
Lob
S
,
Konigsrainer
A
,
Schafer
R
,
Rammensee
HG
,
Opelz
G
,
Terness
P
. 
Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells
.
Blood
2008
;
111
:
2152
4
.
69.
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
.
70.
Pallotta
MT
,
Orabona
C
,
Volpi
C
,
Vacca
C
,
Belladonna
ML
,
Bianchi
R
, et al
Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells
.
Nat Immunol
2011
;
12
:
870
8
.
71.
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
.
72.
Beischlag
TV
,
Luis Morales
J
,
Hollingshead
BD
,
Perdew
GH
. 
The aryl hydrocarbon receptor complex and the control of gene expression
.
Crit Rev Eukaryot Gene Expr
2008
;
18
:
207
50
.
73.
Seymour
RL
,
Ganapathy
V
,
Mellor
AL
,
Munn
DH
. 
A high-affinity, tryptophan-selective amino acid transport system in human macrophages
.
J Leukoc Biol
2006
;
80
:
1320
7
.
74.
Silk
JD
,
Lakhal
S
,
Laynes
R
,
Vallius
L
,
Karydis
I
,
Marcea
C
, et al
IDO induces expression of a novel tryptophan transporter in mouse and human tumor cells
.
J Immunol
2011
;
187
:
1617
25
.
75.
Mezrich
JD
,
Fechner
JH
,
Zhang
X
,
Johnson
BP
,
Burlingham
WJ
,
Bradfield
CA
. 
An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T Cells
.
J Immunol
2010
;
185
:
3190
8
.
76.
Sharma
MD
,
Baban
B
,
Chandler
P
,
Hou
D-Y
,
Singh
N
,
Yagita
H
, 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
.