7,848
Views
83
CrossRef citations to date
0
Altmetric
Review

Trial watch: IDO inhibitors in cancer therapy

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Article: 1777625 | Received 13 May 2020, Accepted 31 May 2020, Published online: 14 Jun 2020

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first, rate-limiting step of the so-called “kynurenine pathway”, which converts the essential amino acid L-tryptophan (Trp) into the immunosuppressive metabolite L-kynurenine (Kyn). While expressed constitutively by some tissues, IDO1 can also be induced in specific subsets of antigen-presenting cells that ultimately favor the establishment of immune tolerance to tumor antigens. At least in part, the immunomodulatory functions of IDO1 can be explained by depletion of Trp and accumulation of Kyn and its derivatives. In animal tumor models, genetic or pharmacological IDO1 inhibition can cause the (re)activation of anticancer immune responses. Similarly, neoplasms expressing high levels of IDO1 may elude anticancer immunosurveillance. Therefore, IDO1 inhibitors represent promising therapeutic candidates for cancer therapy, and some of them have already entered clinical evaluation. Here, we summarize preclinical and clinical studies testing IDO1-targeting interventions for oncologic indications.

Introduction

L-tryptophan (Trp), one of the essential amino acids, is indispensable for protein synthesis and cell survival. The kynurenine pathway catabolizes Trp to active metabolites such as L-kynurenine (Kyn), kynurenic acid, 3-hydroxykynurenine, 3-hydroxyanthranilic acid, picolinic acid and quinolinic acid. This metabolic cascade can be catalyzed by three enzymes, namely, indoleamine 2,3- dioxygenase 1 (IDO1), IDO2, and tryptophan 2,3-dioxygenase (TDO2).Citation1Citation4 IDO1 is by far the best-studied among these enzymes, as it was the first interferon (IFN)-activated gene to be described as early as in the late 1970s.Citation5 The differential distribution and activity of IDO2 and TDO2 calls for further investigation to elucidate to which extent IDO2 and TDO2 contribute to Trp catabolism in vivo.Citation1,Citation5Citation7

Table 1. Clinical trials testing IDO-1 inhibitors in oncological indications.

In 1998 Munn, Mellor and colleagues demonstrated for the first time that IDO1 exerts immunosuppressive functions, as it prevents rejection of allogenic fetuses by the maternal immune system.Citation6,Citation8 This conceptual breakthrough initiated an intense wave of research aimed at understanding the molecular and cellular circuitries implicated in the immunomodulatory functions of IDO1.

Subsequent studies revealed that IDO1 is a central driver of cancer development and progression. In particular, IDO1 mediates pathogenic inflammatory processes in malignant, stromal and immune cells that ultimately lead to immune tolerance to tumor antigens.Citation9,Citation10 According to current knowledge, the pleiotropic role of IDO in cancer includes the suppression of cytotoxic T lymphocytes (CTL)Citation10Citation13 and natural killer (NK) cells,Citation14,Citation15 the generation and activation of regulatory T (TREG) cellsCitation16,Citation17 and myeloid-derived suppressor cells (MDSCs)Citation17Citation20 as well as the promotion of tumor angiogenesis.Citation10,Citation17 The immunomodulatory functions of IDO1 can be attributed to Trp starvation and increased Kyn levels.Citation21,Citation22 More specifically, Trp depletion induces cell cycle arrest of T cells and apoptosis through inhibition of the mechanistic target of rapamycin complex 1 (mTORC1),Citation10,Citation23,Citation24 while inducing a stress response that activates the general control nondepressible 2 (GCN2).Citation25Citation27 Increased levels of Trp metabolites, especially Kyn, activate the transcription factor aryl hydrocarbon receptor (AHR), which in return induces differentiation of CD4+ T cells into immunosuppressive TREG cells.Citation28Citation30 Alongside, IDO1-epxressing dendritic cells (DCs) have been shown to mediate immunosuppressive functions independent of Trp depletion and Kyn accumulation.Citation31Citation35 Moreover, IDO1 has been recently implicated in the microbiota-dependent control of obesity by shifting Trp metabolism from indole derivatives and interleukin 22 (IL-22) synthesis toward kynurenine production.Citation28,Citation36

IDO1 is widely overexpressed in tumor cells, which has been predominantly associated with poor prognosis.Citation8,Citation37Citation39 Similarly, increased circulating levels of Trp metabolites, such as Kyn, have been detected in patients with various cancers and have been attributed a poor predictive value in some cohorts.Citation40Citation42 Also, IDO1 expression in tumor cells has been linked to the status of the oncosuppressor gene bridging integrator 1 (BIN1).Citation43,Citation44 BIN1 is one of the most frequently down-regulated genes in human cancer,Citation45 due to either abnormal RNA splicing patterns compromising its tumor suppressor function,Citation46Citation48 or increased gene methylation abolishing its expression.Citation49,Citation50 In particular, BIN1 is absent or underexpressed in various human neoplasms, such as neuroblastoma,Citation51 melanoma,Citation46 as well as breast, lung, colorectal and prostate carcinoma.Citation46,Citation52,Citation53 The loss of BIN1 triggers the interferon gamma (IFNγ)-induced expression of IDO1, ultimately favoring tumor growth in immunocompetent, but not in immunodeficient, mice.Citation43 High levels of IDO not only correlate with poor outcome in some malignancies but they may also be implicated in drug resistance, as this has been reported for IDO1-expressing ovarian cancer patients.Citation54 Likewise, higher Kyn/Trp ratio have been shown to predict resistance to programmed cell death 1 (PDCD1, best known as PD-1) blockade in patients with non-small cell lung carcinoma (NSCLC).Citation55,Citation56 At last, profiling of advanced melanoma and renal cell carcinoma (RCC) patients showed that Kyn/Trp alterations correlated with overall survival upon administration of nivolumab (a PD-1 blocker).Citation57

Thus, IDO inhibition stands out as a promising strategy to (re)instance cancer immunosurveillance. Indeed, IDO inhibitors demonstrated their ability to successfully cooperate with immunotherapy, radiotherapy or chemotherapy even in tumors that are normally resistant to conventional treatments.Citation10,Citation58,Citation59 In this setting, preclinical studies have revealed an interesting paradox: while IDO inhibitors have a negligible effect on established tumors as single-agent, combination of IDO inhibitors and immunotherapies including checkpoint blockers targeting cytotoxic T lymphocyte-associated protein 4 (CTLA4) or PD-1 yields a synergistic effect to control cancer burden and favor survival.Citation17,Citation60Citation63 Here, we discuss recent progresses on the use of IDO1 agonists in preclinical and clinical settings as a strategy for the (re)activation of antitumor immune responses.

Preclinical advances

In this section we summarize the findings of key preclinical studies on the ability of IDO1 inhibitors to (re)instate anticancer immunosurveillance since the publication by Hornyák et al. dealing with this topic.Citation64

Indoximod

The simple racemic compound 1-methyl-tryptophan (1MT) was first described as a competitive inhibitor of the IDO1 enzyme in the early 1990s.Citation65 It is by far the most employed IDO inhibitor in the preclinical literature. Unlike its L isomer, which has shown weak inhibitory activity, D-1MT isomer neither binds nor inhibits the purified IDO1 enzyme while demonstrating anticancer activity.Citation66Citation68 Therefore, clinical development focused on D-1MT (best known as indoximod or NLG8189).Citation69 In contrast to direct enzymatic inhibitors of IDO1, indoximod acts downstream of IDO1 to stimulate mTORC1, possibly lowering risks of drug resistance.Citation69,Citation70 Several combinatorial regimens have been developed to increase the antineoplastic effects of indoximod, some of which demonstrated pronounced therapeutic activity in preclinical models of hepatocellular carcinoma (HCC),Citation71 advanced prostateCitation72 and lung cancer.Citation73 Indeed, IDO1 inhibition with 1MT, synergized with radiotherapy to downregulate TREG cells, reduce expression of PD-1 or its ligand CD274 (best known as PD-L1), and to prevent T cell exhaustion in Lewis lung cancer (LLC)-bearing mice.Citation73 D-1MT and CTLA4 blockers administration mediated improved therapeutic effects in treatment resistant IDO1-overexpressing HCCs in both subcutaneous and hepatic orthotopic models.Citation71 Additionally, CTLA4 blockade induced the IFNγ-dependent upregulation of IDO1 in chemoresistant (but not sensitive) HCCs in mice.Citation71 At last, IDO activity positively correlates with disease stage in prostate cancer patients,Citation72 and both a DNA vaccine encoding the tumor-associated antigen acid phosphatase 3 (ACP3, best known as PAP) and PD-1 blockade with pembrolizumab promotes IDO expression and activity in these individuals.Citation72 Consistent with the immunosuppressive activity of IDO in this setting, ex vivo stimulation of peripheral blood cells with 1-MT increased T cell responses to vaccination.Citation72 Recently, Hu et al. also demonstrated that a methyltryptophan-paclitaxel (MP) albumin-bound drug conjugate (that links indoximod to the microtubular poison paclitaxelCitation74-Citation77 through an ester bond) not only significantly elevates the tumor levels of indoximod and local CD8+ T populations, but reduces granulocyte-like myeloid derived suppressor cells (G-MDSCs) and TREG cells.Citation78

Epacadostat (INCB024360)

Epacadostat, also known as INCB024360, is an orally available reversible competitive IDO1 inhibitor. Wachowska and colleagues reported that photodynamic therapy (PDT)Citation79Citation82 induced IDO1 expression within neoplasms as well as in tumor draining lymph nodes in murine orthotopic breast cancer models.Citation83 Mechanistically, granulocytic CD11b+Ly6G+ myeloid cells were the major source of IDO1 and strongly infiltrated the tumor bed following PDT.Citation83 Although less abundant after PDT, monocytic CD11b+Ly6C+ myeloid cells, could also upregulate IDO1.Citation83 Interestingly, depending on the therapeutic scheme of PDT administration, IDO-induced immunosuppression can either be beneficial or lead to systemic toxicity.Citation83 Although IL-6 neutralization restored antitumor efficacy, it abolished the synergistic effect of epacadostat and PDT.Citation83 This might be explained by the fact that constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, signal transducer and activator of transcription 3 (STAT3)Citation84Citation87 and the AHR.Citation88

Navoximod (GDC-0919, NLG-919)

Navoximod (also known as GDC-0919 or NLG-919) was initially developed as an orally bioavailable IDO1/TDO inhibitor with an improved pharmacokinetic and toxicity profile, based on 4-phenylimidazole, a compound that binds the heme moiety within the catalytic site of IDO1.Citation89 IDO1 inhibition by navoximod has been shown to decrease plasmatic Kyn/Trp ratios and tumor Kyn levels.Citation90 In sarcoma-bearing mice, navoximod used alone or combined with a PD-L1 blocker could neither efficiently control tumor growth nor affect the tumor immune cell infiltrate.Citation90 However, in the 4T1 murine breast tumor model, navoximod synergizes with doxorubicinCitation91-Citation93 to elicit an antitumoral immune response and to control tumor growth.Citation94,Citation95

PF-06840003 and BGS-5777

PF-06840003 is a highly selective IDO1 inhibitor with favorable pharmacokinetic characteristics and a prolonged half-life in humans, which enable single-dose daily administration. Additionally, its ability to enter the central nervous system (CNS) allows for its use against brain metastases.Citation96 In several preclinical tumor models in mice, PF-06840003 strongly reduced intratumoral Kyn levels and inhibited tumor growth in both monotherapy and, with an increased efficacy, in combinatorial regimens with PD-L1 or CTLA4 blockers.Citation97 Recently, BGB-5777, a potent CNS-penetrating IDO1 inhibitor, enabled a durable survival benefit in a fraction of patients with advanced glioblastoma when combined with nivolumab and radiation therapy.Citation98,Citation99

BMS-986205

BMS-986205 is an orally available irreversible inhibitor of IDO1. Current clinical studies have shown its dose-dependent efficacy, coupled to better efficiency and pharmacokinetics than epacadostat.Citation10 Even at a low concentrations, BMS-986205 successfully inhibits IDO1 and lowers Kyn serum levels.Citation100

Other IDO1 inhibitors

A few additional IDO1 inhibitors are in preclinical development, including Trp analogs,Citation1 imidazoles,Citation101 phenyl benzenesulfonylhydrazides,Citation102 N-hydroxyamidinesCitation103 and LW106.Citation104 Other IDO1 inhibitors being developed by pharmaceutical groups in late preclinical settings, which include IOM2983 (Merck/IO-Met) and RG-70099 (Roche/CuraDev), have not yet publicly disclosed. In contrast, SHR9146 (also known as HTI-1090), an inhibitor of IDO1 and TDO, and KHK2455, an IDO1 inhibitor, have recently entered early clinical development.Citation1,Citation105 Overall, these compounds offer abundant possibilities for exploring the effects of specific IDO1 inhibition in the clinics.

Translational and clinical progress

A number of translational and clinical results addressing the safety and therapeutic potential of IDO1 inhibitors have been published since the latest survey on this topic (January 2018).Citation64 Here, we discuss some of these recent studies to recapitulate the current state-of-the-art.

Translational studies

Recent immunohistochemical analyses demonstrate that patchy expression of IDO1 within cervical cancers is associated with an increased systemic Kyn/Trp ratio and poor disease outcome, whereas marginal IDO1 expression pattern in the tumor predicts favorable outcome.Citation106 At least in part, these observations could be related to T cell infiltration and IFNγ release in the cervical tumor microenvironment.Citation106 Along similar lines, analyses of 144 cervical tumor samples from The Cancer Genome Atlas (TCGA) revealed a strong and positive correlation between IDO1 and IFNG mRNA expression levels, as well as significantly improved disease-free survival for patients with high IDO1 and IFNG levels.Citation106

Li and colleagues demonstrated that serum Kyn/Trp ratio increases as an adaptive resistance mechanism associated with worse overall survival in advanced melanoma and RCC patients treated with nivolumab.Citation57 They further established a correlation in melanoma samples between Kyn/Trp ratio and IDO1 but not TDO mRNA levels 4 weeks after nivolumab administration,Citation57,Citation107 suggesting that IDO1 may be the major source of Kyn in this setting. At last, two studies described synergistic effects of agents targeting erb-b2 receptor tyrosine kinase 2 (ERBB2, best known as HER2),Citation108,Citation109 IDO1 and PD-1.Citation110,Citation111 Upon antibody-dependent cellular phagocytosis (ADCP), macrophages inhibit NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) and T cell-mediated cytotoxicity in breast cancers and lymphomas.Citation2Citation11,Citation110,Citation112 Mechanistically, following ADCP, absent in melanoma 2 (AIM2) is recruited to the phagosomes by FcγR signaling and activated by DNA from phagocytosed tumor cells.Citation111,Citation113 Upon activation, AIM2 upregulates PD-L1 and IDO to cause immunosuppression. Combined treatment with anti-HER2 antibodies and inhibitors of PD-L1 and IDO enhances anti-tumor immunity and anti-HER2 therapeutic efficacy in vitroCitation111 as well as in mouse models of HER2+ breast carcinoma.Citation110 Additionally, neoadjuvant trastuzumabCitation114-Citation120 treatment significantly upregulates PD-L1 and IDO on tumor-associated macrophages (TAMs) from HER2+ breast cancer patients, correlating with poor trastuzumab responses.Citation110 Collectively, these findings suggest that IDO inhibitors may provide synergistic effects with other targeted immunotherapies.

Published clinical trials

Komrokji et al. reported preliminary results for the sole published clinical trial monitoring the efficacy of epacadostat administered as standalone intervention.Citation121 In particular, this Phase II study aimed at evaluating the pharmacodynamics and activity of epacadostat in heavily pre-treated transfusion-dependent patients with myelodysplastic syndrome (MDS) after hypomethylating agent (HMA) failure.Citation122Citation124 The IDO1 inhibitor was well tolerated, as no Grade 3 or 4 treatment-related adverse events (TRAEs) were recorded. Only one patient (among the 15 included in the trial) developed grade 2 adrenal insufficiency and hypothyroidism, while another showed low testosterone levels. Eighty percent of individuals exhibited s table disease and 20% progressive disease, largely in line with the poor prognosis of this patient population (overall survival of ~18 months in low-risk disease and 4–6 months in high-risk disease). All these findings suggest that future studies should consider to test epacadostat earlier in the disease course, before HMA failure (since expansion of MDSCs probably contribute to myelosuppression).Citation121

All other clinical studies recently published on IDO1 inhibitors tested these agents in combination with immune checkpoint blockers. In particular, Gibney et al. reported the results for the Phase I/II clinical trial NCT01604889 enrolling patients with unresectable or metastatic melanoma and receiving epacadostat together with ipilimumab.Citation125Citation127 Among the 50 participants, 20 discontinued treatment due to disease progression and 48 experienced TRAEs including hypothyroidism (10%), pruritus (28%), alanine aminotransferase elevation (28%), rash (50%), and aspartate aminotransferase elevation (24%). Dose-limiting toxicities occurred in 11 patients, and doses ≥100 mg BID were not tested due to hepatotoxicity. Among immunotherapy-naive patients (n = 39), objective response rate was 23% by response evaluation criteria in solid tumors (RECIST) and 26% by immune-related response criteria (iRECIST). No objective response was observed in the 11 patients previously treated with immunotherapy. According to the authors, these preliminary findings support continuing the evaluation of epacadostat plus ipilimumab in patients with unresectable or metastatic melanoma.Citation128 Unfortunately the study was prematurely terminated due to the sponsor’s decision, and only the Phase I portion of the trial was completed.

The NCT02298153 ECHO-11O Phase Ib trial evaluated the efficacy, tolerability and safety of the epacadostat administered together with the PD-L1 blocker atezolizumab,Citation129Citation131 to 29 patients with stage IIIB/IV NSCLC previously treated with platinum derivativesCitation132-Citation136 chemotherapy in conjunction with a folic acid analogue.Citation137Citation139 Seventy-nine percent of enrolled patients experienced TRAEs, 17% discontinued treatment due to such effects, one patient showed anticancer partial response, and the maximum tolerated dose (MTD) was not achieved. Thus, the clinical activity of epacadostat plus atezolizumab against NSCLC was deceptive, in line with the hitherto unclear significance of IDO1 expression in this setting.Citation140 Ultimately, the ECHO-110 study was prematurely terminated due to slow recruitment.

Additional results have recently lent further support to the controversial efficacy of epacadostat administered in combination with immune checkpoint blockers.Citation141,Citation142 In particular, Mitchell et al. reported the results of the Phase I KEYNOTE-037/ECHO-202 (NCT02178722) trial, enrolling 62 individuals with several solid tumors, including 22 melanomas, 12 NSCLCs and 11 RCCs. Eighty-four percent of the patients exhibited tolerable Grade 1/2 TRAEs (such as nausea, pruritus, rash, fatigue and arthralgia), 11% of the subjects discontinued the therapy, and the MTD was not attained.Citation141 An objective response was observed in 55% of melanoma patients and in some patients with urothelial carcinoma, RCC, head and neck squamous cell carcinoma (HNSCC), endometrial adenocarcinoma or NSCLC (in all cases, independently of PD-L1 expression levels). Altogether, these results suggest an encouraging and durable antitumor activity for this combinatorial regimen that has be to confirmed in additional Phase II studies.Citation141,Citation143,Citation144 Long and colleagues published the first results for the KEYNOTE-252/ECHO-301 (NCT02752074) assay, a phase III randomized, double-blind study evaluating the efficacy of epacadostat combined with pembrolizumab versus placebo plus pembrolizumab in 706 patients with untreated, unresectable or metastatic melanoma.Citation142 At odds with the findings from ECHO-202 and despite promising preliminary observations, no evidence of improved progression free survival could be documented (4.7 months in the epacadostat plus pembrolizumab arm versus 4.9 months in the pembrolizumab only arm). Overall survival was 74% in both groups, and objective response rates were similar in the two arms. Additionally, the most common TRAE, a lipase increase, occurred with a similar frequency in both groups (9% of patients receiving pembrolizumab monotherapy versus 10% in individuals receiving the combinatorial regimen).Citation142

These disappointing results suggested that this combinatorial therapy did not improve the clinical outcome of melanoma patients receiving pembrolizumab, confirming that the role of epacadostat (or IDO1 inhibitors in general) in advanced solid tumors with robust PD-1 signaling remains unclear. No less than twelve Phase III clinical assays testing this IDO1 selective inhibitor, alone or in combinatorial regimen in different cancer contexts, have been recently been withdrawn, downsized or suspended.Citation145 Indeed, it remains to be elucidated whether IDO1 constitutes a robust target for the development of anticancer agents. The results of ongoing clinical trials (see below) may clarify whether IDO1 inhibitors are an option to improve the therapeutic activity of PD-1 blockade in some cancer patient populations.Citation146

The results of two studies investigating the clinical profile of indoximod have recently been reported. Soliman and colleagues showed that indoximod plus an adenoviral DC vaccine targeting tumor protein p53 (TP53)Citation147Citation149 was well tolerated by metastatic breast cancer patients enrolled in a Phase I/II clinical trial. Patients who did not exhibit particular side effects (none of the toxicities required treatment discontinuation) achieved a median progression-free survival of ~13 weeks and a median overall survival of ~21 weeks, suggesting the absence of a statistically significant effect of indoximod.Citation150 Moreover, preliminary results from the Phase II NCT02077881 assay, enrolling 104 metastatic pancreatic cancer patients treated with indoximod plus gemcitabine and paclitaxel, have been disclosed by Bahary et al. Median overall survival was ~11 months, overall response rate was 46% (including one patient experiencing a complete response), and no significant toxicities were documented (anemia, nausea and fatigue being the most common).Citation151

Navoximod has been tested as a standalone intervention in patients with advanced or recurrent solid tumors in a Phase I study aiming to assess the antitumor activity, safety, pharmacokinetics and pharmacodynamics of the IDO inhibitor (NCT02048709). Preliminary results by Nayak-Kapoor and colleagues indicate that the MTD was not reached in the 22 enrolled patients, with a single dose-limiting Grade 4 toxicity (lower gastrointestinal hemorrhage). In ≥20% of patients, regardless of causality, TRAEs included vomiting (27%), nausea (36%), pruritus, cough, decreased appetite (41% of each) and fatigue (59%). Grade ≥ 3 TRAEs, reported in 64% of patients, could be attributed to navoximod in two patients (9%). Overall, navoximod was well tolerated at doses up to 800 mg BID and, among patients evaluated for efficacy, 8 (36%) had stable disease and 10 (46%) progressed.Citation152

Results from two clinical trials, testing navoximod in combination with the PD-L1 inhibitor atezolizumab, in patients with advanced cancer, have recently been published.Citation153,Citation154 Jung et al. reported preliminary results from the NCT02471846 trial, consisting of a 3 + 3 dose-escalation (n = 66) and a tumor-specific expansion (n = 92) phase. Navoximod was given orally every 12 hours for 21 consecutive days of each cycle except for cycle 1, where navoximod administration started on day −1 to measure pharmacokinetics. The maximum administered dose was 1000 mg BID, and the MTD was not reached. Navoximod demonstrated a linear pharmacokinetic profile as plasma Kyn levels decreased in a dose-dependent manner. The most common TRAEs were rash (22%), chromaturia (20%) and fatigue (22%). Some degree of antitumoral activity was observed at all dose levels in various tumor types including breast, cervical, HNSCC, melanoma, neural sheath, NSCLC, ovarian, pancreatic, prostate, RCC and urothelial bladder cancer. Of note, 6 (9%) dose-escalation patients partially responded, and 10 (11%) expansion patients achieved partial or complete responses. Together, these findings proved that this regimen was safe and well tolerated, although there was no clear evidence of benefit from adding navoximod to atezolizumab.Citation154

At last, results from a dose-escalation study assessing navoximod alone or in combination with atezolizumab, in Japanese individuals with advanced solid tumors, were reported by Ebata and colleagues.Citation153 Patients received either navoximod alone in stage 1 (n = 10) or in combination with atezolizumab in stage 2 (n = 10). No dose-limiting toxicities were observed. In stage 1, chromaturia (50%) and maculopapular rash (20%) occurred in ≥20% of patients and Grade ≥ 3 TRAEs were reported in two patients (20%; maculopapular rash and increased lipase). In stage 2, chromaturia (60%) and decreased appetite (40%) occurred in ≥30% of patients, while Grade ≥ 3 TRAEs were reported in three patients (30%; alanine and aspartate aminotransferase increased, hyponatremia, lympho- and neutro-paenia). Stable disease was observed in 5 patients (50%) in stage 1 and 8 patients (80%) in stage 2. Overall, these results suggested that navoximod, as monotherapy and in combination with atezolizumab, was well tolerated in patients with advanced solid tumors.

Similarly, Riccuiti, Luke and colleaguesCitation155-Citation158 reported results for the NCT02658890 study which aimed at testing BMS-986205 administered as monotherapy once daily for 2 weeks followed by nivolumab in advanced bladder cancer. TRAEs (all grades) were reported in 57% of patients with 12% of Grade 3–4 side effects. The most common side effects of any grade were fatigue (15%) and nausea (12%). Nineteen patients (4%) discontinued treatment due to TRAEs, and 3 patients died due to a TRAE (hepatic failure, myocarditis and Stevens-Johnson syndrome). The combination of BMS-986205 and nivolumab was well tolerated in heavily pretreated patients and enhanced tolerability was observed with the 100 mg dose. Preliminary evidence of efficacy was observed in advanced bladder cancer, supporting further evaluation of this combinatorial regimen.

Ongoing clinical trials

When this Trial Watch was being redacted (May 2020), official sources listed 22 clinical trials launched after January 2018 () to evaluate the safety and efficacy of IDO1 targeting intervention in cancer patients (source http://www.clinicaltrials.gov). Ten of these studies involve BMS-986205, 9 epacadostat, 1 indoximod, 1 KHK2455 and 1 SHR9146.

In particular, epacadostat is being tested together with a brachyury-targeted antitumor vaccine, a transforming growth factor beta (TGFβ) trap-anti-PD-L1 antibody (M7824), and an IL-15/IL-15RA superagonist (ALT-803) in patients affected by metastatic castration-resistant prostate cancer (NCT03493945).Citation159

BN-Brachyury is a novel recombinant vector-based therapeutic cancer vaccine that enhances an immune response against brachyury,Citation37 a transcription factor that plays a key role in epithelial-mesenchymal transition (EMT) and is overexpressed in prostate adenocarcinoma.Citation160Citation163 M7824, a bifunctional fusion protein composed by 2 extracellular domains of a TGFβ trap and a human IgG1 anti PD-L1 mAb,Citation164,Citation165 is able to reverse the EMT, to promote ADCC in vitro,Citation165 and promising evidence of immunostimulatory and clinical activity in solid tumors has been provided.Citation166,Citation167 ALT-803 is a fusion protein that stimulates both T and NK cells via agonism of the IL-2 and IL-15 receptors, thus supporting ADCC induction in synergy with M7824.Citation168,Citation169 This combinatorial regimen is a promising therapeutic option because of the activation of vaccine-derived tumor-specific T cells (by ALT-803) that is boosted by M7824 and epacadostat.

NCT03532295 is the only trial testing the safety and preliminary efficacy of epacadostat in subjects affected by brain tumors. The synergy among this IDO1 inhibitor, radiotherapy, the vascular endothelial growth factor A (VEGFA)-targeting antibody bevacizumabCitation170,Citation171 and the humanized, hinge-stabilized IgG4, targeting the interaction of PD-1 with PD-L1 and PD-L2, INCMGA00012 (also known as MGA012),Citation172 might activate a pronounced anti-cancer immune response thus leading to tumor regression and improved outcome. Along similar lines, the remaining clinical trials that involve epacadostat assess safety and preliminary efficacy of the IDO1 inhibitor combined with pembrolizumab (and other immunotherapeutic regimens).Citation173Citation175 In particular, NCT03823131 evaluates the efficacy of tavokinogene telseplasmid (tavo) electroporation (EP), pembrolizumab, and epacadostat against unresectable HNSCC (as compared to pembrolizumab monotherapy). Moreover, the tolerability, safety and preliminary efficacy of epacadostat and pembrolizumab were tested in patients affected by (i) advanced pancreatic cancer with chromosomal instability or homologous recombination repair deficiency (HRD) (NCT03432676), (ii) esophageal squamous cell carcinoma (ESCC), esophageal adenocarcinoma and gastroesophageal adenocarcinoma (NCT03592407), (iii) HNSCC recurring after PD-1/PD-L1 therapy (NCT03463161), and (iv) ovarian clear cell carcinoma (NCT03602586). However, two of these studies are currently listed as “Withdrawn” (NCT03432676, because the trial is no longer financed by the main supporter, and NCT03592407, due to safety concerns), while two other studies have been “Suspended” (NCT03602586, for scheduled interim monitoring) or “Terminated” (NCT03463161, due to a conflict of interest among the investigators). In the Phase II NCT03592407 study, the administration of neoadjuvant epacadostat plus pembrolizumab (followed by standard chemoradiation) aimed at verifying the capacity of this combinatorial regimen to ameliorate the lymphoid compartment of the tumor, thus increasing the abundance of CD8+ CTLs expressing the effector molecule granzyme B (GZMB),Citation176 and reducing the relative amount of tumor-infiltrating CD4+CD25+FOXP3+ TREG cellsCitation177-Citation182 (with respect to CD8+ cells). Of note, the therapeutic profile of this neoadjuvant combinatorial regimen is currently assessed in a Phase II study (NCT03832673) enrolling patients with muscle-invasive bladder cancer (MIBC). Indeed, the published literature lends robust support to the notion that pembrolizumab not only is an encouraging neoadjuvant therapy for the treatment of PD-L1+ MIBC and neoplasm with high mutational burden,Citation183Citation185 but also increases overall survival (by ~3 months) in advanced urothelial carcinomaCitation185,Citation186 and exhibits good tolerability when administered to patients with advanced solid tumors in combination with epacadostat.Citation141 Moreover, Chu et al. have recently demonstrated that the manipulation of the immune microenvironment with IDO1 inhibition enhances patient responses to existing therapies.Citation187

Finally, the Phase I trial NCT03516708, evaluating the efficacy of epacadostat administered to locally advanced rectal cancer patients, in the context of the so-called XELOX regimen (capecitabine plus oxaliplatin)Citation188Citation191 for preoperative chemoradiotherapy, has been suspended to ensure patient safety during the Covid19 epidemics.Citation192Citation194

BMS-986205 is mainly being administered to cancer patients simultaneously receiving nivolumabCitation195 (NCT03792750, NCT03459222, NCT03519256, NCT03661320, NCT04106414, NCT04047706, NCT03695250, NCT03854032, NCT04007588, NCT03417037). Patients affected by advanced solid tumors are treated with BMS-986205 plus nivolumab alone (NCT03792750) or combined with the anti-lymphocyte activating 3 (LAG3) agent relatlimabCitation54,Citation196Citation199 (NCT03459222). The Phase II study NCT03519256, enrolling subjects with high-risk, non-MIBC, is monitoring the therapeutic profile of BMS-986205 combined with two drugs already approved for some types of bladder cancer such as nivolumabCitation200-Citation204 and the toll like receptor 2 (TLR2)/TLR4 agonistCitation205-Citation209 bacillus Calmette–Guérin (BCG).Citation210Citation212 Along similar lines, the Phase III study NCT03661320 compared the efficacy, tolerability and safety of three therapeutic regimens for MIBC: neoadjuvant standard of care chemotherapy with cisplatinCitation213-Citation217 and gemcitabine,Citation218,Citation219 (NAC) versus NAC combined with nivolumab or nivolumab plus BMS-986205, followed by continuation of adjuvant immunotherapy (nivolumab with or without the IDO1 inhibitor) post radical cystectomy.Citation220

Additionally, four ongoing studies aim at elucidating the therapeutic profile of BMS-986205 in combination with nivolumab in patients affected by endometrial carcinoma or endometrial carcinosarcoma (NCT04106414), unresectable or metastatic HCC (NCT03695250), stage II to IV HNSCC (NCT03854032), as well as stage III or IV melanoma (NCT04007588). In particular, the NCT04007588 assay was planned to compare the effectiveness of neoadjuvant PD-1 blockade alone or combined with IDO1 inhibition or with the CTLA4 checkpoint blockers ipilimumab.Citation125Citation127,Citation221,Citation222 However, NCT04007588 has been “Withdrawn” (due to slow accrual). Untreated stage IV or recurrent NSCLC patients were also to be enrolled in NCT03417037, planned to test the combination of BMS-986205 and nivolumab given with or without chemotherapy. Also, NCT03417037 is currently listed as “Withdrawn” (due to changes in the business objectives of the investors). Finally, the safety, side effects and preliminary efficacy of BMS-986205, nivolumab and standard radiation therapy, with or without temozolomide, are being assessed in individuals with newly diagnosed glioblastoma (NCT04047706).

The NCT03915405 clinical assay explores the therapeutic potential of KHK2455 combined with the PD-L1 blocker avelumabCitation223-Citation225 in individuals with locally advanced or metastatic urothelial carcinoma. In particular, the tolerability and safety of the regimen will be evaluated during the first phase of the study (dose-escalation), while pharmacodynamics, pharmacokinetics, and preliminary antitumor activity will be assessed during the expansion phase.

The principal purpose of NCT04049669 is to monitor the efficacy of daily oral administration of indoximodCitation10,Citation226,Citation227 concomitant to chemo-immunotherapy and radiotherapy (for eligible individuals) in subverting immune tolerance and improving clinical outcome of pediatric patients affected by relapsed or refractory glioblastoma, medulloblastoma or ependymoma, as well as by newly-diagnosed diffuse intrinsic pontine glioma (DIPG). In particular, the core therapeutic regimen consists of oral administration of temozolomideCitation228-Citation230 and indoximod, preceded by either low-dose or partial-field radiation (cohort B), or full-dose radiation (cohort C, including all newly diagnosed DIPG patients and some relapsed ependymoma patients), or corresponded to the starting treatment (cohort A, patients not eligible to re-irradiation). If patients accept continuing the indoximod treatment they undergo two “salvage” regimens including either cyclophosphamideCitation189,Citation190,Citation231,Citation232 plus etoposideCitation233,Citation234 or the DNA alkylating agents lomustineCitation235-Citation237 and temozolomide.

Finally, the safety and efficacy of SHR9146 (also known as HTI-1090),Citation238,Citation239 combined with the experimental PD-1 inhibitor SHR-1210 with or without the vascular endothelial growth factor receptor (VEGFR) inhibitor apatinib,Citation240Citation243 are being assessed in patients with advanced or metastatic solid tumors (NCT03491631).

Concluding remarks

Most investigators in the field agree that IDO1 inhibition can synergize with immune checkpoint blockers. While immune checkpoint blockers remove molecular brakes on cytotoxic T cells, they also stimulate the production of IDO1, which, in a negative feedback loop involving AHR activation, shuts down the immune response. Thus, IDO1-targeting drugs should enhance immune checkpoint blockers efficacy. Although our understanding of the biological effects of IDO1 inhibitors is incomplete, these compounds appear to trigger efficient antineoplastic effects along with the re(activation) of anticancer immunosurveillance, at least in preclinical tumor models. However, clinical efficacy remains limited. The exact mechanisms by which IDO1 restrains the immune system as well as the nature of the immune cells affected by IDO1 remains unclear. In particular, precisely determining to which extent IDO1 inhibitors operate on-target may allow for the development of novel agents that would exclusively trigger tumor-targeting immune response without systemic side effects. Indeed, some IDO1 inhibitors directly bind to the AHRCitation88 and could therefore have immunosuppressive effects as Kyn does, which would be the opposite of the drug’s intent. Along the same line, the failure of numerous trials implicating epacadostat has highlighted the need of in-depth research of modes of action before launching combinatorial regimens. Therefore, it appears urgent to disentangle the signaling pathways and metabolic circuitries influenced by IDO1.

Abbreviations

1MT=

1-methyl-tryptophan

ADCC=

Antibody-dependent cellular cytotoxicity

CTL=

Cytotoxic T lymphocyte

EMT=

Epithelial-mesenchymal transition

HCC=

Hepatocellular carcinoma

HNSCC=

Head and neck squamous cell carcinoma

IDO=

Indoleamine 2,3-dioxygenase

IFN=

Interferon

IL=

Interleukin

Kyn=

L-kynurenine

mAb=

Monoclonal antibody

MDSC=

Myeloid-derived suppressor cells

MDT=

Maximum tolerated dose

MIBC=

Muscle-invasive bladder cancer

NK=

Natural killer

NSCLC=

Non-small cell lung cancer

PDT=

Photodynamic therapy

RCC=

Renal cell carcinoma

TRAE=

Treatment-related adverse event

TREG=

Regulatory T

Trp=

L-tryptophan

Acknowledgments

LG is supported by a Breakthrough Level 2 grant from the US Department of Defense (DoD), Breast Cancer Research Program (BRCP) (#BC180476P1), by the 2019 Laura Ziskin Prize in Translational Research (#ZP-6177, PI: Formenti) from the Stand Up to Cancer (SU2C), by a Mantle Cell Lymphoma Research Initiative (MCL-RI, PI: Chen-Kiang) grant from the Leukemia and Lymphoma Society (LLS), by a startup grant from the Dept. of Radiation Oncology at Weill Cornell Medicine (New York, US), by a Rapid Response Grant from the Functional Genomics Initiative (New York, US), by industrial collaborations with Lytix (Oslo, Norway) and Phosplatin (New York, US), and by donations from Phosplatin (New York, US), the Luke Heller TECPR2 Foundation (Boston, US) and Sotio a.s. (Prague, Czech Republic). GK is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; AMMICa US23/CNRS UMS3655; Association pour la recherche sur le cancer (ARC); Association “Le Cancer du Sein, Parlons-en!”; Cancéropôle Ile-de-France; Chancelerie des universités de Paris (Legs Poix), Fondation pour la Recherche Médicale (FRM); a donation by Elior; European Research Area Network on Cardiovascular Diseases (ERA-CVD, MINOTAUR); Gustave Roussy Odyssea, the European Union Horizon 2020 Project Oncobiome; Fondation Carrefour; High-end Foreign Expert Program in China (GDW20171100085), Institut National du Cancer (INCa); Inserm (HTE); Institut Universitaire de France; LeDucq Foundation; the LabEx Immuno-Oncology (ANR-18- IDEX-0001); the RHU Torino Lumière; the Seerave Foundation; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and the SIRIC Cancer Research and Personalized Medicine (CARPEM).

References

  • Dounay AB, Tuttle JB, Verhoest PR. Challenges and opportunities in the discovery of new therapeutics targeting the kynurenine pathway. J Med Chem. 2015;58(22):8762–16. doi:10.1021/acs.jmedchem.5b00461.
  • Mellor AL, Munn DH. Tryptophan catabolism and regulation of adaptive immunity. J Immunol. 2003;170:5809–5813. doi:10.4049/jimmunol.170.12.5809.
  • Van der Leek AP, Yanishevsky Y, Kozyrskyj AL. The kynurenine pathway as a novel link between allergy and the gut microbiome. Front Immunol. 2017;8:1374. doi:10.3389/fimmu.2017.01374.
  • Platten M, Nollen EAA, Rohrig UF, Fallarino F, Opitz CA. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat Rev Drug Discov. 2019;18:379–401.
  • Yoshida R, Imanishi J, Oku T, Kishida T, Hayaishi O. Induction of pulmonary indoleamine 2,3-dioxygenase by interferon. Proc Natl Acad Sci U S A. 1981;78:129–132. doi:10.1073/pnas.78.1.129.
  • Munn DH,  Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, Brown C, Mellor AL. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281:1191–1193. doi:10.1126/science.281.5380.1191.
  • Sidransky H. Tryptophan and carcinogenesis: review and update on how tryptophan may act. Nutr Cancer. 1997;29:181–194. doi:10.1080/01635589709514623.
  • Uyttenhove C, Pilotte L, Théate I, Stroobant V, Colau D, Parmentier N, Boon T, Van den Eynde BJ. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med. 2003;9(10):1269–1274. doi:10.1038/nm934.
  • 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;189:1363–1372. doi:10.1084/jem.189.9.1363.
  • Prendergast GC, Malachowski WJ, Mondal A, Scherle P, Muller AJ. Indoleamine 2,3-dioxygenase and its therapeutic inhibition in cancer. Int Rev Cell Mol Biol. 2018;336:175–203.
  • Prendergast GC, Smith C, Thomas S, Mandik-Nayak L, Laury-Kleintop L, Metz R, Muller AJ. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother. 2014;63(7):721–735. doi:10.1007/s00262-014-1549-4.
  • Terness P, Bauer TM, Röse L, Dufter C, Watzlik A, Simon H, Opelz G. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med. 2002;196:447–457. doi:10.1084/jem.20020052.
  • Rao S, Gharib K, Han A. Cancer Immunosurveillance by T Cells. Int Rev Cell Mol Biol. 2019;342:149–173.
  • 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;196:459–468. doi:10.1084/jem.20020121.
  • Della Chiesa M, Carlomagno S, Frumento G, Balsamo M, Cantoni C, Conte R, Moretta L, Moretta A, Vitale M. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function. Blood. 2006;108(13):4118–4125. doi:10.1182/blood-2006-03-006700.
  • Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Alegre M-L, Puccetti P, et al. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol. 2003;4(12):1206–1212. doi:10.1038/ni1003.
  • Schaaf MB, Garg AD, Agostinis P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018;9:115. doi:10.1038/s41419-017-0061-0.
  • Smith C, Chang MY, Parker KH, Beury DW, DuHadaway JB, Flick HE, Boulden J, Sutanto-Ward E, Soler AP, Laury-Kleintop LD, et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov. 2012;2:722–735. doi:10.1158/2159-8290.CD-12-0014.
  • Bronte V, Brandau S, Chen S-H, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7(1):12150. doi:10.1038/ncomms12150.
  • Elliott LA, Doherty GA, Sheahan K, Ryan EJ. Human tumor-infiltrating myeloid cells: phenotypic and functional diversity. Front Immunol. 2017;8:86. doi:10.3389/fimmu.2017.00086.
  • Gaber T, Chen Y, Krauss PL, Buttgereit F. Metabolism of T lymphocytes in health and disease. Int Rev Cell Mol Biol. 2019;342:95–148.
  • van Baren N, Van den Eynde BJ. Tryptophan-degrading enzymes in tumoral immune resistance. Front Immunol. 2015;6:34. doi:10.3389/fimmu.2015.00034.
  • Eichner R, Fernandez-Saiz V, Targosz BS, Bassermann F. Cross talk networks of mammalian target of rapamycin signaling with the ubiquitin proteasome system and their clinical implications in multiple myeloma. Int Rev Cell Mol Biol. 2019;343:219–297.
  • Bilir C, Sarisozen C. Indoleamine 2,3-dioxygenase (IDO): only an enzyme or a checkpoint controller? J Oncol Sci. 2017;3(2):52–56. doi:10.1016/j.jons.2017.04.001.
  • Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, Ron D, Mellor AL. GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity. 2005;22:633–642. doi:10.1016/j.immuni.2005.03.013.
  • Fougeray S, Mami I, Bertho G, Beaune P, Thervet E, Pallet N. Tryptophan depletion and the kinase GCN2 mediate IFN-gamma-induced autophagy. J Immunol. 2012;189:2954–2964. doi:10.4049/jimmunol.1201214.
  • McGaha TL. IDO-GCN2 and autophagy in inflammation. Oncotarget. 2015;6:21771–21772. doi:10.18632/oncotarget.4846.
  • Nguyen NT, Kimura A, Nakahama T, Chinen I, Masuda K, Nohara K, Fujii-Kuriyama Y, Kishimoto T. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A. 2010;107:19961–19966. doi:10.1073/pnas.1014465107.
  • 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(6):3190–3198. doi:10.4049/jimmunol.0903670.
  • Grohmann U, Puccetti P. The Coevolution of IDO1 and AhR in the emergence of regulatory T-Cells in mammals. Front Immunol. 2015;6:58. doi:10.3389/fimmu.2015.00058.
  • Kotsias F, Cebrian I, Alloatti A. Antigen processing and presentation. Int Rev Cell Mol Biol. 2019;348:69–121.
  • Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. Int Rev Cell Mol Biol. 2019;348:1–68.
  • Derks RA, Jankowska-Gan E, Xu Q, Burlingham WJ. Dendritic cell type determines the mechanism of bystander suppression by adaptive T regulatory cells specific for the minor antigen HA-1. J Immunol. 2007;179:3443–3451. doi:10.4049/jimmunol.179.6.3443.
  • Mellor AL, Baban B, Chandler P, Marshall B, Jhaver K, Hansen A, Koni PA, Iwashima M, Munn DH. Cutting edge: induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. J Immunol. 2003;171:1652–1655. doi:10.4049/jimmunol.171.4.1652.
  • Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL, et al. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114(2):280–290. doi:10.1172/JCI21583.
  • Laurans L, Venteclef N, Haddad Y, Chajadine M, Alzaid F, Metghalchi S, Sovran B, Denis RGP, Dairou J, Cardellini M, et al. Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat Med. 2018;24(8):1113–1120. doi:10.1038/s41591-018-0060-4.
  • Fernando RI, Litzinger M, Trono P, Hamilton DH, Schlom J, Palena C. The T-box transcription factor Brachyury promotes epithelial-mesenchymal transition in human tumor cells. J Clin Invest. 2010;120:533–544. doi:10.1172/JCI38379.
  • Kiyozumi Y, Baba Y, Okadome K, Yagi T, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Watanabe M, Komohara Y, et al. IDO1 expression is associated with immune tolerance and poor prognosis in patients with surgically resected esophageal cancer. Ann Surg. 2019;269:1101–1108. doi:10.1097/SLA.0000000000002754.
  • Yu CP, Fu S-F, Chen X, Ye J, Ye Y, Kong L-D, Zhu Z. The clinicopathological and prognostic significance of IDO1 expression in human solid tumors: evidence from a systematic review and meta-analysis. Cell Physiol Biochem. 2018;49:134–143. doi:10.1159/000492849.
  • Berthon C,Fontenay M, Corm S, Briche I, Allorge D, Hennart B, Lhermitte M, Quesnel B. Metabolites of tryptophan catabolism are elevated in sera of patients with myelodysplastic syndromes and inhibit hematopoietic progenitor amplification. Leuk Res. 2013;37:573–579. doi:10.1016/j.leukres.2013.02.001.
  • Creelan BC, Antonia SJ, Bepler G, Garrett TJ, Simon GR, Soliman HH. Indoleamine 2,3-dioxygenase activity and clinical outcome following induction chemotherapy and concurrent chemoradiation in Stage III non-small cell lung cancer. Oncoimmunology. 2013;2(3):e23428. doi:10.4161/onci.23428.
  • Yoshikawa T, Hara T, Tsurumi H, Goto N, Hoshi M, Kitagawa J, Kanemura N, Kasahara S, Ito H, Takemura M, et al. Serum concentration of L-kynurenine predicts the clinical outcome of patients with diffuse large B-cell lymphoma treated with R-CHOP. Eur J Haematol. 2010;84(4):304–309. doi:10.1111/j.1600-0609.2009.01393.x.
  • 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;11:312–319. doi:10.1038/nm1196.
  • Pan K,Liang X, Zhang H, Zhao J, Wang D, Li J, Lian Q, Chang AE, Li Q, Xia J. Characterization of bridging integrator 1 (BIN1) as a potential tumor suppressor and prognostic marker in hepatocellular carcinoma. Mol Med. 2012;18:507–518. doi:10.2119/molmed.2011.00319.
  • Prendergast GC, Muller AJ, Ramalingam A, Chang MY. BAR the door: cancer suppression by amphiphysin-like genes. Biochim Biophys Acta. 2009;1795:25–36. doi:10.1016/j.bbcan.2008.09.001.
  • Ge K, Duhadaway J, Du W, Herlyn M, Rodeck U, Prendergast GC. Mechanism for elimination of a tumor suppressor: aberrant splicing of a brain-specific exon causes loss of function of Bin1 in melanoma. Proc Natl Acad Sci U S A. 1999;96:9689–9694. doi:10.1073/pnas.96.17.9689.
  • Karni R, de Stanchina E, Lowe SW, Sinha R, Mu D, Krainer AR.The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nat Struct Mol Biol. 2007;14:185–193. doi:10.1038/nsmb1209.
  • Pineda-Lucena A, Ho CSW, Mao DYL, Sheng Y, Laister RC, Muhandiram R, Lu Y, Seet BT, Katz S, Szyperski T, et al. A structure-based model of the c-Myc/Bin1 protein interaction shows alternative splicing of Bin1 and c-Myc phosphorylation are key binding determinants. J Mol Biol. 2005;351(1):182–194. doi:10.1016/j.jmb.2005.05.046.
  • McKenna ES, Tamayo P, Cho Y-J, Tillman EJ, Mora-Blanco EL, Sansam CG, Koellhoffer EC, Pomeroy SL, Roberts CWM. Epigenetic inactivation of the tumor suppressor BIN1 drives proliferation of SNF5-deficient tumors. Cell Cycle. 2012;11(10):1956–1965. doi:10.4161/cc.20280.
  • Radpour R, Barekati Z, Kohler C, Lv Q, Bürki N, Diesch C, Bitzer J, Zheng H, Schmid S, Zhong XY, et al. Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer. PLoS One. 2011;6(1):e16080. doi:10.1371/journal.pone.0016080.
  • Tajiri T, Liu X, Thompson PM, Tanaka S, Suita S, Zhao H, Maris JM, Prendergast GC, Hogarty MD. Expression of a MYCN-interacting isoform of the tumor suppressor BIN1 is reduced in neuroblastomas with unfavorable biological features. Clin Cancer Res. 2003;9:3345–3355.
  • Chang MY, Boulden J, Sutanto-Ward E, Duhadaway JB, Soler AP, Muller AJ, Prendergast GC. Bin1 ablation in mammary gland delays tissue remodeling and drives cancer progression. Cancer Res. 2007;67:100–107. doi:10.1158/0008-5472.CAN-06-2742.
  • Ge K, Duhadaway J, Sakamuro D, Wechsler-Reya R, Reynolds C, Prendergast GC. Losses of the tumor suppressor BIN1 in breast carcinoma are frequent and reflect deficits in programmed cell death capacity. Int J Cancer. 2000;85:376–383.
  • Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. 2018;11:39. doi:10.1186/s13045-018-0582-8.
  • Botticelli A, Cerbelli B, Lionetto L, Zizzari I, Salati M, Pisano A, Federica M, Simmaco M, Nuti M, Marchetti P, et al. Can IDO activity predict primary resistance to anti-PD-1 treatment in NSCLC? J Transl Med. 2018;16:219. doi:10.1186/s12967-018-1595-3.
  • Moon PK, Tran S, Minhas PS. Revisiting IDO and its value as a predictive marker for anti-PD-1 resistance. J Transl Med. 2019;17:31. doi:10.1186/s12967-019-1784-8.
  • Li H, Bullock K, Gurjao C, Braun D, Shukla SA, Bossé D, Lalani AKA, Gopal S, Jin C, Horak C, et al. Metabolomic adaptations and correlates of survival to immune checkpoint blockade. Nat Commun. 2019;10:4346. doi:10.1038/s41467-019-12361-9.
  • Muller AJ, Prendergast GC. Marrying immunotherapy with chemotherapy: why say IDO? Cancer Res. 2005;65:8065–8068. doi:10.1158/0008-5472.CAN-05-2213.
  • Pilotte L, Larrieu P, Stroobant V, Colau D, Dolusic E, Frederick R, De Plaen E, Uyttenhove C, Wouters J, Masereel B, et al. Reversal of tumoral immune resistance by inhibition of tryptophan 2,3-dioxygenase. Proc Natl Acad Sci U S A. 2012;109(7):2497–2502. doi:10.1073/pnas.1113873109.
  • Aris M, Mordoh J, Barrio MM. Immunomodulatory monoclonal antibodies in combined immunotherapy trials for cutaneous melanoma. Front Immunol. 2017;8:1024. doi:10.3389/fimmu.2017.01024.
  • Wang D, Lin J, Yang X, Long J, Bai Y, Yang X, Mao Y, Sang X, Seery S, Zhao H, et al. Combination regimens with PD-1/PD-L1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol. 2019;12(1):42. doi:10.1186/s13045-019-0730-9.
  • Khair DO, Bax HJ, Mele S, Crescioli S, Pellizzari G, Khiabany A, Nakamura M, Harris RJ, French E, Hoffmann RM, et al. Combining immune checkpoint inhibitors: established and emerging targets and strategies to improve outcomes in melanoma. Front Immunol. 2019;10:453. doi:10.3389/fimmu.2019.00453.
  • Sheridan C. IDO inhibitors move center stage in immuno-oncology. Nat Biotechnol. 2015;33:321–322.
  • Hornyak L, Dobos N, Koncz G, Karányi Z, Páll D, Szabó Z, Halmos G, Székvölgyi L, Hellmann MD, Gettinger S. The role of indoleamine-2,3-dioxygenase in cancer development, diagnostics, and therapy. Front Immunol. 2018;9:151. doi:10.3389/fimmu.2018.00151.
  • Cady SG, Sono M. 1-Methyl-DL-tryptophan, beta-(3-benzofuranyl)-DL-alanine (the oxygen analog of tryptophan), and beta-[3-benzo(b)thienyl]-DL-alanine (the sulfur analog of tryptophan) are competitive inhibitors for indoleamine 2,3-dioxygenase. Arch Biochem Biophys. 1991;291:326–333. doi:10.1016/0003-9861(91)90142-6.
  • Hou DY, Muller AJ, Sharma MD, DuHadaway J, Banerjee T, Johnson M, Mellor AL, Prendergast GC, Munn DH. 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.
  • Lob S, Konigsrainer A, Schafer R, Rammensee H-G, Opelz G, Terness P. Levo- but not dextro-1-methyl tryptophan abrogates the IDO activity of human dendritic cells. Blood. 2008;111(4):2152–2154. doi:10.1182/blood-2007-10-116111.
  • Qian F, Villella J, Wallace PK, Mhawech-Fauceglia P, Tario JD Jr, Andrews C, Matsuzaki J, Valmori D, Ayyoub M, Frederick PJ, et al. Efficacy of levo-1-methyl tryptophan and dextro-1-methyl tryptophan in reversing indoleamine-2,3-dioxygenase-mediated arrest of T-cell proliferation in human epithelial ovarian cancer. Cancer Res. 2009;69:5498–5504. doi:10.1158/0008-5472.CAN-08-2106.
  • Fox E, Oliver T, Rowe M, Thomas S, Zakharia Y, Gilman PB, Muller AJ, Prendergast GC. Indoximod: an immunometabolic adjuvant that empowers T cell activity in cancer. Front Oncol. 2018;8:370. doi:10.3389/fonc.2018.00370.
  • Sorgdrager FJH, Naude PJW, Kema IP, Nollen EA, Deyn PP. Tryptophan metabolism in inflammaging: from biomarker to therapeutic target. Front Immunol. 2019;10:2565. doi:10.3389/fimmu.2019.02565.
  • Brown ZJ, Yu SJ, Heinrich B, Ma C, Fu Q, Sandhu M, Agdashian D, Zhang Q, Korangy F, Greten TF, et al. Indoleamine 2,3-dioxygenase provides adaptive resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunol Immunother. 2018;67(8):1305–1315. doi:10.1007/s00262-018-2190-4.
  • Zahm CD, Johnson LE, McNeel DG. Increased indoleamine 2,3-dioxygenase activity and expression in prostate cancer following targeted immunotherapy. Cancer Immunol Immunother. 2019;68:1661–1669. doi:10.1007/s00262-019-02394-w.
  • Liu M, Li Z, Yao W, Zeng X, Wang L, Cheng J, Ma B, Zhang R, Min W, Wang H. IDO inhibitor synergized with radiotherapy to delay tumor growth by reversing T cell exhaustion. Mol Med Rep. 2020;21:445–453. doi:10.3892/mmr.2019.10816.
  • Alqahtani FY, Aleanizy FS, El Tahir E, Alkahtani HM, AlQuadeib BT. Paclitaxel. Profiles Drug Subst Excip Relat Methodol. 2019;44:205–238.
  • Martins I, Raza SQ, Voisin L, Dakhli H, Allouch A, Law F, Sabino D, De Jong D, Thoreau M, Mintet E, et al. Anticancer chemotherapy and radiotherapy trigger both non-cell-autonomous and cell-autonomous death. Cell Death Dis. 2018;9:716. doi:10.1038/s41419-018-0747-y.
  • Castedo M, Perfettini JL, Roumier T, Kroemer G. Cyclin-dependent kinase-1: linking apoptosis to cell cycle and mitotic catastrophe. Cell Death Differ. 2002;9:1287–1293. doi:10.1038/sj.cdd.4401130.
  • Hoffmann J, Vitale I, Buchmann B, Galluzzi L, Schwede W, Senovilla L, Skuballa W, Vivet S, Lichtner RB, Vicencio JM, et al. Improved cellular pharmacokinetics and pharmacodynamics underlie the wide anticancer activity of sagopilone. Cancer Res. 2008;68:5301–5308. doi:10.1158/0008-5472.CAN-08-0237.
  • Hu Z, Zheng B, Xu J, Gao S, Lu W. An albumin-bound drug conjugate of paclitaxel and indoleamine-2,3-dioxygenase inhibitor for enhanced cancer chemo-immunotherapy. Nanotechnology. 2020;31:295101. doi:10.1088/1361-6528/ab824d.
  • Galluzzi L, Kepp O, Kroemer G. Enlightening the impact of immunogenic cell death in photodynamic cancer therapy. Embo J. 2012;31:1055–1057. doi:10.1038/emboj.2012.2.
  • Garg AD, Agostinis P. ER stress, autophagy and immunogenic cell death in photodynamic therapy-induced anti-cancer immune responses. Photochem Photobiol Sci. 2014;13:474–487. doi:10.1039/C3PP50333J.
  • Garg AD, Dudek AM, Ferreira GB, Verfaillie T, Vandenabeele P, Krysko DV, Mathieu C, Agostinis P. ROS-induced autophagy in cancer cells assists in evasion from determinants of immunogenic cell death. Autophagy. 2013;9:1292–1307. doi:10.4161/auto.25399.
  • Tatsuno K, Yamazaki T, Hanlon D, Han P, Robinson E, Sobolev O, Yurter A, Rivera-Molina F, Arshad N, Edelson RL, et al. Extracorporeal photochemotherapy induces bona fide immunogenic cell death. Cell Death Dis. 2019;10(8):578. doi:10.1038/s41419-019-1819-3.
  • Wachowska M, Stachura J, Tonecka K, Fidyt K, Braniewska A, Sas Z, Kotula I, Rygiel TP, Boon L, Golab J, et al. Inhibition of IDO leads to IL-6-dependent systemic inflammation in mice when combined with photodynamic therapy. Cancer Immunol Immunother. 2020. doi:10.1007/s00262-020-02528-5.
  • Shen S, Niso-Santano M, Adjemian S, Takehara T, Malik S, Minoux H, Souquere S, Mariño G, Lachkar S, Senovilla L, et al. Cytoplasmic STAT3 represses autophagy by inhibiting PKR activity. Mol Cell. 2012;48(5):667–680. doi:10.1016/j.molcel.2012.09.013.
  • Yang H, Yamazaki T, Pietrocola F, Zhou H, Zitvogel L, Ma Y, Kroemer G, Gust KM, Shariat SF. STAT3 inhibition enhances the therapeutic efficacy of immunogenic chemotherapy by stimulating type 1 interferon production by cancer cells. Cancer Res. 2015;75(18):3812–3822. doi:10.1158/0008-5472.CAN-15-1122.
  • Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer. 2009;9:798–809. doi:10.1038/nrc2734.
  • Johnson DE, O’Keefe RA, Grandis JR. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 2018;15:234–248. doi:10.1038/nrclinonc.2018.8.
  • Litzenburger UM, Opitz CA, Sahm F, Rauschenbach KJ, Trump S, Winter M, Ott M, Ochs K, Lutz C, Liu X, 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(4):1038–1051. doi:10.18632/oncotarget.1637.
  • Yan D, Lin YW, Tan X. Heme-containing enzymes and inhibitors for tryptophan metabolism. Metallomics. 2017;9:1230–1240. doi:10.1039/C7MT00105C.
  • Nafia I, Toulmonde M, Bortolotto D, Chaibi A, Bodet D, Rey C, Velasco V, Larmonier CB, Cerf L, Adam J, et al. IDO targeting in sarcoma: biological and clinical implications. Front Immunol. 2020;11:274. doi:10.3389/fimmu.2020.00274.
  • Ma Y, Adjemian S, Mattarollo S, Yamazaki T, Aymeric L, Yang H, Portela Catani J, Hannani D, Duret H, Steegh K, et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38(4):729–741. doi:10.1016/j.immuni.2013.03.003.
  • Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, Vitale I, Goubar A, Baracco EE, Remédios C, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20:1301–1309. doi:10.1038/nm.3708.
  • Vacchelli E, Ma Y, Baracco EE, Sistigu A, Enot DP, Pietrocola F, Yang H, Adjemian S, Chaba K, Semeraro M, et al. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science. 2015;350(6263):972–978. doi:10.1126/science.aad0779.
  • Gao J, Deng F, Jia W. Inhibition of Indoleamine 2,3-dioxygenase enhances the therapeutic efficacy of immunogenic chemotherapeutics in breast cancer. J Breast Cancer. 2019;22:196–209. doi:10.4048/jbc.2019.22.e23.
  • Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2005;202:1691–1701. doi:10.1084/jem.20050915.
  • Tumang J, Gomes B, Wythes M, Crosignani S, Bingham P, Bottemanne P, Cannelle H, Cauwenberghs S, Chaplin J, Dalvie D, et al. PF-06840003: a highly selective IDO-1 inhibitor that shows good in vivo efficacy in combination with immune checkpoint inhibitors. Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4863..
  • Gomes B, Driessens G, Bartlett D, Cai D, Cauwenberghs S, Crosignani S, Dalvie D, Denies S, Dillon CP, Fantin VR, et al. Characterization of the selective indoleamine 2,3-dioxygenase-1 (IDO1) catalytic inhibitor EOS200271/PF-06840003 Supports IDO1 as a critical resistance mechanism to PD-(L)1 blockade therapy. Mol Cancer Ther. 2018;17(12):2530–2542. doi:10.1158/1535-7163.MCT-17-1104.
  • Ladomersky E, Zhai L, Lenzen A, Lauing KL, Qian J, Scholtens DM, Gritsina G, Sun X, Liu Y, Yu F, et al. IDO1 inhibition synergizes with radiation and pd-1 blockade to durably increase survival against advanced glioblastoma. Clin Cancer Res. 2018;24(11):2559–2573. doi:10.1158/1078-0432.CCR-17-3573.
  • Deutsch E, Chargari C, Galluzzi L, Kroemer G. Optimising efficacy and reducing toxicity of anticancer radioimmunotherapy. Lancet Oncol. 2019;20:e452–e463. doi:10.1016/S1470-2045(19)30171-8.
  • Davar D, Bahary N. Modulating tumor immunology by inhibiting indoleamine 2,3-dioxygenase (IDO): recent developments and first clinical experiences. Target Oncol. 2018;13:125–140. doi:10.1007/s11523-017-0547-9.
  • Bai Z, Huang H, Chen J, Zhang X, Ding Y. Identification of novel imidazoles as IDO1 inhibitors through microwave-assisted one-pot multicomponent reactions. Arch Pharm (Weinheim). 2019;352:e1900165. doi:10.1002/ardp.201900165.
  • Cheng MF, Hung M-S, Song J-S, Lin S-Y, Liao F-Y, Wu M-H, Hsiao W, Hsieh C-L, Wu J-S, Chao Y-S, et al. Discovery and structure-activity relationships of phenyl benzenesulfonylhydrazides as novel indoleamine 2,3-dioxygenase inhibitors. Bioorg Med Chem Lett. 2014;24:3403–3406. doi:10.1016/j.bmcl.2014.05.084.
  • Winters M, DuHadaway JB, Pham KN, Lewis-Ballester A, Badir S, Wai J, Sheikh E, Yeh S-R, Prendergast GC, Muller AJ, et al. Diaryl hydroxylamines as pan or dual inhibitors of indoleamine 2,3-dioxygenase-1, indoleamine 2,3-dioxygenase-2 and tryptophan dioxygenase. Eur J Med Chem. 2019;162:455–464. doi:10.1016/j.ejmech.2018.11.010.
  • Fu R, Zhang Y-W, Li H-M, Lv W-C, Zhao L, Guo Q-L, Lu T, Weiss SJ, Li Z-Y, Wu Z-Q, et al. LW106, a novel indoleamine 2,3-dioxygenase 1 inhibitor, suppresses tumour progression by limiting stroma-immune crosstalk and cancer stem cell enrichment in tumour micro-environment. Br J Pharmacol. 2018;175(14):3034–3049. doi:10.1111/bph.14351.
  • Prendergast GC, Malachowski WP, DuHadaway JB, Muller AJ. Discovery of IDO1 Inhibitors: from Bench to Bedside. Cancer Res. 2017;77:6795–6811. doi:10.1158/0008-5472.CAN-17-2285.
  • Heeren AM, van Dijk I, Berry DRAI, Khelil M, Ferns D, Kole J, Musters RJP, Thijssen VL, Mom CH, Kenter GG, et al. Indoleamine 2,3-dioxygenase expression pattern in the tumor microenvironment predicts clinical outcome in early stage cervical cancer. Front Immunol. 2018;9:1598. doi:10.3389/fimmu.2018.01598.
  • Li H, Ning S, Ghandi M, Kryukov GV, Gopal S, Deik A, Souza A, Pierce K, Keskula P, Hernandez D, et al. The landscape of cancer cell line metabolism. Nat Med. 2019;25:850–860. doi:10.1038/s41591-019-0404-8.
  • Wong DJ, Hurvitz SA. Recent advances in the development of anti-HER2 antibodies and antibody-drug conjugates. Ann Transl Med. 2014;2:122.
  • Baselga J, Albanell J. Mechanism of action of anti-HER2 monoclonal antibodies. Ann Oncol. 2001;12(Suppl 1):S35–41. doi:10.1093/annonc/12.suppl_1.S35.
  • Su S, Zhao J, Xing Y, Zhang X, Liu J, Ouyang Q, Chen J, Su F, Liu Q, Song E, et al. Immune checkpoint inhibition overcomes ADCP-induced immunosuppression by macrophages. Cell. 2018;175(442–457):e423. doi:10.1016/j.cell.2018.09.007.
  • Antibody-dependent cellular cytotoxicity renders macrophages immunosuppressive. Cancer Discov. October 12 2018. doi:10.1158/2159-8290.CD-RW2018-176
  • Lopez-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK Cells. Cancer Cell. 2017;32:135–154. doi:10.1016/j.ccell.2017.06.009.
  • Vanpouille-Box C, Demaria S, Formenti SC, Galluzzi L. Cytosolic DNA sensing in organismal tumor control. Cancer Cell. 2018;34:361–378. doi:10.1016/j.ccell.2018.05.013.
  • Kroemer G, Senovilla L, Galluzzi L, Andre F, Zitvogel L. Natural and therapy-induced immunosurveillance in breast cancer. Nat Med. 2015;21:1128–1138. doi:10.1038/nm.3944.
  • Mittal D, Vijayan D, Neijssen J, Kreijtz J, Habraken MMJM, Van Eenennaam H, Van Elsas A, Smyth MJ. Blockade of ErbB2 and PD-L1 using a bispecific antibody to improve targeted anti-ErbB2 therapy. Oncoimmunology. 2019;8:e1648171. doi:10.1080/2162402X.2019.1648171.
  • Triulzi T, Forte L, Regondi V, Di Modica M, Ghirelli C, Carcangiu ML, Sfondrini L, Balsari A, Tagliabue E. HER2 signaling regulates the tumor immune microenvironment and trastuzumab efficacy. Oncoimmunology. 2019;8:e1512942. doi:10.1080/2162402X.2018.1512942.
  • Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, de Azambuja E, Castro G, Untch M, Smith I, Gianni L, et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. Lancet. 2017;389:1195–1205. doi:10.1016/S0140-6736(16)32616-2.
  • Tolaney SM, Wardley AM, Zambelli S, Hilton JF, Troso-Sandoval TA, Ricci F, Im S-A, Kim S-B, Johnston SR, Chan A, et al. Abemaciclib plus trastuzumab with or without fulvestrant versus trastuzumab plus standard-of-care chemotherapy in women with hormone receptor-positive, HER2-positive advanced breast cancer (monarcHER): a randomised, open-label, phase 2 trial. Lancet Oncol. 2020;21(6):763–775. doi:10.1016/S1470-2045(20)30112-1.
  • Dumas A, Vaz Luis I, Bovagnet T, El Mouhebb M, Di Meglio A, Pinto S, Charles C, Dauchy S, Delaloge S, Arveux P, et al. Impact of breast cancer treatment on employment: results of a multicenter prospective cohort study (CANTO). J Clin Oncol. 2020;38:734–743. doi:10.1200/JCO.19.01726.
  • Modi S, Saura C, Yamashita T, Park YH, Kim S-B, Tamura K, Andre F, Iwata H, Ito Y, Tsurutani J, et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N Engl J Med. 2020;382(7):610–621. doi:10.1056/NEJMoa1914510.
  • Komrokji RS, Wei S, Mailloux AW, Zhang L, Padron E, Sallman D, Lancet JE, Tinsley S, Nardelli LA, Pinilla-Ibarz J, et al. A phase II study to determine the safety and efficacy of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO) enzyme INCB024360 in patients with myelodysplastic syndromes. Clin Lymphoma Myeloma Leuk. 2019;19(3):157–161. doi:10.1016/j.clml.2018.12.005.
  • Cseh AM, Niemeyer CM, Yoshimi A, Catala A, Frühwald MC, Hasle H, van den Heuvel-eibrink MM, Lauten M, De Moerloose B, Smith OP, et al. Therapy with low-dose azacitidine for MDS in children and young adults: a retrospective analysis of the EWOG-MDS study group. Br J Haematol. 2016;172(6):930–936. doi:10.1111/bjh.13915.
  • Jabbour E, Short NJ, Montalban-Bravo G, Huang X, Bueso-Ramos C, Qiao W, Yang H, Zhao C, Kadia T, Borthakur G, et al. Randomized phase 2 study of low-dose decitabine vs low-dose azacitidine in lower-risk MDS and MDS/MPN. Blood. 2017;130(13):1514–1522. doi:10.1182/blood-2017-06-788497.
  • Scott LJ. Azacitidine: a review in myelodysplastic syndromes and acute myeloid leukaemia. Drugs. 2016;76:889–900. doi:10.1007/s40265-016-0585-0.
  • Kverneland AH, Enevold C, Donia M, Bastholt L, Svane IM, Nielsen CH. Development of anti-drug antibodies is associated with shortened survival in patients with metastatic melanoma treated with ipilimumab. Oncoimmunology. 2018;7(5):e1424674. doi:10.1080/2162402X.2018.1424674.
  • Madonna G, Ballesteros-Merino C, Feng Z, Bifulco C, Capone M, Giannarelli D, Mallardo D, Simeone E, Grimaldi AM, Caracò C, et al. PD-L1 expression with immune-infiltrate evaluation and outcome prediction in melanoma patients treated with ipilimumab. Oncoimmunology. 2018;7(12):e1405206. doi:10.1080/2162402X.2017.1405206.
  • Wu X, Giobbie-Hurder A, Connolly EM, Li J, Liao X, Severgnini M, Zhou J, Rodig S, Hodi FS. Anti-CTLA-4 based therapy elicits humoral immunity to galectin-3 in patients with metastatic melanoma. Oncoimmunology. 2018;7(7):e1440930. doi:10.1080/2162402X.2018.1440930.
  • Gibney GT, Hamid O, Lutzky J, Olszanski AJ, Mitchell TC, Gajewski TF, Chmielowski B, Hanks BA, Zhao Y, Newton RC, et al. Phase 1/2 study of epacadostat in combination with ipilimumab in patients with unresectable or metastatic melanoma. J Immunother Cancer. 2019;7(1):80. doi:10.1186/s40425-019-0562-8.
  • Mohan N, Hosain S, Zhao J, Shen Y, Luo X, Jiang J, Endo Y, Wu WJ. Atezolizumab potentiates Tcell-mediated cytotoxicity and coordinates with FAK to suppress cell invasion and motility in PD-L1+triple negative breast cancer cells. Oncoimmunology. 2019;8(9):e1624128. doi:10.1080/2162402X.2019.1624128.
  • Nie W, Qian J, Xu M-D, Gu K, Qian -F-F, Hu M-J, Lu J, Gan L, Zhang X-Y, Cao S-H, et al. A non-linear association between blood tumor mutation burden and prognosis in NSCLC patients receiving atezolizumab. Oncoimmunology. 2020;9(1):1731072. doi:10.1080/2162402X.2020.1731072.
  • Vitale I, Sistigu A, Manic G, Rudqvist N-P, Trajanoski Z, Galluzzi L. Mutational and antigenic landscape in tumor progression and cancer immunotherapy. Trends Cell Biol. 2019;29(5):396–416. doi:10.1016/j.tcb.2019.01.003.
  • Michels J, Vitale I, Galluzzi L, Adam J, Olaussen KA, Kepp O, Senovilla L, Talhaoui I, Guegan J, Enot DP, et al. Cisplatin resistance associated with PARP hyperactivation. Cancer Res. 2013;73(7):2271–2280. doi:10.1158/0008-5472.CAN-12-3000.
  • Vacchelli E, Galluzzi L, Rousseau V, Rigoni A, Tesniere A, Delahaye N, Schlemmer F, Menger L, Sukkurwala AQ, Adjemian S, et al. Loss-of-function alleles ofP2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology. 2012;1(3):271–278. doi:10.4161/onci.18684.
  • Filipits M, Pirker R, Dunant A, Lantuejoul S, Schmid K, Huynh A, Haddad V, André F, Stahel R, Pignon J-P, et al. Cell cycle regulators and outcome of adjuvant cisplatin-based chemotherapy in completely resected non-small-cell lung cancer: the international adjuvant lung cancer trial biologic program. J Clin Oncol. 2007;25:2735–2740. doi:10.1200/JCO.2006.08.2867.
  • Yamazaki T, Buque A, Ames TD, Galluzzi L. PT-112 induces immunogenic cell death and synergizes with immune checkpoint blockers in mouse tumor models. Oncoimmunology. 2020;9:1721810. doi:10.1080/2162402X.2020.1721810.
  • Olaussen KA, Dunant A, Fouret P, Brambilla E, André F, Haddad V, Taranchon E, Filipits M, Pirker R, Popper HH, et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med. 2006;355:983–991. doi:10.1056/NEJMoa060570.
  • Al-Batran SE, Homann N, Pauligk C, Goetze TO, Meiler J, Kasper S, Kopp H-G, Mayer F, Haag GM, Luley K, et al. Perioperative chemotherapy with fluorouracil plus leucovorin, oxaliplatin, and docetaxel versus fluorouracil or capecitabine plus cisplatin and epirubicin for locally advanced, resectable gastric or gastro-oesophageal junction adenocarcinoma (FLOT4): a randomised, phase 2/3 trial. Lancet. 2019;393:1948–1957. doi:10.1016/S0140-6736(18)32557-1.
  • Fuchs CS, Niedzwiecki D, Mamon HJ, Tepper JE, Ye X, Swanson RS, Enzinger PC, Haller DG, Dragovich T, Alberts SR, et al. Adjuvant Chemoradiotherapy with epirubicin, cisplatin, and fluorouracil compared with adjuvant chemoradiotherapy with fluorouracil and leucovorin after curative resection of gastric cancer: results from CALGB 80101 (Alliance). J Clin Oncol. 2017;35:3671–3677. doi:10.1200/JCO.2017.74.2130.
  • Kang YK, Cho H. Perioperative FLOT: new standard for gastric cancer? Lancet. 2019;393:1914–1916. doi:10.1016/S0140-6736(18)33189-1.
  • Hellmann MD, Gettinger S, Chow LQM, Gordon M, Awad MM, Cha E, Gong X, Zhou G, Walker C, Leopold L, et al. Phase 1 study of epacadostat in combination with atezolizumab for patients with previously treated advanced nonsmall cell lung cancer. Int J Cancer. 2020. doi:10.1002/ijc.32951.
  • Mitchell TC, Hamid O, Smith DC, Bauer TM, Wasser JS, Olszanski AJ, Luke JJ, Balmanoukian AS, Schmidt EV, Zhao Y, et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase i results from a multicenter, open-label phase I/II Trial (ECHO-202/KEYNOTE-037). J Clin Oncol. 2018;36:3223–3230. doi:10.1200/JCO.2018.78.9602.
  • Long GV, Dummer R, Hamid O, Gajewski TF, Caglevic C, Dalle S, Arance A, Carlino MS, Grob -J-J, Kim TM, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20:1083–1097. doi:10.1016/S1470-2045(19)30274-8.
  • Luther C, Swami U, Zhang J, Milhem M, Zakharia Y. Advanced stage melanoma therapies: detailing the present and exploring the future. Crit Rev Oncol Hematol. 2019;133:99–111. doi:10.1016/j.critrevonc.2018.11.002.
  • Lara P, Bauer TM, Hamid O, Smith DC, Gajewski TF, Gangadhar TC, Somer BG, Schmidt EV, Zhang Y, Gowda H, et al. Epacadostat plus pembrolizumab in patients with advanced RCC: preliminary phase I/II results from ECHO-202/KEYNOTE-037. J Clin Oncol. 2017;35:15_suppl, 4515–4515
  • Mazzarella L, Duso BA, Trapani D, Belli C, D’Amico P, Ferraro E, Viale G, Curigliano G. The evolving landscape of ‘next-generation’ immune checkpoint inhibitors: A review. Eur J Cancer. 2019;117:14–31. doi:10.1016/j.ejca.2019.04.035.
  • Garber K. A new cancer immunotherapy suffers a setback. Science. 2018;360:588. doi:10.1126/science.360.6389.588.
  • Hafner A, Bulyk ML, Jambhekar A, Lahav G. The multiple mechanisms that regulate p53 activity and cell fate. Nat Rev Mol Cell Biol. 2019;20:199–210. doi:10.1038/s41580-019-0110-x.
  • Galluzzi L, Morselli E, Kepp O, Tajeddine N, Kroemer G. Targeting p53 to mitochondria for cancer therapy. Cell Cycle. 2008;7:1949–1955. doi:10.4161/cc.7.13.6222.
  • Bykov VJN, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 2018;18:89–102. doi:10.1038/nrc.2017.109.
  • Soliman H, Khambati F, Han HS, Ismail-Khan R, Bui MM, Sullivan DM, Antonia S. A phase-1/2 study of adenovirus-p53 transduced dendritic cell vaccine in combination with indoximod in metastatic solid tumors and invasive breast cancer. Oncotarget. 2018;9(11):10110–10117. doi:10.18632/oncotarget.24118.
  • Bahary N, Wang-Gillam A, Somer BG, Lee JS, O’Rourke MA, Nayak-Kapoor A, Beatty GL, Liu M, Delman D, Rossi GR, et al. Phase 2 trial of the IDO pathway inhibitor indoximod plus gemcitabine/nab-paclitaxel for the treatment of patients with metastatic pancreas cancer. J Clin Oncol. 2018; 36:4015–4015.
  • Nayak-Kapoor A, Hao Z, Sadek R, Dobbins R, Marshall L, Vahanian NN, Jay Ramsey W, Kennedy E, Mautino MR, Link CJ, et al. Phase Ia study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) in patients with recurrent advanced solid tumors. J Immunother Cancer. 2018;6(1):61. doi:10.1186/s40425-018-0351-9.
  • Ebata T, Shimizu T, Fujiwara Y, Tamura K, Kondo S, Iwasa S, Yonemori K, Shimomura A, Kitano S, Koyama T, et al. Phase I study of the indoleamine 2,3-dioxygenase 1 inhibitor navoximod (GDC-0919) as monotherapy and in combination with the PD-L1 inhibitor atezolizumab in Japanese patients with advanced solid tumours. Invest New Drugs. 2020;38(2):468–477. doi:10.1007/s10637-019-00787-3.
  • Jung KH, LoRusso P, Burris H, Gordon M, Bang Y-J, Hellmann MD, Cervantes A, de Olza MO, Marabelle A, Hodi FS, et al. Phase I Study of the Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitor Navoximod (GDC-0919) Administered with PD-L1 Inhibitor (Atezolizumab) in Advanced Solid Tumors. Clin Cancer Res. 2019;25(11):3220–3228. doi:10.1158/1078-0432.CCR-18-2740.
  • Ricciuti B, Leonardi GC, Puccetti P, Fallarino F, Bianconi V, Sahebkar A, Baglivo S, Chiari R, Pirro M. Targeting indoleamine-2,3-dioxygenase in cancer: scientific rationale and clinical evidence. Pharmacol Ther. 2019;196:105–116. doi:10.1016/j.pharmthera.2018.12.004.
  • Luke JJ, Tabernero J, Joshua A, Desai J, Varga AI, Moreno V, Gomez-Roca CA, Markman B, De Braud FG, Patel SP, et al. BMS-986205, an indoleamine 2, 3-dioxygenase 1 inhibitor (IDO1i), in combination with nivolumab (nivo): updated safety across all tumor cohorts and efficacy in advanced bladder cancer (advBC). J Clin Oncol. 2019;37:7. doi:10.1200/JCO.2019.37.7_suppl.358.
  • Tabernero J, Luke JJ, Joshua AM, Varga AI, Moreno V, Desai J, Markman B, Gomez-Roca CA, De Braud FG, Patel SP, et al. BMS-986205, an indoleamine 2,3-dioxygenase 1 inhibitor (IDO1i), in combination with nivolumab (NIVO): updated safety across all tumor cohorts and efficacy in pts with advanced bladder cancer (advBC). J Clin Oncol. 2018;36(15_suppl):4512. doi:10.1200/JCO.2018.36.15_suppl.4512.
  • Siu LL, Gelmon K, Chu Q, Pachynski R, Alese O, Basciano P, Walker J, Mitra P, Zhu L, Phillips P, et al. BMS-986205, an optimized indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor, is well tolerated with potent pharmacodynamic (PD) activity, alone and in combination with nivolumab (nivo) in advanced cancers in a phase 1/2a trial.
  • Redman JM, Steinberg SM, Gulley JL. Quick efficacy seeking trial (QuEST1): a novel combination immunotherapy study designed for rapid clinical signal assessment metastatic castration-resistant prostate cancer. J Immunother Cancer. 2018;6:91. doi:10.1186/s40425-018-0409-8.
  • Nouri M, Ratther E, Stylianou N, Nelson CC, Hollier BG, Williams ED. Androgen-targeted therapy-induced epithelial mesenchymal plasticity and neuroendocrine transdifferentiation in prostate cancer: an opportunity for intervention. Front Oncol. 2014;4:370. doi:10.3389/fonc.2014.00370.
  • Palena C, Polev DE, Tsang KY, Fernando RI, Litzinger M, Krukovskaya LL, Baranova AV, Kozlov AP, Schlom J. The human T-box mesodermal transcription factor Brachyury is a candidate target for T-cell-mediated cancer immunotherapy. Clin Cancer Res. 2007;13:2471–2478. doi:10.1158/1078-0432.CCR-06-2353.
  • Pinto F, Pertega-Gomes N, Pereira MS, Vizcaino JR, Monteiro P, Henrique RM, Baltazar F, Andrade RP, Reis RM. T-box transcription factor brachyury is associated with prostate cancer progression and aggressiveness. Clin Cancer Res. 2014;20:4949–4961. doi:10.1158/1078-0432.CCR-14-0421.
  • Thoma C. Prostate cancer: brachyury–a biomarker for progression and prognosis? Nat Rev Urol. 2014;11:485. doi:10.1038/nrurol.2014.184.
  • David JM, Dominguez C, McCampbell KK, Gulley JL, Schlom J, Palena C. A novel bifunctional anti-PD-L1/TGF-beta Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. Oncoimmunology. 2017;6:e1349589. doi:10.1080/2162402X.2017.1349589.
  • Lan Y, Zhang D, Xu C, Hance KW, Marelli B, Qi J, Yu H, Qin G, Sircar A, Hernández VM, et al. Enhanced preclinical antitumor activity of M7824, a bifunctional fusion protein simultaneously targeting PD-L1 and TGF-beta. Sci Transl Med. 2018;10. doi:10.1126/scitranslmed.aan5488
  • Strauss J, Heery CR, Schlom J, Madan RA, Cao L, Kang Z, Lamping E, Marté JL, Donahue RN, Grenga I, et al. Phase I Trial of M7824 (MSB0011359C), a Bifunctional Fusion Protein Targeting PD-L1 and TGFbeta, in Advanced Solid Tumors. Clin Cancer Res. 2018;24:1287–1295. doi:10.1158/1078-0432.CCR-17-2653.
  • Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, Ratikan JA, Felix C, Hwang L, Faull KF, et al. Focal irradiation and systemic TGFbeta blockade in metastatic breast cancer. Clin Cancer Res. 2018;24:2493–2504. doi:10.1158/1078-0432.CCR-17-3322.
  • Kim PS, Kwilas AR, Xu W, Alter S, Jeng EK, Wong HC, Schlom J, Hodge JW. IL-15 superagonist/IL-15RalphaSushi-Fc fusion complex (IL-15SA/IL-15RalphaSu-Fc; ALT-803) markedly enhances specific subpopulations of NK and memory CD8+ T cells, and mediates potent anti-tumor activity against murine breast and colon carcinomas. Oncotarget. 2016;7:16130–16145. doi:10.18632/oncotarget.7470.
  • Fucikova J, Kline JP, Galluzzi L, Spisek R. Calreticulin arms NK cells against leukemia. Oncoimmunology. 2020;9:1671763. doi:10.1080/2162402X.2019.1671763.
  • Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 2016;15:385–403. doi:10.1038/nrd.2015.17.
  • Kazazi-Hyseni F, Beijnen JH, Schellens JH. Bevacizumab. Oncologist. 2010;15:819–825. doi:10.1634/theoncologist.2009-0317.
  • Mehnert JM, Joshua AM, Lakhani N, Banerji U, Rasco DW, Lugowska I, Tomaszewska-Kiecana M, Garralda E, Kornacki DL, Sumrow BJ, et al. First-in-Human Phase 1 Study of INCMGA00012 in patients with advanced solid tumors: interim results of the cohort expansion phase. 33rd Annual meeting of the society for immunotherapy of cancer poster 669(2018).Washington, DC, USA [2018 November 7–11].
  • Ansell SM. Pembrolizumab: living up to expectations. Blood. 2019;134:1114–1115. doi:10.1182/blood.2019002417.
  • Kroemer G, Galluzzi L. Combinatorial immunotherapy with checkpoint blockers solves the problem of metastatic melanoma-An exclamation sign with a question mark. Oncoimmunology. 2015;4:e1058037. doi:10.1080/2162402X.2015.1058037.
  • Kuryk L, Moller AW, Jaderberg M. Combination of immunogenic oncolytic adenovirus ONCOS-102 with anti-PD-1 pembrolizumab exhibits synergistic antitumor effect in humanized A2058 melanoma huNOG mouse model. Oncoimmunology. 2019;8:e1532763. doi:10.1080/2162402X.2018.1532763.
  • Nowacki TM, Kuerten S, Zhang W, Shive CL, Kreher CR, Boehm BO, Lehmann PV, Tary-Lehmann M. Granzyme B production distinguishes recently activated CD8(+) memory cells from resting memory cells. Cell Immunol. 2007;247:36–48. doi:10.1016/j.cellimm.2007.07.004.
  • Semeraro M, Adam J, Stoll G, Louvet E, Chaba K, Poirier-Colame V, Sauvat A, Senovilla L, Vacchelli E, Bloy N, et al. The ratio of CD8 +/FOXP3 T lymphocytes infiltrating breast tissues predicts the relapse of ductal carcinoma in situ. Oncoimmunology. 2016;5:e1218106. doi:10.1080/2162402X.2016.1218106.
  • Senovilla L, Vitale I, Martins I, Tailler M, Pailleret C, Michaud M, Galluzzi L, Adjemian S, Kepp O, Niso-Santano M, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science. 2012;337:1678–1684. doi:10.1126/science.1224922.
  • Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16:356–371. doi:10.1038/s41571-019-0175-7.
  • Fridman WH, Zitvogel L, Sautes-Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717–734. doi:10.1038/nrclinonc.2017.101.
  • Vacchelli E, Semeraro M, Adam J, Dartigues P, Zitvogel L, Kroemer G. Immunosurveillance in esophageal carcinoma: the decisive impact of regulatory T cells. Oncoimmunology. 2016;5:e1064581. doi:10.1080/2162402X.2015.1064581.
  • Vacchelli E, Semeraro M, Enot DP, Chaba K, Colame VP, Dartigues P, Perier A, Villa I, Rusakiewicz S, Gronnier C, et al. Negative prognostic impact of regulatory T cell infiltration in surgically resected esophageal cancer post-radiochemotherapy. Oncotarget. 2015;6(25):20840–20850. doi:10.18632/oncotarget.4428.
  • Necchi A, Anichini A, Raggi D, Briganti A, Massa S, Lucianò R, Colecchia M, Giannatempo P, Mortarini R, Bianchi M, et al. Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase II Study. J Clin Oncol. 2018;36:3353–3360. doi:10.1200/JCO.18.01148.
  • Gust KM, Shariat SF. Re: pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase II study. Eur Urol. 2019;75:695–696. doi:10.1016/j.eururo.2018.12.034.
  • Zhang S. Neoadjuvant Immunotherapy in muscle-invasive bladder cancer: time to change clinical practice? J Clin Oncol. 2019;37:939. doi:10.1200/JCO.18.01864.
  • Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee J-L, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015–1026. doi:10.1056/NEJMoa1613683.
  • Chu CE, Porten SP, Grossfeld GD, Meng MV. Role of indoleamine-2,3-dioxygenase inhibitors in salvage therapy for non-muscle invasive bladder cancer. Urol Clin North Am. 2020;47:111–118. doi:10.1016/j.ucl.2019.09.013.
  • Hoehler T, von Wichert G, Schimanski C, Kanzler S, Moehler MH, Hinke A, Seufferlein T, Siebler J, Hochhaus A, Arnold D, et al. Phase I/II trial of capecitabine and oxaliplatin in combination with bevacizumab and imatinib in patients with metastatic colorectal cancer: AIO KRK 0205. Br J Cancer. 2013;109:1408–1413. doi:10.1038/bjc.2013.409.
  • Pol J, Vacchelli E, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautès-Fridman C, Fucikova J, Galon J, Spisek R, et al. Trial Watch: immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology. 2015;4(4):e1008866. doi:10.1080/2162402X.2015.1008866.
  • Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2014;3(3):e27878. doi:10.4161/onci.27878.
  • Vanmeerbeek I, Sprooten J, De Ruysscher D, Tejpar S, Vandenberghe P, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L, et al. Trial watch: chemotherapy-induced immunogenic cell death in immuno-oncology. Oncoimmunology. 2020;9(1):1703449. doi:10.1080/2162402X.2019.1703449.
  • Ciotti M, Angeletti S, Minieri M, Giovannetti M, Benvenuto D, Pascarella S, Sagnelli C, Bianchi M, Bernardini S, Ciccozzi M, et al. COVID-19 outbreak: an overview. Chemotherapy. 2020:1–9. doi:10.1159/000507423
  • Raoult D, Zumla A, Locatelli F, Ippolito G, Kroemer G. Coronavirus infections: epidemiological, clinical and immunological features and hypotheses. Cell Stress. 2020;4:66–75. doi:10.15698/cst2020.04.216.
  • Rothan HA, Byrareddy SN. The epidemiology and pathogenesis of coronavirus disease (COVID-19) outbreak. J Autoimmun. 2020;109:102433. doi:10.1016/j.jaut.2020.102433.
  • Costantini A, Julie C, Dumenil C, Hélias-Rodzewicz Z, Tisserand J, Dumoulin J, Giraud V, Labrune S, Chinet T, Emile J-F, et al. Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. Oncoimmunology. 2018;7:e1452581. doi:10.1080/2162402X.2018.1452581.
  • Ruffo E, Wu RC, Bruno TC, Workman CJ, Vignali DAA. Lymphocyte-activation gene 3 (LAG3): the next immune checkpoint receptor. Semin Immunol. 2019;42:101305. doi:10.1016/j.smim.2019.101305.
  • Solinas C, Migliori E, De Silva P, Willard-Gallo K. LAG3: the biological processes that motivate targeting this immune checkpoint molecule in human cancer. Cancers (Basel). 2019;11(8):1213. doi:10.3390/cancers11081213.
  • Long L, Zhang X, Chen F, Pan Q, Phiphatwatchara P, Zeng Y, Chen H. The promising immune checkpoint LAG-3: from tumor microenvironment to cancer immunotherapy. Genes Cancer. 2018;9:176–189. doi:10.18632/genesandcancer.180.
  • Yu X, Huang X, Chen X, Liu J, Wu C, Pu Q, Wang Y, Kang X, Zhou L. Characterization of a novel anti-human lymphocyte activation gene 3 (LAG-3) antibody for cancer immunotherapy. MAbs. 2019;11:1139–1148. doi:10.1080/19420862.2019.1629239.
  • Nivolumab gets FDA nod for bladder cancer. Cancer Discov. 2017;7:OF7. doi:10.1158/2159-8290.CD-NB2017-021. Epub 2017 Feb 9.
  • Ghatalia P, Zibelman M, Geynisman DM, Plimack E. Approved checkpoint inhibitors in bladder cancer: which drug should be used when? Ther Adv Med Oncol. 2018;10:1758835918788310. doi:10.1177/1758835918788310.
  • Hakenberg OW. Nivolumab for the treatment of bladder cancer. Expert Opin Biol Ther. 2017;17:1309–1315. doi:10.1080/14712598.2017.1353076.
  • Hahn NM, Chang SS, Meng M, Shore ND, Konety BR, Steinberg GD, Gschwend JE, Nishiyama H, Redorta JP, Taylor JA, et al. A phase II, randomized study of nivolumab (nivo) or nivo plus BMS-986205 with or without intravesical Bacillus Calmette-Guerin (BCG) in BCG-unresponsive, high-risk, non-muscle invasive bladder cancer (NMIBC): checkMate 9UT. Journal of Clinical Oncology. 2019;37(7_suppl):TPS493-TPS493
  • Narayan V, Gladney W, Plesa G, Vapiwala N, Carpenter E, Maude SL, Lal P, Lacey SF, Melenhorst JJ, Sebro R, et al. A phase I clinical trial of PSMA-directed/TGFβ-insensitive CAR-T cells in metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37:7. doi:10.1200/JCO.2019.37.7_suppl.TPS347.
  • Smith M, García-Martínez E, Pitter MR, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: toll-like receptor agonists in cancer immunotherapy. Oncoimmunology. 2018;7:e1526250. doi:10.1080/2162402X.2018.1526250.
  • Vanpouille-Box C, Hoffmann JA, Galluzzi L. Pharmacological modulation of nucleic acid sensors - therapeutic potential and persisting obstacles. Nat Rev Drug Discov. 2019;18:845–867. doi:10.1038/s41573-019-0043-2.
  • Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science. 2007;316:1628–1632. doi:10.1126/science.1138963.
  • Uehori J, Matsumoto M, Tsuji S, Akazawa T, Takeuchi O, Akira S, Kawata T, Azuma I, Toyoshima K, Seya T, et al. Simultaneous blocking of human toll-like receptors 2 and 4 suppresses myeloid dendritic cell activation induced by mycobacterium bovis bacillus calmette-guerin peptidoglycan. Infect Immun. 2003;71:4238–4249. doi:10.1128/IAI.71.8.4238-4249.2003.
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology. 2012;1:699–716. doi:10.4161/onci.20696.
  • Ji N, Mukherjee N, Morales EE, Tomasini ME, Hurez V, Curiel TJ, Abate G, Hoft DF, Zhao X-R, Gelfond J, et al. Percutaneous BCG enhances innate effector antitumor cytotoxicity during treatment of bladder cancer: a translational clinical trial. Oncoimmunology. 2019;8(8):1614857. doi:10.1080/2162402X.2019.1614857.
  • Martinez R, Tapia G, De Muga S, Hernández A, Cao MG, Teixidó C, Urrea V, García E, Pedreño-López S, Ibarz L, et al. Combined assessment of peritumoral Th1/Th2 polarization and peripheral immunity as a new biomarker in the prediction of BCG response in patients with high-risk NMIBC. Oncoimmunology. 2019;8(8):1602460. doi:10.1080/2162402X.2019.1602460.
  • Rentsch CA, Bosshard P, Mayor G, Rieken M, Puschel H, Wirth G, Cathomas R, Parzmair GP, Grode L, Eisele B, et al. Results of the phase I open label clinical trial SAKK 06/14 assessing safety of intravesical instillation of VPM1002BC, a recombinant mycobacterium Bacillus Calmette Guerin (BCG), in patients with non-muscle invasive bladder cancer and previous failure of conventional BCG therapy. Oncoimmunology. 2020;9:1748981.
  • Maekawa H, Inoue T, Ouchi H, Jao T-M, Inoue R, Nishi H, Fujii R, Ishidate F, Tanaka T, Tanaka Y, et al. Mitochondrial damage causes inflammation via cGAS-STING signaling in acute kidney injury. Cell Rep. 2019;29(1261–1273):e1266. doi:10.1016/j.celrep.2019.09.050.
  • Grabosch S, Bulatovic M, Zeng F, Ma T, Zhang L, Ross M, Brozick J, Fang Y, Tseng G, Kim E, et al. Cisplatin-induced immune modulation in ovarian cancer mouse models with distinct inflammation profiles. Oncogene. 2019;38(13):2380–2393. doi:10.1038/s41388-018-0581-9.
  • von der Maase H, Sengelov L, Roberts JT, Ricci S, Dogliotti L, Oliver T, Moore MJ, Zimmermann A, Arning M. Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. 2005;23:4602–4608. doi:10.1200/JCO.2005.07.757.
  • Yuh BE, Ruel N, Wilson TG, Vogelzang N, Pal SK. Pooled analysis of clinical outcomes with neoadjuvant cisplatin and gemcitabine chemotherapy for muscle invasive bladder cancer. J Urol. 2013;189:1682–1686. doi:10.1016/j.juro.2012.10.120.
  • Obrist F, Michels J, Durand S, Chery A, Pol J, Levesque S, Joseph A, Astesana V, Pietrocola F, Wu GS, et al. Metabolic vulnerability of cisplatin-resistant cancers. Embo J. 2018;37. doi:10.15252/embj.201798597
  • Gravett AM, Trautwein N, Stevanovic S, Dalgleish AG, Copier J. Gemcitabine alters the proteasome composition and immunopeptidome of tumour cells. Oncoimmunology. 2018;7:e1438107. doi:10.1080/2162402X.2018.1438107.
  • Shimizu T, Tomogane M, Miyashita M, Ukimura O, Ashihara E. Low dose gemcitabine increases the cytotoxicity of human Vgamma9Vdelta2 T cells in bladder cancer cells in vitro and in an orthotopic xenograft model. Oncoimmunology. 2018;7:e1424671. doi:10.1080/2162402X.2018.1424671.
  • Sonpavde G, Necchi A, Gupta S, Steinberg GD, Gschwend JE, Van Der Heijden MS, Garzon N, Ibrahim M, Raybold B, Liaw D, et al. ENERGIZE: a Phase III study of neoadjuvant chemotherapy alone or with nivolumab with/without linrodostat mesylate for muscle-invasive bladder cancer. Future Oncol. 2020;16(2):4359–4368. doi:10.2217/fon-2019-0611.
  • Hellmann MD, Ciuleanu T-E, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, Minenza E, Linardou H, Burgers S, Salman P, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378:2093–2104. doi:10.1056/NEJMoa1801946.
  • Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–1290. doi:10.1056/NEJMoa1712126.
  • Hicks KC, Fantini M, Donahue RN, Schwab A, Knudson KM, Tritsch SR, Jochems C, Clavijo PE, Allen CT, Hodge JW, et al. Epigenetic priming of both tumor and NK cells augments antibody-dependent cellular cytotoxicity elicited by the anti-PD-L1 antibody avelumab against multiple carcinoma cell types. Oncoimmunology. 2018;7(11):e1466018. doi:10.1080/2162402X.2018.1466018.
  • Patel MR, Ellerton J, Infante JR, Agrawal M, Gordon M, Aljumaily R, Britten CD, Dirix L, Lee K-W, Taylor M, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018;19(1):51–64. doi:10.1016/S1470-2045(17)30900-2.
  • Tripathi A, Plimack ER. Immunotherapy for urothelial carcinoma: current evidence and future directions. Curr Urol Rep. 2018;19:109. doi:10.1007/s11934-018-0851-7.
  • Watanabe T, Gaedicke S, Guffart E, Firat E, Niedermann G. Adding indoximod to hypofractionated radiotherapy with anti-PD-1 checkpoint blockade enhances early NK and CD8(+) T-cell-dependent tumor activity. Clin Cancer Res. 2020;26:945–956. doi:10.1158/1078-0432.CCR-19-0476.
  • Zhai L, Spranger S, Binder DC, Gritsina G, Lauing KL, Giles FJ, Wainwright DA. Molecular pathways: targeting IDO1 and other tryptophan dioxygenases for cancer immunotherapy. Clin Cancer Res. 2015;21(24):5427–5433. doi:10.1158/1078-0432.CCR-15-0420.
  • Park J, Kim CG, Shim J-K, Kim JH, Lee H, Lee JE, Kim MH, Haam K, Jung I, Park S-H, et al. Effect of combined anti-PD-1 and temozolomide therapy in glioblastoma. Oncoimmunology. 2019;8(1):e1525243. doi:10.1080/2162402X.2018.1525243.
  • Pellegatta S, Eoli M, Cuccarini V, Anghileri E, Pollo B, Pessina S, Frigerio S, Servida M, Cuppini L, Antozzi C, et al. Survival gain in glioblastoma patients treated with dendritic cell immunotherapy is associated with increased NK but not CD8+T cell activation in the presence of adjuvant temozolomide. Oncoimmunology. 2018;7:e1412901. doi:10.1080/2162402X.2017.1412901.
  • Suryadevara CM, Desai R, Abel ML, Riccione KA, Batich KA, Shen SH, Chongsathidkiet P, Gedeon PC, Elsamadicy AA, Snyder DJ, et al. Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma. Oncoimmunology. 2018;7:e1434464. doi:10.1080/2162402X.2018.1434464.
  • Schiavoni G, Mattei F, Di Pucchio T, Santini SM, Bracci L, Belardelli F, Proietti E. Cyclophosphamide induces type I interferon and augments the number of CD44(hi) T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer. Blood. 2000;95:2024–2030. doi:10.1182/blood.V95.6.2024.
  • Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, Sanchez M, Lorenzi S, D’Urso MT, Belardelli F, et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 2011;71:768–778. doi:10.1158/0008-5472.CAN-10-2788.
  • Brown RA, Herzig RH, Wolff SN, Frei-Lahr D, Pineiro L, Bolwell BJ, Lowder JN, Harden EA, Hande KR, Herzig GP, et al. High-dose etoposide and cyclophosphamide without bone marrow transplantation for resistant hematologic malignancy. Blood. 1990;76:473–479. doi:10.1182/blood.V76.3.473.473.
  • Sirachainan N, Pakakasama S, Anurathapan U, Hansasuta A, Dhanachai M, Khongkhatithum C, Jinawath A, Mahachoklertwattana P, Hongeng S. Outcome of newly diagnosed high risk medulloblastoma treated with carboplatin, vincristine, cyclophosphamide and etoposide. J Clin Neurosci. 2018;56:139–142. doi:10.1016/j.jocn.2018.06.028.
  • Jakacki RI, Cohen KJ, Buxton A, Krailo MD, Burger PC, Rosenblum MK, Brat, DJ, Hamilton RL, Eckel SP, Zhou T, et al. Phase 2 study of concurrent radiotherapy and temozolomide followed by temozolomide and lomustine in the treatment of children with high-grade glioma: a report of the Children’s Oncology Group ACNS0423 study. Neuro Oncol 18, 1442–1450 (2016).
  • van den Bent MJ, Brandes AA, Taphoorn MJB, Kros JM, Kouwenhoven MCM, Delattre J-Y, Bernsen HJJA, Frenay M, Tijssen CC, Grisold W, et al. Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol. 2013;31(3):344–350. doi:10.1200/JCO.2012.43.2229.
  • Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, Brandes AA, Taal W, Domont J, Idbaih A, et al. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017;377(20):1954–1963. doi:10.1056/NEJMoa1707358.
  • Komiya T, Huang CH. Updates in the clinical development of epacadostat and other indoleamine 2,3-dioxygenase 1 inhibitors (IDO1) for human cancers. Front Oncol. 2018;8:423. doi:10.3389/fonc.2018.00423.
  • Xu X, Ren J, Ma Y, Liu H, Rong Q, Feng Y, Wang Y, Cheng Y, Ge R, Li Z, et al. Discovery of cyanopyridine scaffold as novel indoleamine-2,3-dioxygenase 1 (IDO1) inhibitors through virtual screening and preliminary hit optimisation. J Enzyme Inhib Med Chem. 2019;34(1):250–263. doi:10.1080/14756366.2018.1480614.
  • Liu K, Ren T, Huang Y, Sun K, Bao X, Wang S, Zheng B, Guo W. Apatinib promotes autophagy and apoptosis through VEGFR2/STAT3/BCL-2 signaling in osteosarcoma. Cell Death Dis. 2017;8(8):e3015. doi:10.1038/cddis.2017.422.
  • Scott LJ. Apatinib: a review in advanced gastric cancer and other advanced cancers. Drugs. 2018;78(7):747–758. doi:10.1007/s40265-018-0903-9.
  • Xu J, Zhang Y, Jia R, Yue C, Chang L, Liu R, Zhang G, Zhao C, Zhang Y, Chen C, et al. Anti-PD-1 antibody SHR-1210 combined with apatinib for advanced hepatocellular carcinoma, gastric, or esophagogastric junction cancer: an open-label, dose escalation and expansion study. Clin Cancer Res. 2019;25:515–523. doi:10.1158/1078-0432.CCR-18-2484.
  • Feng H, Cheng X, Kuang J, Chen L, Yuen S, Shi M, Liang J, Shen B, Jin Z, Yan J, et al. Apatinib-induced protective autophagy and apoptosis through the AKT-mTOR pathway in anaplastic thyroid cancer. Cell Death Dis. 2018;9:1030. doi:10.1038/s41419-018-1054-3.