5,348
Views
45
CrossRef citations to date
0
Altmetric
Review

Trial Watch—Small molecules targeting the immunological tumor microenvironment for cancer therapy

, , , , , , , , , , , & show all
Article: e1149674 | Received 29 Jan 2016, Accepted 29 Jan 2016, Published online: 23 May 2016

ABSTRACT

Progressing malignancies establish robust immunosuppressive networks that operate both systemically and locally. In particular, as tumors escape immunosurveillance, they recruit increasing amounts of myeloid and lymphoid cells that exert pronounced immunosuppressive effects. These cells not only prevent the natural recognition of growing neoplasms by the immune system, but also inhibit anticancer immune responses elicited by chemo-, radio- and immuno therapeutic interventions. Throughout the past decade, multiple strategies have been devised to counteract the accumulation or activation of tumor-infiltrating immunosuppressive cells for therapeutic purposes. Here, we review recent preclinical and clinical advances on the use of small molecules that target the immunological tumor microenvironment for cancer therapy. These agents include inhibitors of indoleamine 2,3-dioxigenase 1 (IDO1), prostaglandin E2, and specific cytokine receptors, as well as modulators of intratumoral purinergic signaling and arginine metabolism.

Introduction

Solid neoplastic lesions are not formed by cancer cells only, but contain elevated amounts of non-transformed cells, making up a highly interconnected system that encompasses malignant, endothelial, stromal and immunological components.Citation1-3 Thus, neoplastic lesions arise and evolve in the context of a physical and functional crosstalk with various populations of non-transformed cells, some of which de facto promote tumor progression and resistance to treatment.Citation4,5 Tumor-infiltrating myeloid and lymphoid cells play a particularly important role in this context.Citation3,6-11 Accumulating evidence indicates indeed that tumors can become clinically manifest only when the immunological mechanisms that are in place to recognize and eliminate potentially oncogenic cells (which are cumulatively referred to as natural anticancer immunosurveillance) fail.Citation12-15 Such a failure reflects the accumulation of genetic and/or epigenetic defects that ultimately renders (pre-)malignant cells undetectable by the immune system, or endows them with the capacity to block immune effector functions.Citation16-18 One of the main strategies whereby cancer cells maintain the immune system at bay consists in the establishment of robust immunosuppressive networks that operate both systemically (i.e., in the circulation and lymphoid organs) and locally (i.e., within the tumor microenvironment).Citation16,19-23

Extensive tumor infiltration by immunosuppressive cells (and/or limited infiltration by immune effector cells with antitumor functions) support tumor progression as it inhibits natural immunosurveillance.Citation20,21 Accordingly, high intratumoral levels of particularly immunosuppressive cells like CD4+CD25+FOXP3+ regulatory T cells (Tregs) and so-called M2 tumor-associated macrophages (TAMs) have been attributed negative prognostic value in patients affected by a wide panel of solid neoplasms.Citation3,7,24,25 Moreover, the accumulation of immunosuppressive cells, rather than immune effector cells, within the tumor microenvironment limits the clinical benefits provided not only by anticancer immunotherapy, but also by various radio- and chemo therapeutic regimens.Citation3,7,24-26 Indeed, the long-term efficacy of radio- and chemo-therapy appears to rely, at least in part, on the (re-)instatement of anticancer immunosurveillance.Citation27-32 In line with this notion, elevated intratumoral levels of Tregs, M2 TAMs and other immunosuppressive cells have been shown to predict dismal therapeutic outcome in multiple cohorts of individuals bearing solid malignancies.Citation3,7,24,27 Taken together, these observations provide a strong rationale to the development of clinically implementable strategies aimed at reverting immunosuppression within the tumor microenvironment, a wave of investigation that has been intensively pursued throughout the past decade, both preclinically and clinically.Citation33,34

As it stands, a number of approaches is available to counteract immunosuppressive systems established within the tumor microenvironment for therapeutic purposes.Citation35-38 Schematically, we can classify these strategies into four major (not mutually-exclusive) groups, based on their main mechanism of action: (1) direct immunostimulatory agents (i.e., interventions that mediate immunostimulatory effects by acting on immune effectors); (2) indirect immunostimulatory interventions (i.e., interventions that mediate immunostimulatory effects by operating on immunosuppressive cells); (3) enriching/depleting agents (i.e., interventions that exert immunostimulatory activity by influencing the recruitment of tumor-infiltrating immune cells); and (4) repolarizing interventions (i.e., interventions that exert immunostimulatory activity by converting immunosuppressive cells into immune effectors). Thus, enriching/depleting and repolarizing agents de facto alter the relative composition of the tumor infiltrate, whereas immunostimulatory interventions (at least initially) fail to do so, but only alter its activation state.

Prominent examples of direct immunostimulatory agents include Toll-like receptor (TLR) agonists, which potently activate dendritic cells (DCs) and other antigen-presenting cells (APCs) to prime tumor-targeting adaptive immune responses,Citation34,39,40 immunostimulatory cytokines such as granulocyte-macrophage colony stimulating factor (GM-CSF) and interleukin (IL)-15,Citation41 as well as a wide panel of monoclonal antibodies (mAbs) that operate as agonists of activatory receptors expressed on CD8+ cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells, like tumor necrosis factor receptor superfamily, member 9 (TNFRSF9, best known as CD137), TNFRSF4 (best known as OX40), or TNFRSF18 (best known as GITR).Citation42-45 This said, the molecules that are best known for their direct immunostimulatory effects are so-called “checkpoint blockers,” i.e., mAbs that antagonize immunosuppressive receptors expressed by activated CTLs like cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) or their ligands, like CD274 (best known as PD-L1) and programmed cell death 1 ligand 2 (PDCD1LG2, best known as PD-L2).Citation36,46,47 No less than three checkpoint blockers are nowadays approved by the US Food and Drug Administration (FDA) and other regulatory agencies worldwide for use in cancer patients, including one CTLA4-specific molecule (ipilimumab) and two PD-1-targeting agents (nivolumab and pembrolizumab).Citation46-48 Good examples of indirect immunostimulatory agents are provided by mAbs that neutralize potent immunosuppressive cytokines, including transforming growth factor β 1 (TGFB1) and vascular endothelial growth factor A (VEGFA).Citation44,49,50 Various chemotherapeutic agents, especially when used according to a metronomic schedule, have been shown to promote tumor infiltration by immune effector cells or to limit the recruitment of immunosuppressive cells to the tumor microenvironment.Citation27,30 For example, this applies to doxorubicin and oxaliplatin, which kill cancer cells in a way that promotes the recruitment of DCs and CTLs coupled to the elicitation of tumor-targeting immune responses,Citation51,52 as well as to cyclophosphamide and gemcitabine,Citation53 both of which limit tumor infiltration by Tregs.Citation54,55 Finally, some agents like IL-21 have been associated with the capacity to promote a functional shift of TAMs from a prominently immunosuppressive (M2) to a mainly immunostimulatory and tumoricidal (M1) state.Citation56

Of note, several of these molecules do not operate according to a single mechanism of action, but exert multipronged immunostimulatory effects. For example, cyclophosphamide not only depletes Tregs from the tumor environment, but also resembles doxorubicin and oxaliplatin in their ability to trigger so-called “immunogenic cell death” (ICD), hence promoting tumor infiltration by DCs and CTLs.Citation52,57-60 Along similar lines, VEGFA-targeting mAbs such as the FDA-approved agent bevacizumabCitation50 prevent VEGFA-dependent T-cell exhaustion and stimulate the normalization of the tumor vasculature, which favors tumor infiltration by immune effector cells.Citation49,61

We have discussed recent progress on tumor-targeting mAbs, immunostimulatory mAbs, immunostimulatory cytokines, TLR agonists, ICD inducers, and their ability to target the immunological tumor microenvironment in recent articles of the Trial Watch series. Here, we present preclinical and clinical advances on small molecules that may be harnessed to boost natural or therapy-elicited immune responses as they act on the tumor infiltrate.

IDO1 inhibitors

IDO1 catalyzes the initial, rate-limiting step of the so-called “kynurenine pathway,” the biochemical cascade that transforms the essential amino acid L-tryptophan (Trp) into L-kynurenine (Kyn).Citation62-64 Two additional enzymes can catalyze the same reaction in humans, namely, indoleamine 2,3-dioxigenase 2 (IDO2), and tryptophan 2,3-dioxigenase (TDO2).Citation62,63 Nonetheless, IDO1 plays a quantitatively superior role in this context and initiates the kynurenine pathway in a majority of settings.Citation62,63 Besides being constitutively expressed by some tissues, IDO1 can be synthesized in an inducible manner by specific subsets of DCs, macrophages and immature monocytes,Citation65-71 and it was originally viewed as an active component of the innate immune response to microbial stimuli.Citation72 More recently, however, it turned out that IDO1 has a key function in the establishment and maintenance of peripheral tolerance as it exerts major immunosuppressive effects.Citation62,63,73 The immunosuppressive activity of IDO1 has first been ascribed to its ability to limit the microenvironmental availability of Trp while favoring the accumulation of Kyn, 3-hydroxykynurenine and 3-hydroxyanthranilic acid.Citation72,74-76 However, several other, less-direct mechanisms may account for the robust immunosuppressive effects of IDO1-expressing cells.Citation66,77-86 Irrespective of the precise mechanisms whereby IDO1 mediates immunosuppressive effects, targeting IDO1 by pharmacological, genetic and immunological means has been associated with therapeutic effects in several distinct rodent models of carcinogenesis, and such effects were invariably accompanied by the (re)elicitation of tumor-targeting immune responses.Citation87 Today, several pharmacological inhibitors of IDO1 are available, some of which have also entered clinical evaluation.Citation88 Interestingly, the FDA-approved agent imatinib appears to limit IDO1 expression in the tumor microenvironment as a consequence of KIT inhibition.Citation89 However, imatinib is currently employed for its ability to directly inhibit oncogenic BCR-ABL1 fusion proteins, KIT and platelet-derived growth factor receptor, β polypeptide (PDGFRB), rather than for its immunostimulatory functions (and hence will not be discussed in further detail here).Citation90-92 TDO2 inhibitors are also being developed, although for the moment only in preclinical settings.Citation93,94

The competitive inhibition of IDO1 (and IDO2) with 1-methyltryptophan (a chiral mixture of 1-methyl-D-tryptophan and 1-methyl-L-tryptophan) as well as genetic interventions specific for IDO1 reportedly re(activate) therapeutically relevant immune responses in rodent tumor models,Citation22,87 especially when employed in combination with chemotherapy or checkpoint blockers.Citation62,95-99 Since in vivo 1-methyl-D-tryptophan (also known as indoximod or NLG8189) has a superior immunostimulatory activity as compared to its L-counterpart,Citation96,100,101 the former has already entered clinical development.Citation87 The first results from such a wave of investigation, however, have become available only recently.Citation102-105 In particular, indoximod was well tolerated in two Phase I trials, one involving 27 patients with metastatic solid tumors, who received indoximod in combination with docetaxel-based chemotherapy,Citation102 and one enrolling 12 adult subjects with recurrent refractory malignant brain tumors, who were treated with indoximod plus the alkylating agent temozolomide (NCT02052648).Citation104,106 Preliminary findings from a Phase I/II trial testing indoximod in support of DC-based vaccination targeting tumor protein 53 (TP53, best known as p53) epitopes in 44 subjects with metastatic solid tumors (NCT01042535) have also been released, although the study is still registered as “Active, not recruiting.”Citation105 Of 41 subjects who were evaluable, 37 (90.24%) experienced treatment-related side effects of any degree, while 14 (31.15%) were affected by Grade 3 or higher toxicities. However, 6 weeks after the completion of treatment, no study participants out of 30 who were evaluable in this sense exhibited objective responses (source www.clinicaltrials.gov). Official sources list nine additional studies aimed at assessing the immunostimulatory effects of indoximod in cancer patients. Five of these trials, including NCT01560923 (a Phase II study investigating the clinical profile of indoximod in prostate carcinoma patients receiving cellular immunotherapy with sipuleucel-T),Citation107 NCT01792050 (a Phase II trial assessing the safety and efficacy of indoximod plus docetaxel-based chemotherapy in women with metastatic breast carcinoma),Citation108 NCT02073123 (a Phase I/II study testing indoximod combined with ipilimumab in Stage III/IV melanoma patients),Citation109 NCT02077881 (a Phase I/II trial investigating the clinical profile of indoximod plus gemcitabine- and paclitaxel-based chemotherapy in subjects with metastatic pancreatic carcinoma), and NCT02502708 (a Phase I study assessing the safety and efficacy of indoximod plus temozolomide and radiation therapy in pediatric patients with primary brain tumors) are currently recruiting participants. Moreover, NCT02460367 (a Phase II trial testing indoximod combined with a cancer cell-based vaccine and docetaxel in subjects with advanced, previously treated non-small cell lung carcinoma, NSCLC) is listed as “Not yet recruiting.” NCT00739609 (a Phase I study investigating the maximum tolerated dose of indoximod in subjects with relapsed or refractory solid tumors has been terminated owing to lack of enrollment. To the best of our knowledge, the results of NCT00567931 and NCT01191216 (two Phase I trials testing indoximod as standalone immunotherapeutic intervention or combined with docetaxel in patients with unresectable advanced solid tumors or breast carcinoma, respectively)Citation103 have not yet been released, even though both studies are listed as “Completed” () (source www.clinicaltrials.gov).

Table 1. Clinical trials recently started to assess the safety and efficacy of small molecules targeting the tumor microenvironment for oncological applications*.

Similar to indoximod, epacadostat (also known INCB024360) and NLG919 (also known as GDC-0919) efficiently inhibit IDO1 (and less so IDO2), resulting in the restoration of therapeutically relevant anticancer immunity in rodent tumor models.Citation110-113 Clinical data from a Phase I trial testing the safety, tolerability, pharmacokinetics and pharmacodynamics of standalone epacadostat in subjects with advanced malignancies (NCT01195311) and from a Phase I/II study investigating the clinical profile of epacadostat plus ipilimumab in patients with unresectable or metastatic melanoma (NCT01604889) suggest that this IDO1 inhibitor is relatively well tolerated and exerts clinical activity, at least to some degree.114-116 Official sources list 12 additional studies aimed at testing the immunostimulatory effects of epacadostat in oncological indications. Eight of these trials, including NCT01961115 (a Phase II study testing epacadostat in support of a multipeptide vaccine in subjects with unresectable or advanced melanoma), NCT02042430 (a trial investigating the safety and efficacy of epacadostat as standalone immunotherapeutic intervention in women with reproductive tract tumors), NCT02118285 (a Phase I study assessing the clinical profile of epacadostat plus adoptively transferred NK cells in patients affected by reproductive tract neoplasms), NCT02166905 (a Phase I/II trial testing epacadostat in combination with a DC-targeted variant of NY-ESO-1 and a TLR3 agonist in women with malignancies of the reproductive system), NCT02178722 (a Phase I/II study investigating the safety and efficacy of epacadostat plus pembrolizumab in subjects with advanced solid tumors), NCT02298153 (a Phase I trial assessing the clinical activity of epacadostat in combination with an experimental checkpoint-blocking mAb in NSCLC patients), NCT02318277 (a Phase I/II study testing epacadostat plus an experimental checkpoint blocker in subjects with advanced solid tumors), and NCT02559492 (a Phase I trial investigating the safety and efficacy of epacadostat combined with a Janus kinase 1 (JAK1) inhibitor in individuals affected by advanced solid tumors) are currently recruiting participants, while NCT02575807 (a Phase II study assessing the therapeutic profile of epacadostat combined with a mesothelin-expressing bacterial vector in women with chemoresistant malignancies of the reproductive tract) is now listed as “Not yet recruiting.” NCT01982487 (a Phase I/II trial assessing the ability of epacadostat to boost the efficacy of a NY-ESO-1-targeting recombinant vaccine in women with cancers of the reproductive tract) has been withdrawn prior to enrollment for undisclosed reasons. To the best of our knowledge, the results of NCT01685255 (a Phase II study comparing the clinical activity of epacadostat to that of tamoxifen in women with biochemically-recurrent malignancies of the reproductive tract that achieved complete remission on first-line chemotherapy) and NCT01822691 (a Phase II study testing epacadostat as standalone immunotherapeutic agent in patients with myelodysplastic syndromes) have not been released yet, even though all these studies are listed as “Completed.” Finally, NCT02048709 (a Phase I trial assessing the safety and efficacy of NLG919 as standalone therapeutic intervention in subjects with advanced solid tumors) and NCT02471846 (a Phase I study investigating the clinical profile of NLG919 plus an experimental checkpoint-blocking antibody in individuals with locally advanced or metastatic solid tumors) are currently recruiting participants () (source www.clinicaltrials.gov).

Various other chemical inhibitors of IDO1 have been developed and tested for their immunostimulatory effects in preclinical settings, including methylthiohydantoin tryptophan,Citation95,117 brassinin and derivatives,Citation118,119 annulin B and derivatives,Citation120,121 exiguamine A and derivatives,Citation122,123 and INCB023843.Citation110 However, the clinical development of these molecules appear to stand at an impasse (source www.clinicaltrials.gov). Along similar lines, the TDO2-specific inhibitor LM10 has been shown to mediate therapeutically relevant immunostimulatory effects in mice with TDO2-expressing tumors.Citation124 However, the actual relevance of TDO2 for tumor-driven immunosuppression in patients has not yet been confirmed, and (at least for the moment) LM10 has not entered clinical development (source www.clinicaltrials.gov).

Modulators of purinergic tumor metabolism

Purinergic signaling plays a fundamental role in the elicitation of tumor-targeting immune responses.Citation52,58,125 Indeed, dying cancer cells actively secrete ATP (especially if they are undergoing ICD),Citation126,127 and acute elevations in extracellular ATP levels have a dual immunostimulatory effect. First, by activating purinergic receptor P2Y, G-protein coupled, 2 (P2RY2), they mediate robust chemotactic functions and recruit APCs and their precursors to site of cancer cell death.Citation128 Second, by acting on purinergic receptor P2X, ligand gated ion channel, 7 (P2RX7), they favor the activation of APCs and boost their ability to cross-prime an adaptive immune response specific for dead cell-associated antigens.Citation129 Moreover, extracellular ATP is normally degraded by the sequential activity of two enzymes, namely, ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1, best known as CD39) and 5′-nucleotidase, ecto (NT5E, best known as CD73).Citation130 In particular, CD39 converts ATP into ADP and AMP, whereas CD73 transforms AMP into adenosine.Citation130,131 Importantly, while extracellular ATP has immunostimulatory effects, extracellular adenosine (and less so AMP) exerts major immunosuppressive functions as it binds adenosine A2a receptor (ADORA2A) and adenosine A2b receptor (ADORA2B).Citation132 ADORA2A limits the ability of NK cells and CTLs to mediate interferon (IFN)γ-dependent cytotoxicity,Citation133,134 promotes CTL anergy,Citation135 and stimulates the immunosuppressive functions of Tregs and M2 TAMs.Citation133,136-138 ADORA2B promotes the recruitment and activation of a heterogeneous populations of relatively immature and potently immunosuppressive myeloid cells cumulatively referred to as myeloid-derived suppressor cells (MDSCs),Citation139,140 and stimulates DCs to express various immunosuppressive molecules including IL-6, IL-10, TGFB1 and IDO1.Citation141 Last, but not least, both ADORA2A and ADORA2B have direct effects on malignant cells, stimulating their survival and proliferation.Citation142 Of note, chronic elevations in extracellular ATP levels may also mediate immunosuppressive (rather than immunostimulatory) effects.Citation143 In particular, chronically high concentrations of extracellular ATP have been shown to promote the development of semi-mature, dysfunctional DCs by signaling via purinergic receptor P2Y, G-protein coupled, 11 (P2RY11),Citation144 and to stimulate the activity of MDSCs.Citation143 Taken together, these observations provide a robust rationale to the development of strategies that target the purinergic system within neoplastic lesions for the potentiation of chemo-, radio- or immuno therapy-driven anticancer immune responses.Citation52,58,145 Two major approaches are being pursued with this objective: (1) blocking the extracellular conversion of ATP into adenosine, and (2) modulating purinergic receptor signaling.

Inhibitors of extracellular ATP degradation

Multiple small molecules that limit the conversion of extracellular ATP into adenosine by inhibiting CD39 or CD73 are currently available for investigational purposes.Citation146 Molecules that inhibit CD39 (with variable degree of selectivity) include N6,N6-diethyl-D-β-γ-dibromomethylene-ATP (ARL-67176), 1-amino-2-sulfo-4-(2-naphthylamino)anthraquinone, suramin, polyoxotungstate (POM-1), reactive blue 2, pyridoxal phosphate-6-azophenyl-2′,4′-disulfonic acid (PPADS), as well as 8-thiobutyl-ATP and derivatives (8-thiobutyl-ADP and 8-thiobutyl-AMP).Citation147,148 CD73 inhibitors encompass α-β-methylene-ADP (AMPCP or APCP) and various derivatives (i.e., PSB-12441, PSB-12425, and PSB-12379).Citation149-151 The pharmacological inhibition of CD39 and/or CD73 has been shown to promote therapeutically relevant tumor-targeting immune responses in several rodent models of oncogenesis and tumor progression.Citation147,152-163 None of these agents, however, has yet entered clinical evaluation for its immunostimulatory activity (source www.clinicaltrials.gov). Indeed, while suramin has been tested as standalone therapeutic interventions or in combination with other treatments modalities in more than 15 clinical trials performed over the past 2 decades, all these studies tested suramin for its ability to operate as broad-spectrum antagonists of P2Y receptors expressed malignant cells and to neutralize some soluble growth factors including various members of the fibroblast growth factor (FGF) family.Citation164-168 Of note, the results of these trials suggest that non-cytotoxic doses of suramin are well-tolerated by cancer patients, but generally fail to improve the clinical activity of chemotherapy.Citation164-168

Modulators of purinergic receptors

Besides suramin and PPADS, both of which operate as broad-spectrum P2Y receptor antagonists, a few specific inhibitors of P2RY11, ADORA2A and ADORA2B are currently available.Citation37 So far, the P2RY11-selective antagonist NF340 has only been employed to confirm the implication of P2RY11 in IL-8 secretion and in the acquisition of a dysfunctional, immunosuppressive phenotype by DCs exposed to ischemia/reperfusion.Citation144,169 It will therefore be interesting to see whether NF340 can also limit the P2RY11-dependent accumulation of semi-mature DCs that may accompany some instances of tumor progression. Two experimental ADORA2A antagonists, namely ZM241365,Citation163 and SCH58261,Citation153,154,170,171 have been shown to exert therapeutically relevant immunostimulatory effects in tumor-bearing mice, but these molecules have not yet entered clinical evaluation (source www.clinicaltrials.gov). Two other small-molecule inhibitors of ADORA2A, namely, preladenant (also known as SCH420814) and istradefylline (also known as KW-6002), have been extensively tested in the clinic as therapeutic options for Parkinson disease,Citation172-174 but not as anticancer agents or immunostimulatory molecules (source www.clinicaltrials.gov). Although Phase III clinical data demonstrated that preladenant is well tolerated by patients with Parkinson disease, there was no therapeutic activity,Citation174,175 and the development of preladenant has been discontinued.Citation37 Conversely, istradefylline demonstrated a good safety profile and modest efficacy, and this was sufficient for obtaining regulatory approval in Japan.Citation176 It will be important to investigate whether preladenant or istradefylline may exert clinically relevant immunostimulatory effects in cancer patients receiving chemo-, radio- or immuno therapeutic regimens. The ADORA2B-selective antagonists PSB603 and PSB1115 have been shown to limit immunosuppression and neoangiogenesis mediated by Tregs and MDSCs in a murine melanoma model,Citation177-179 but have not yet entered clinical development (source www.clinicaltrials.gov). Others ADORA2B inhibitors like CVT-6883 and GS-6201 are being developed for the treatment of non-oncological, inflammatory conditions, like inflammatory bowel disease and chronic obstructive pulmonary disease.Citation180,181 Preliminary clinical data indicate that CVT-6883 is well tolerated.Citation182 To the best of our knowledge, however, whether CVT-6883 or GS-6201 can improve the efficacy of anticancer therapy has not yet been investigated.

PGE2 inhibitors

Especially under hypoxic conditions, several tumors overexpress prostaglandin-endoperoxide synthase 2 (PTGS2, best known as COX2),Citation183 which catalyzes the first and rate-limiting step of the biochemical cascade that converts arachidonic acid into prostaglandins, prostacyclin and thromboxane A2.Citation184 Sustained elevations in local prostaglandin E2 (PGE2) levels not only have a direct mitogenic activity on malignant cells,Citation185,186 but also mediate multipronged immunosuppressive effects by acting on tumor-infiltrating immune cells as well as on the tumor vasculature.Citation187-191 The principal responsible of the immunosuppressive activity of PGE2 are prostaglandin E receptor 2 (PTGER2, best known as EP2) and prostaglandin E receptor 4 (PTGER4, best known as EP4).Citation184 Indeed, EP2 and/or EP4 robustly inhibit the cytotoxic activity of NK cells and CTLs,Citation192-195 suppress the ability of M1 TAMs to secrete tumoricidal molecules like IFNγ and tumor necrosis factor (TNF),Citation193 activate Tregs,Citation196 promote the accumulation of MDSCsCitation194,197-199 and their capacity to secrete TGFB1,Citation194,200 limit inflammasome activation in macrophage,Citation201,202 and stimulate endothelial cells to express Fas ligand (FASLG), hence impeding tumor infiltration by immune effector cells.Citation188 Intriguingly, PGE2 is also overproduced in the course of apoptotic cell death,Citation203,204 owing to the caspase-3-dependent activation of phospholipase A2, group VI (PLA2G6).Citation185 This reinforces the notion that the precise cell death mechanisms activated by chemo- or radio therapy have a major impact on the consequent elicitation of a tumor-targeting immune response.Citation205 Irrespective of this aspect, two approaches have been pursued to limit the immunosuppressive activity of PGE2: (1) inhibiting PGE2 synthesis, and (2) antagonizing EP2 and EP4.

Inhibitors of PGE2 synthesis

A wide panel of COX2 inhibitor (with various degree of selectivity) is currently approved by the US FDA and equivalent agencies worldwide for use as non-steroidal anti-inflammatory agents, including (but not limited to) acetylsalicylic acid (best known as aspirin) and celecoxib (source www.fda.gov/Drugs/default.htm). Aspirin is a relatively non-selective COX1 and COX2 inhibitor commonly available over the counter for the treatment of pain, fever and inflammation.Citation184 Low-dose aspirin is often prescribed as maintenance therapy to patients surviving myocardial infarction and other ischemic events, owing to its capacity to inhibit platelet aggregation.Citation206 Prolonged aspirin intake has also been associated with a reduced incidence of colorectal cancer, an on-target effect presumably linked to the oncogenic effects of chronic inflammation in large intestine as well as to phosphoinositide-3-kinase (PI3K) signaling (which is directly connected to EP2 and EP4).Citation207,208 Celecoxib is a COX2-specific inhibitor, available on prescription for the treatment of various inflammatory disorders associated with pain including osteoarthritis, rheumatoid arthritis and ankylosing spondylitis.Citation209 Aspirin as well as celecoxib have been shown to favor the (re-)establishment of an immunostimulatory tumor microenvironment,Citation188,210-218 hence limiting natural tumor progression or improving the therapeutic effects of various treatments. Taken together with the well-established safety profile of aspirin and celecoxib, these findings prompted an intense wave of investigation aimed at elucidating the ability of COX2 inhibitors to provide clinical benefits to cancer patients. Official sources list indeed 95 and 202 studies aimed at assessing the therapeutic profile of aspirin and celecoxib, respectively, in cancer patients. Only a minority of these trials, however, specifically focused/focuses on the immunostimulatory potential of aspirin (three studies) and celecoxib (21 studies) (source www.clinicaltrials.gov). In particular, the ability of aspirin to mediate clinically relevant immunostimulatory effects has been/is being assessed in (1) breast carcinoma patients, who are treated with aspirin plus neoadjuvant metronomic cyclophosphamide and methotrexate (NCT01612247: Phase I, status “Unknown”);Citation219,220 (2) women with ovarian carcinoma, who received aspirin in the context of a multimodal immunotherapeutic regimen (NCT01312376: Phase I, status “Completed”); and (3) subjects with reproductive tract neoplasms, receiving aspirin plus bevacizumab and metronomic cyclophosphamide in support of DC-based vaccination (NCT01132014: Phase 0, status “Recruiting”). To the best of our knowledge, the results of NCT01312376 have not been released yet () (source www.clinicaltrials.gov). Moreover, the immunostimulatory potential of celecoxib has been/is being investigated in (1) colorectal cancer patients, receiving celecoxib either as standalone pre-operative intervention (NCT01284504: Phase n.a., status “Withdrawn;” NCT00473980: Phase IV, status “Completed”), or in the context of a chemokine-modulatory regimen involving IFN-α2b and an experimental TLR agonist given alone (NCT01545141: Phase I/II, status “Recruiting”) or in support to DC-based vaccination (NCT02615574: Phase II, status “not yet recruiting); (2) subjects with NSCLC, who were treated with celecoxib as standalone immunotherapeutic intervention (NCT00104767: Phase I, status “Completed;” NCT00108186: Phase I, status “Withdrawn”) or in support of DC-vaccination (NCT00442754: Phase I, status “Completed”); (3) patients with oral or nasopharyngeal neoplasms, who received celecoxib alone (NCT00953849: Phase I/II, Status “Recruiting”), combined with 1,25-dihydroxyvitamin D3 (NCT00953849: Phase I/II, Status “Recruiting”), or as an adjuvant to a genetically modified DC-based vaccine (NCT00589186: Phase II, Status “Unknown”); (4) individuals with hepatocellular carcinoma, receiving celecoxib in support of a sequential strategy of virus-based anticancer vaccination (NCT00081848: Phase I, status “Completed”);Citation221,222 (5) melanoma patients, who received celecoxib plus metronomic cyclophosphamide and IL-2 as adjuvant to autologous DC-based vaccination (NCT00197912: Phase I/II, status “Completed”);Citation223 (6) children and adolescents with recurrent or progressive high-risk neuroblastoma, who are treated with celecoxib in the context of multimodal metronomic chemotherapy (NCT02641314: Phase II, status “Recruiting”); and (7) women with ovarian carcinoma, receiving celecoxib in the context of intensive locoregional chemoimmunotherapy plus intranodal DC-based vaccination (NCT02432378: Phase I/II, status “Recruiting”) () (source www.clinicaltrials.gov). In addition, the immunostimulatory activity of celecoxib has been/is being evaluated in several cohorts of patients with locally advanced or metastatic solid neoplasms, receiving celecoxib (1) together with metronomic cyclophosphamide and/or the particulate adjuvant commonly known as ISCOMATRIX™ in support of vaccination with autologous or heterologous cancer cell lysates (NCT01143545: Phase I, status “Completed;” NCT01258868: Phase I, status “Suspended;” NCT01313429: Phase I, status “Completed;” NCT01341496: Phase I, status “Suspended;” NCT02054104: Phase I/II, status “Suspended”); (2) in the context of a chemokine-modulatory regimen involving IFN-α2b and an experimental TLR agonist in support to DC-based vaccination (NCT02151448: Phase I/II, status “Recruiting”); and (3) combined with the epigenetic modifier decitabine (NCT00037817: Phase I, status “Completed”) () (source www.clinicaltrials.gov). Both NCT00108186 and NCT01284504 were withdrawn owing to low accrual rate. NCT01258868, NCT01341496 and NCT02054104 suspended enrollment for undisclosed reasons. Preliminary findings from NCT00081848 suggest that celecoxib does not cause unexpected side effects in cancer patients.Citation221,222 Moreover, the results of NCT00197912 suggest that celecoxib plus metronomic cyclophosphamide may considerable improve the efficacy of DC-based vaccination, although no decrease in tumor-infiltrating Tregs was detected in this setting.Citation223 To the best of our knowledge, the findings of NCT00037817, NCT00104767, NCT00442754, NCT00473980, NCT00953849, NCT01143545, and NCT01313429 have not yet been disclosed (source www.clinicaltrials.gov).

Pge2 receptor antagonists

Various small-molecule antagonists of EP2 and EP4 are currently available, including the EP2-specific inhibitor PF-04418948,Citation224,225 and the EP4-specific agents RQ-15986,Citation200,217 AH 23848,Citation198,226 and BGC201531.Citation227 The safety of PF-04418948 has been tested in a single clinical trial enrolling healthy volunteers (NCT01002963), but the development of PF-04418948 has subsequently been discontinued.Citation37 In murine models of breast carcinoma, RQ-15986 has been shown to protect NK cells from PGE2-dependent immunosuppression,Citation200 and to inhibit tumor-infiltration by immunosuppressive macrophages,Citation217 resulting in limited tumor growth and reduced metastatic dissemination. AH 23848 has been demonstrated to resemble RQ-15986 in its capacity to protect NK cells from immunosuppression,Citation226 and to inhibit the ability of MDSCs to infiltrate neoplastic lesions upon the downregulation of chemokine (C–X–C motif) receptor 4 (CXCR4).Citation198 To the best of our knowledge, neither RQ-15986 nor AH 23848 has been tested in the clinic so far (source www.clinicaltrials.gov). Finally, BGC20-1531 has never been investigated for its immunostimulatory activity, but preliminary clinical data indicate that it is well tolerated by healthy subjects.Citation227 Preclinical experiments testing whether BGC20-1531 mediates therapeutically relevant immunostimulatory activity in rodent tumor models are urgently awaited.

Cytokine receptor inhibitors

The establishment of an immunostimulatory versus immunosuppressive immune infiltrate is determined by several factors, among which cytokine signaling has a prominent role.Citation228-231 Indeed, cytokines and chemokines not only guide the recruitment of immunostimulatory versus immunosuppressive cells to the tumor microenvironment, but also influence the local acquisition of immunostimulatory versus immunosuppressive functions.Citation228-231 One strategy to suppress immunosuppressive cytokine signaling consists in the administration of neutralizing mAbs, like the TGFB1-targeting molecule fresolimumab,Citation232-234 or mAbs that operate as receptor antagonists, like the CXCR4-specific molecule BMS-936564.Citation235 Cytokine receptors, however, are common G protein-coupled receptors and hence can be also be targeted with small molecules that operate as antagonists.Citation145,236,237

CXCR4 inhibitors

CXCR4 is main receptor for chemokine (C–X–C motif) ligand 12 (CXCL12), and the CXCL12–CXCR4 signaling axis appears to play a central role in the recruitment of immunosuppressive cells to the tumor microenvironment.Citation238,239 Several CXCR4 antagonists have been developed, some of which have already entered clinical evaluation.Citation240,241 Of note, part of this wave of investigation has been driven by the fact that CXCR4 also operates as co-receptor for the entry of T-tropic HIV-1 strains into CD4+ cells.Citation242-244 Moreover, various CXCR4 antagonists including plerixafor (also known as AMD3100 or JM3100 and commercialized under the trade name of Mozobil™), burixafor (also known as TG0054), LY2510924, and POL6326 and BKT140 (also known as BL-8040) have been developed (and are currently being tested in clinical settings) because of their ability to rapidly mobilize CD34+ cells.Citation245 This has two major clinical applications: (1) the mobilization of healthy haematopoietic stem cells for collection and subsequent autologous or heterologous transplantation,Citation246,247 and (2) the disruption of beneficial interactions between haematopoietic cancer cells and their microenvironment, resulting in chemosensitization.Citation248,249 Nonetheless, official sources list no less than seven studies testing CXCR4 antagonists for their anticancer (as opposed as CD34+ cell-mobilizing) effects. NCT01339039 (a Phase I trial assessing the clinical profile of plerixaforCitation250 plus bevacizumab in individuals with recurrent high-grade glioma), NCT01977677 (a Phase I/II study testing plerixafor plus temozolomide and radiation therapy in patients with newly diagnosed high-grade glioma), and NCT02179970 (a Phase I trial investigating the safety of continuous intravenous plerixafor in subjects with advanced pancreatic, ovarian and colorectal tumors) are currently recruiting participants. NCT01391130 (a Phase II study assessing the therapeutic profile of LY2510924Citation251 combined with the multi-target tyrosine kinase inhibitor sunitinib in patients with metastatic renal cell carcinoma) and NCT01439568 (a Phase II trial investigating the safety and activity of LY2510924 plus carboplatin and etoposide in subjects with extensive stage small cell lung carcinoma) are currently listed as “Active, not recruiting.” Preliminary results from NCT01391130 indicate that the addition of LY2510924 to sunitinib-based chemotherapy is safe, but does not improve efficacy,Citation252 while early findings from NCT01439568 suggest that the clinical activity of LY2510924 may not involve immunological effects in this setting.Citation253,254 NCT00591682 (a Phase I study testing oral MSX122Citation255,256 in individuals with metastatic or locally advanced solid tumors) has suspended the recruitment of participants for undisclosed reasons. Finally, NCT01837095 (a Phase I trial evaluating the clinical profile of POL6326Citation241 plus eribulin in women with metastatic breast carcinoma) is currently recruiting participants () (source www.clinicaltrials.gov).

Additional CXCR4 antagonists including CTCE9908, and POL5551 have been shown to limit neoplastic growth and/or metastatic dissemination in mice, in some instances coupled to reduced tumor infiltration by MDSCs or immunosuppressive TAMs.Citation257-263 To the best of our knowledge, however, none of these agents has entered clinical development yet (source www.clinicaltrials.gov).

CCR2 inhibitors

Similar to CXCR4 and CXCL12, chemokine (C–C motif) receptor 2 (CCR2) and its cognate ligand chemokine (C–C motif) ligand 2 (CCL2) have been implicated in tumor infiltration by immunosuppressive cells, notably M2 TAMs and MDSCs.Citation264-269 However, CCR2 has also been involved in the recruitment of CD4+ TH1 lymphocytes and DC precursors to the tumor bed, underlying the local activation of anticancer immunity.Citation270-273 Thus, even though targeting the CCL2-CCR2 axis may have context-dependent effects, several CCR2 inhibitors are currently being evaluated for their immunostimulatory activity, mostly in preclinical settings.Citation274 The majority of these molecules have indeed been tested clinically for the therapy of non-oncological conditions, but their development has come to an impasse.Citation274 This applies to AZD2423, which has been studied for the treatment of chronic obstructive pulmonary disease and neurotic pain;Citation275 BMS741672, which has been investigated for safety and efficacy in patients with diabetes or neurotic pain; BMS813160 and CCX140, which have been assessed as therapeutic measures in subjects experiencing diabetic kidney disease;Citation276-278 JNJ17166864, which has been tested as a treatment for allergic rhinitis; MK0812, which has been investigated as a therapy for multiple sclerosis, rheumatoid arthritis and Alzheimer disease;Citation279 as well as PF04634817, which has been intensively tested in subjects with diabetic kidney disease (source www.clinicaltrials.gov). Cenicriviroc, a mixed CCR2/CCR5 antagonist, is still being assessed for its activity against HIV-1 infection, primary sclerosing cholangitis, and hepatic disorders, but not as an immunostimulatory agent in cancer patients (source www.clinicaltrials.gov). Conversely, the safety and efficacy of PF4136309 (which mediated promising therapeutic activity in preclinical settings)Citation265 are being evaluated in subjects with borderline resectable and locally advanced pancreatic adenocarcinoma under the FOLFIRINOX chemotherapeutic regimen (folinic acid plus 5-fluorouracil plus irinotecan plus oxaliplatin), in the context of a Phase I clinical trial that is listed by official sources as “Active, not recruiting” (NCT01413022) () (source www.clinicaltrials.gov).

CCR5 inhibitors

Chemokine (C–C motif) receptor 5 (CCR5) is expressed by a plethora of immune cells, including CTLs, CD4+ TH1 cells, monocytes, macrophages, MDSCs and Tregs, and is well known for its ability to operate a co-receptor for the entry of M-tropic HIV-1 strains into CD4+ cells.Citation280,281 In addition, CCR5 has been implicated in the recruitment of Tregs and MDSCs to the tumor microenvironment.Citation282,283 A bunch of CCR5 antagonists have been developed so far, mostly for antiretroviral applications.Citation37,284 Some of these molecules, however, are also being evaluated as immunostimulatory agents for oncological indications, mostly in preclinical settings. In mice, maraviroc has been shown to limit diet-induced hepatocellular tumorigenesis,Citation285 to interrupt the metastatic dissemination of basal breast cancer cells,Citation286 and to impede the recruitment of immunosuppressive TAMs to triple-negative breast carcinomas.Citation90,287,288 An intense wave of investigation, which has not yet come to an and, evaluated the safety, tolerability and pharmacodynamics of maraviroc in healthy subjects, as well as its therapeutic activity in subjects affected by HIV-1 and associated complications (e.g., Kaposi sarcoma, dementia).Citation289,290 More recently, maraviroc has been tested for its capacity to limit graft-versus-host disease in transplanted patients,Citation291,292 and as a single immunotherapeutic intervention in subjects affected by advanced colorectal cancer patients with hepatic metastases (NCT01736813). To the best of our knowledge, the results of this Phase I trial (MARACON-001) have not yet been released, even though the study is listed as “Completed” by official sources () (source www.clinicaltrials.gov). The safety and efficacy of additional CCR5 antagonists including INCB009471,Citation293 aplaviroc and vicriviroc have been assessed in multiple clinical studied enrolling HIV-1+ patients or subjects at increased risk for HIV-1 infection, but not in oncological indications (source www.clinicaltrials.gov). Similarly, the development of CCR5 antagonists of the spiropiperidine family appears to stand at an impasse.Citation294,295

CSF1R inhibitors

Colony stimulating factor 1 receptor (CSF1R) signaling favors tumor infiltration by TAMs and their polarization toward an M2 (rather than an M1) phenotype.Citation296-299 A few chemical inhibitors of CSF1R are currently being assessed for their immunostimulatory activity, both in preclinical and clinical settings. PLX3397, a CSF1R inhibitor that also target the oncogenic receptors KIT and fms-related tyrosine kinase 3 (FLT3), reportedly mediates therapeutically relevant immunostimulatory effects in various rodent models of tumorigenesis, both as standalone immunotherapeutic intervention and combined with other forms of treatment.Citation300-304 Importantly, PLX3397 has already entered clinical development, and official sources list no less than 17 studies aimed at investigating the safety and efficacy of this agent in cancer patients (source www.clinicaltrials.gov). Three of these trials (NCT01004861: Phase I, status “Active, not recruiting;” NCT02371369: Phase III, status “Recruiting;” NCT02584647: Phase I/II, status “Recruiting”) are assessing the ability of PLX3397 to inhibit the growth of synovial tumors, which are known to overexpress CSF1R. Preliminary results from the Phase II extension of NCT01004861 indicate that PLX3397 is well tolerated and promote objective responses or disease stabilization in majority of patients.Citation305 Moreover, five studies have investigated/are investigating the ability of PLX3397 to exert direct antineoplastic effects in patients with KIT- or FMS3-expressing neoplasms, including gastrointestinal stromal tumors (NCT02401815: Phase I/II, status “Suspended”), various forms of leukemia (NCT01349049: Phase I/II, status “Active, not recruiting;” NCT02390752: Phase I/II, status “Recruiting”), Hodgkin lymphoma (NCT01217229: Phase II, status “Completed”), and acral and mucosal melanoma (NCT02071940: Phase II, status “Recruiting) (source www.clinicaltrials.gov). The remaining nine studies have evaluated/are evaluating the capacity of PLX3397 to mediate therapeutic effects as standalone agent, or to improve the efficacy of other treatments in patients with neoplasms that do not express CSF1R, KIT or FMS3, or do not rely on these receptors for growth (source www.clinicaltrials.gov). NCT01826448 (a Phase I trial testing PLX3397 in combination with a BRAFV600E inhibitor in individuals with metastatic or unresectable melanoma) has been terminated for undisclosed reasons. Results from NCT01349036 (a Phase I study investigating the safety of oral PLX3397 in subjects with recurrent glioblastoma) suggest that this immunotherapeutic regimen is well tolerated but has no clinical efficiency.Citation306,307 Although it is listed as “Completed,” the results of NCT01499043 (a Phase II study assessing the clinical profile of single-agent PLX3397 in patients with advanced castration-resistant prostate cancer) are not publicly available. NCT01790503 (a Phase I/II trial testing PLX3397 plus temozolomide and radiation therapy in individuals with newly diagnosed glioblastoma) is currently listed as “Active, not recruiting.” Moreover, NCT01042379 (a Phase II study evaluating the safety and efficacy of PLX3397 combined with paclitaxel in breast carcinoma patients), NCT01525602 (a Phase I trial investigating the therapeutic profile of PLX3397 plus paclitaxel in subjects with advanced solid tumors),Citation308 NCT01596751 (a Phase I/II study testing PLX3397 in combination with eribulin in women with metastatic breast carcinoma), NCT02452424 (a Phase I/II trial assessing the safety and efficacy of PLX3397 plus pembrolizumab in individuals with advances solid neoplasms), and NCT02472275 (a Phase I study investigating the clinical profile of PLX3397 plus radiotherapy and androgen ablation therapy in patients with intermediate- or high-risk prostate cancer) are currently recruiting participants () (source www.clinicaltrials.gov).

The administration of BLZ945 as a standalone immunotherapeutic agent has been shown to limit the turnover of TAMs in murine models of transgene-driven cervical cancer, breast carcinoma and glioma, resulting in increased infiltration by CTLs, tumor growth inhibition, and improved survival.Citation296,309 GW2580 has been reported to limit tumor-infiltration by M2 TAMs and/or MDSCs, resulting in the inhibition of neoplastic progression in various rodent models of tumorigenesis.Citation310-315 Along similar lines, GW2580 remarkably increased the efficacy of radiotherapy in mice bearing prostate carcinomas,Citation316 gemcitabine in a transgenic mouse model of pancreatic adenocarcinoma,Citation317 and an mAb targeting the VEGFA receptor in mice bearing subcutaneous melanomas, prostate carcinomas or lung carcinomas.Citation318 It will be interesting to see whether these CSF1R inhibitors are well tolerated and exert clinical activity in cancer patients.

Kinase inhibitors

Some intracellular signal transduction pathways have been implicated in the establishment of an immunosuppressive tumor microenvironment, and hence may constitute promising targets for the development of immunostimulatory interventions based on small molecules.

ALK5 inhibitors

TGFB1-driven immunosuppression relies on the engagement of transforming growth factor, β receptor 1 (TGFBR1, also known as ALK5), resulting in the activating phosphorylation of the transcriptional regulators SMAD family member 2 (SMAD2) and SMAD family member 3 (SMAD3).Citation231,319-321 First-generation ALK5 inhibitors (e.g., AZ12601011, AZ12799734) were associated with prominent cardiotoxicity,Citation322 and their development was discontinued.Citation37 Nowadays, a few second-generation ALK5 inhibitors are being evaluated in preclinical and clinical settings. Galunisertib (also known as LY2157299) is well known to exert therapeutically relevant immunostimulatory effects in several models of tumorigenesis in mice.Citation320,323-325 These findings prompted an intense wave of investigation aimed at assessing the safety and pharmacodynamics of galunisertib in healthy individuals and its therapeutic profile in patients affected by advanced solid tumors, including glioma.Citation326 Preliminary findings from these Phase I studies, which include NCT01722825 (testing galunisertib monotherapy in Japanese patients with advanced or metastatic neoplasms) and NCT01682187 (investigating the clinical profile of galunisertib alone or combined with lomustine in glioma patients), indicate that the administration of galunisertib as standalone immunotherapeutic intervention is well tolerated, does not cause medically relevant cardiac toxicity, and exerts antineoplastic effects (at least in a proportion of glioma patients).Citation327-331 Official sources list 15 additional studies aimed at investigating the clinical profile of galunisertib (source www.clinicaltrials.gov). To the best of our knowledge the results of NCT01746004 and NCT01965808 (two Phase I trials assessing the safety and pharmacodynamics of galunisertib in healthy volunteers) have not been released yet, even though both studies appear as “Completed.” NCT01220271 (a Phase I/II study testing galunisertib plus temozolomide and radiation therapy in subjects with newly diagnosed glioma),Citation332 NCT01373164 (a Phase I/II trial assessing the clinical profile of galunisertib combined with gemcitabine in individuals with locally advanced or metastatic pancreatic cancer),Citation333 NCT01582269 (a Phase II study evaluating the safety and efficacy of galunisertib plus lomustine in patients with recurrent glioblastoma),Citation334,335 and NCT02008318 (a Phase II/III trial investigating the therapeutic activity of galunisertib in subjects affected by myelodysplastic syndromes) are currently listed as “Active, not recruiting,” while NCT02452008 (a Phase II study testing galunisertib plus androgen-receptor inhibition in patients with metastatic, castration-resistant prostate carcinoma) appears as “Not yet recruiting.” Finally, NCT01246986, NCT02178358, NCT02240433 (three Phase I/II trials assessing the therapeutic profile of galunisertib combined with the multi-target tyrosine kinase inhibitor sorafenib in subjects with hepatocellular carcinoma),Citation336 NCT02154646 (a Phase I study evaluating the safety and efficacy of galunisertib plus gemcitabine in individuals with advanced or metastatic pancreatic cancer), NCT02304419 (a Phase I trial specifically investigating the immunological effects of galunisertib in cancer patients), NCT02423343 (a Phase I/II trial testing galunisertib plus nivolumab in subjects with advanced chemorefractory solid tumors) and NCT02538471 (a Phase II study assessing the therapeutic activity of galunisertib combined with radiation therapy in women with metastatic breast carcinoma) are currently recruiting participants (source www.clinicaltrials.gov). The safety an pharmacodynamics of another ALK5 inhibitor, i.e., TEW-7197,Citation37 are also being tested in subjects with advanced stage solid tumors (NCT02160106: Phase I, status “Recruiting”) (source www.clinicaltrials.gov). However, we were unable to find additional information on the activity of this compound in preclinical tumor models.

RON inhibitors

Besides being directly involved in some instances of tumorigenesis, macrophage stimulating 1 receptor (MST1R, best known as RON) has recently been shown to favor the polarization of TAMs toward the M2 phenotype.Citation337,338 However, the clinical development of a relatively non-selective RON inhibitor, i.e., BMS-777607 (previously known as ASLAN002), appears to stand at an impasse. Indeed, official sources list only two trials evaluating the safety and efficacy of BMS-777607 in cancer patients. NCT01721148 (a Phase I trial testing BMS-777607 monotherapy in subjects with advanced or metastatic solid neoplasm) appears as “Active, not recruiting,” while NCT00605618 (a Phase I/II study investigating the clinical profile of BMS-777607 as standalone therapeutic agent in a similar patient cohort) is listed as “Completed,” yet results are unavailable () (source www.clinicaltrials.gov). It will be interesting to see whether the development of immunostimulatory regimens based on known or novel RON inhibitors will be pursued or not.

PI3K inhibitors

PI3K signaling is hyperactivated in several tumors, promoting the survival and proliferation of malignant cells.Citation339-341 This led to the development of various PI3K inhibitors for cancer therapy, including the FDA-approved PI3Kδ-selective agent idelalisib (also known as GS-1101 or CAL-101) and PI-3065.Citation342-344 Recent findings indicate that PI3Kδ inhibitors may also exert robust immunostimulatory effects by inhibiting Tregs and MDSCs.Citation344,345 Along similar lines, PI3Kγ inhibitors like TG100-115 and AS605240 have been shown to mediate anticancer effects by virtue of their capacity to target tumor-supporting myeloid cells.Citation346 Currently, no clinical trials are assessing the possibility that PI3K inhibitors may exert therapeutically relevant immunostimulatory effects in cancer patients (source www.clinicaltrials.gov). Conversely, idelalisib is being extensively assessed for its ability to directly target PI3Kδ-dependent malignancies, including various forms of leukemia, follicular lymphoma, and small lymphocytic lymphoma (SLL).Citation342,343,347-352

Modulators of arginine metabolism

Normal immunological functions rely on physiological levels of L-arginine in the tumor microenvironment.Citation353 Indeed, L-arginine withdrawal results in the depletion of the CD3ζ chain of the T-cell receptor, and the consequent suppression of T-cell responses to antigenic stimulation.Citation354 L-arginine can be hydrolyzed to ornithine and urea by arginase 1 (ARG1), which is highly expressed (and secreted) by hepatocytes, MDSCs and TAMs, or ARG2, a mitochondrial isoform of ARG1 that is expressed by a wide panel of cells.Citation355 Alternatively, L-arginine can be employed by nitric oxide synthase 2, inducible (NOS2, best known as iNOS) to generate nitric oxide (NO).Citation356 NO also exerts immunosuppressive effects, mostly because it favors the S-nitrosylation of key cysteine residues in proteins required for normal immunological functions and because it affects the enzymatic activity of guanylyl cyclases and cyclic GMP-dependent kinases.Citation357,358 Interestingly, MDSCs and TAMs are among the few cell types that co-express ARG1 and iNOS.Citation37 In this setting, iNOS switches to the production of superoxide, hence initiating the generation of immunosuppressive reactive oxygen and nitrogen species.Citation37,359 Various inhibitors of ARG1 and iNOS have been developed for investigational purposes, including the arginine analog N-hydroxy-nor-L-arginine (an ARG1/ARG2 inhibitor),Citation360 NCX-4016 and AT38 (two nitroaspirins that inhibit ARG1, ARG2 and iNOS),Citation358,361,362 and compound 9 (another ARG1/ARG2 inhibitor),Citation363 but none of these molecules is currently under clinical evaluation in cancer patients (source www.clinicaltrials.gov). In addition, it seems that the phosphodiesterase 5A, cGMP-specific (PDE5A) inhibitor tadalafil (which is approved by the US FDA and commercialized as Cialis® for the treatment of erectile dysfunction) mediates immunostimulatory effects as it reduces ARG1 and iNOS expression in MDSCs.Citation364 Recent clinical data from NCT00843635 (Phase II, status “Completed”) and NCT00894413 (Phase II, status “Completed”) demonstrate that adjuvant tadalafil is well tolerated by patients with resected head and neck squamous cell carcinoma (HNSCC) and efficiently lowers the amounts of circulating MDSCs and tumor-infiltrating Tregs, resulting in the recovery of antitumor T-cell functions.Citation365,366 Official sources list six additional trials aimed at investigating the ability of tadalafil to exert therapeutically relevant immunostimulatory activity in cancer patients (source www.clinicaltrials.gov). NCT01374217 (a Phase I study assessing the therapeutic profile of tadalafil plus lenalidomideCitation367 and dexamethasone in subjects with multiple myeloma) has been terminated according to an early stopping rule. The status of NCT01342224 (a Phase I trial evaluating the immunostimulatory activity of tadalafil in pancreatic cancer patients receiving a peptide-based vaccine and radiation therapy) is currently “Unknown.” NCT01903083 (a Phase I study testing neoadjuvant tadalafil plus gemcitabine and radiation therapy in subjects with borderline resectable and locally advanced pancreatic adenocarcinoma) is listed as “Active, not recruiting”, while NCT02544880 (a Phase I/II trial assessing the immunostimulatory activity of tadalafil in subjects with HNSCC receiving a peptide-based vaccine) appears as “Not yet recruiting.” Finally, NCT01697800 (a Phase II study evaluating the clinical profile of tadalafil monotherapy in subjects with HNSCC of the upper aerodigestive tract) and NCT01858558 (a Phase II trial investigating tadalafil plus lenalidomide as maintenance therapy in multiple myeloma patients) are currently recruiting participants (source www.clinicaltrials.gov).

Concluding remarks

Small molecules offer several advantages over other immunotherapeutic regimens (including tumor-targeting and immunomodulatory mAbs, adoptive cell transfer, DNA-, peptide- and DC-based vaccination, and recombinant cytokines), such as an improved stability (ex vivo as well as in patients), limited manufacturing problems (implying reduced production costs), and optimal batch reproducibility.Citation368,369 Some of these agents are already approved for use in humans by the US FDA and equivalent regulatory agencies worldwide, implying that their evaluation as immunostimulatory agents in cancer patients presents limited safety concerns.Citation370 Along similar lines, some of these molecules have been (or are being) developed for other (oncological and non-oncological) indications, and have already been tested in Phase I clinical studies that demonstrated their safety.Citation370 It is therefore no surprise that small molecules targeting the immunological tumor microenvironment have attracted considerable attention over the past decade, resulting in the initiation of several clinical studies testing this immunotherapeutic paradigm in cancer patients.Citation37 Based on the number of ongoing clinical trials, IDO1 inhibitors (mainly indoximod and epacadostat), PLX3397 and galunisertib stand out as the most promising agents to target the immunological tumor microenvironment for therapeutic purposes. Nonetheless, the vast majority of clinical studies investigating these molecules in cancer patients are Phase I/II trials, perhaps suggesting that we will have to wait a little more to see a small molecule that mediates anticancer effects by acting on the immunological tumor microenvironment licensed for use in humans by regulatory agencies. We are looking very much forward to that moment.

Abbreviations

ADORA2A,=

adenosine A2a receptor;

ADORA2B,=

adenosine A2b receptor;

APC,=

antigen-presenting cell;

ARG,=

arginase;

CCL,=

chemokine (C–C motif) ligand;

CCR,=

chemokine (C–C motif) receptor;

CSF1R,=

colony-stimulating factor 1 receptor;

CTL,=

CD8+ cytotoxic T lymphocyte;

CTLA4,=

cytotoxic T lymphocyte-associated protein 4;

CXCL,=

chemokine (C–X–C motif) ligand;

CXCR,=

chemokine (C–X–C motif) receptor;

DC,=

dendritic cell;

FDA,=

Food and Drug Administration;

FLT3,=

fms-related tyrosine kinase 3;

ICD,=

immunogenic cell death;

IDO,=

indoleamine 2,3-dioxigenase;

IFN,=

interferon; IL, interleukin;

mAb,=

monoclonal antibody;

MDSC,=

myeloid-derived suppressor cell;

NK,=

natural killer;

NO,=

nitric oxide;

NSCLC,=

non-small cell lung carcinoma;

PGE2,=

prostaglandin E2;

PI3K,=

phosphoinositide-3-kinase;

TAM,=

tumor-associated macrophage;

TDO2,=

tryptophan 2,3-dioxigenase;

TGFB1,=

transforming growth factor β 1;

TLR,=

Toll-like receptor;

Treg,=

CD4+CD25+FOXP3+regulatory T cell;

VEGFA,=

vascular endothelial growth factor A.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Funding

Authors are supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

References

  • Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer 2004; 4:839-49; PMID:15516957; http://dx.doi.org/10.1038/nrc1477
  • Albini A, Tosetti F, Li VW, Noonan DM, Li WW. Cancer prevention by targeting angiogenesis. Nat Rev Clin Oncol 2012; 9:498-509; PMID:22850752; http://dx.doi.org/10.1038/nrclinonc.2012.120
  • Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12:298-306; PMID:22419253; http://dx.doi.org/10.1038/nrc3245
  • Albini A, Sporn MB. The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer 2007; 7:139-47; PMID:17218951; http://dx.doi.org/10.1038/nrc2067
  • Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. Cancer drug resistance: an evolving paradigm. Nat Rev Cancer 2013; 13:714-26; PMID:24060863; http://dx.doi.org/10.1038/nrc3599
  • Di Mitri D, Toso A, Alimonti A. Tumor-infiltrating myeloid cells drive senescence evasion and chemoresistance in tumors. Oncoimmunology 2015; 4:e988473; PMID:26405613; http://dx.doi.org/10.4161/2162402X.2014.988473
  • Fridman WH, Galon J, Pages F, Tartour E, Sautes-Fridman C, Kroemer G. Prognostic and predictive impact of intra- and peritumoral immune infiltrates. Cancer Res 2011; 71:5601-5; PMID:21846822; http://dx.doi.org/10.1158/0008-5472.CAN-11-1316
  • Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013; 14:1014-22; PMID:24048123; http://dx.doi.org/10.1038/ni.2703
  • Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 2008; 8:618-31; PMID:18633355; http://dx.doi.org/10.1038/nrc2444
  • Tjin EP, Luiten RM. Tumor-infiltrating T-cells: important players in clinical outcome of advanced melanoma patients. Oncoimmunology 2014; 3:e954862; PMID:25941603; http://dx.doi.org/10.4161/21624011.2014.954862
  • Issa-Nummer Y, Loibl S, von Minckwitz G, Denkert C. Tumor-infiltrating lymphocytes in breast cancer: A new predictor for responses to therapy. Oncoimmunology 2014; 3:e27926; PMID:25340002; http://dx.doi.org/10.4161/onci.27926
  • Semeraro M, Rusakiewicz S, Zitvogel L, Kroemer G. Natural killer cell mediated immunosurveillance of pediatric neuroblastoma. Oncoimmunology 2015; 4:e1042202; PMID:26451315; http://dx.doi.org/10.1080/2162402X.2015.1042202
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331:1565-70; PMID:21436444; http://dx.doi.org/10.1126/science.1203486
  • Kroemer G, Galluzzi L, Zitvogel L, Fridman WH. Colorectal cancer: the first neoplasia found to be under immunosurveillance and the last one to respond to immunotherapy? Oncoimmunology 2015; 4:e1058597; PMID:26140250; http://dx.doi.org/10.1080/2162402X.2015.1058597
  • Kallies A. T cell immunosurveillance controls B lymphoma development. Oncoimmunology 2014; 3:e28697; PMID:25050223; http://dx.doi.org/10.4161/onci.28697
  • Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol 2006; 6:715-27; PMID:16977338; http://dx.doi.org/10.1038/nri1936
  • Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002; 3:991-8; PMID:12407406; http://dx.doi.org/10.1038/ni1102-991
  • Matsushita H, Vesely MD, Koboldt DC, Rickert CG, Uppaluri R, Magrini VJ, Arthur CD, White JM, Chen YS, Shea LK et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012; 482:400-4; PMID:22318521; http://dx.doi.org/10.1038/nature10755
  • Danelli L, Frossi B, Pucillo CE. Mast cell/MDSC a liaison immunosuppressive for tumor microenvironment. Oncoimmunology 2015; 4:e1001232; PMID:26137400; http://dx.doi.org/10.1080/2162402X.2014.1001232
  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9:162-74; PMID:19197294; http://dx.doi.org/10.1038/nri2506
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12:253-68; PMID:22437938; http://dx.doi.org/10.1038/nri3175
  • Wu AA, Drake V, Huang HS, Chiu S, Zheng L. Reprogramming the tumor microenvironment: tumor-induced immunosuppressive factors paralyze T cells. Oncoimmunology 2015; 4:e1016700; PMID:26140242; http://dx.doi.org/10.1080/2162402X.2015.1016700
  • Pillarisetty VG. The pancreatic cancer microenvironment: an immunologic battleground. Oncoimmunology 2014; 3:e950171; PMID:25610740; http://dx.doi.org/10.4161/21624011.2014.950171
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323-43; PMID:23243596; http://dx.doi.org/10.4161/onci.22009
  • Tripathi C, Tewari BN, Kanchan RK, Baghel KS, Nautiyal N, Shrivastava R, Kaur H, Bhatt ML, Bhadauria S. Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget 2014; 5:5350-68; PMID:25051364; http://dx.doi.org/10.18632/oncotarget.2110
  • Balermpas P, Rodel F, Weiss C, Rodel C, Fokas E. Tumor-infiltrating lymphocytes favor the response to chemoradiotherapy of head and neck cancer. Oncoimmunology 2014; 3:e27403; PMID:24711959; http://dx.doi.org/10.4161/onci.27403
  • Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015; 28:690-714; PMID:26678337; http://dx.doi.org/10.1016/j.ccell.2015.10.012
  • Golden EB, Apetoh L. Radiotherapy and immunogenic cell death. Semin Radiat Oncol 2015; 25:11-7; PMID:25481261; http://dx.doi.org/10.1016/j.semradonc.2014.07.005
  • Grimaldi AM, Simeone E, Giannarelli D, Muto P, Falivene S, Borzillo V, Giugliano FM, Sandomenico F, Petrillo A, Curvietto M et al. Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 2014; 3:e28780; PMID:25083318; http://dx.doi.org/10.4161/onci.28780
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74-88; PMID:23890065; http://dx.doi.org/10.1016/j.immuni.2013.06.014
  • Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015; 520:373-7; PMID:25754329; http://dx.doi.org/10.1038/nature14292
  • Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014; 3:e28518; PMID:25071979; http://dx.doi.org/10.4161/onci.28518
  • Bloy N, Pol J, Manic G, Vitale I, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Radioimmunotherapy for oncological indications. Oncoimmunology 2014; 3:e954929; PMID:25941606; http://dx.doi.org/10.4161/21624011.2014.954929
  • Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I, Henrik Ter Meulen J, Zitvogel L, Kroemer G et al. Trial Watch: Toll-like receptor agonists in oncological indications. Oncoimmunology 2014; 3:e29179; PMID:25083332; http://dx.doi.org/10.4161/onci.29179
  • von Boehmer H, Daniel C. Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer. Nat Rev Drug Discov 2013; 12:51-63; PMID:23274471; http://dx.doi.org/10.1038/nrd3683
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12:252-64; PMID:22437870; http://dx.doi.org/10.1038/nrc3239
  • Adams JL, Smothers J, Srinivasan R, Hoos A. Big opportunities for small molecules in immuno-oncology. Nat Rev Drug Discov 2015; 14:603-22; PMID:26228631; http://dx.doi.org/10.1038/nrd4596
  • Hardwick N, Chung V, Cristea M, Ellenhorn JD, Diamond DJ. Overcoming immunosuppression to enhance a p53MVA vaccine. Oncoimmunology 2014; 3:e958949; PMID:25941580; http://dx.doi.org/10.4161/21624011.2014.958949
  • Hennessy EJ, Parker AE, O'Neill LA. Targeting Toll-like receptors: emerging therapeutics? Nat Rev Drug Discov 2010; 9:293-307; PMID:20380038; http://dx.doi.org/10.1038/nrd3203
  • O'Neill LA, Golenbock D, Bowie AG. The history of Toll-like receptors - redefining innate immunity. Nat Rev Immunol 2013; 13:453-60; PMID:23681101; http://dx.doi.org/10.1038/nri3446
  • Vacchelli E, Aranda F, Obrist F, Eggermont A, Galon J, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immunostimulatory cytokines in cancer therapy. Oncoimmunology 2014; 3:e29030; PMID:25083328; http://dx.doi.org/10.4161/onci.29030
  • Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer 2007; 7:95-106; PMID:17251916; http://dx.doi.org/10.1038/nrc2051
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27297; PMID:24701370; http://dx.doi.org/10.4161/onci.27297
  • Buque A, Bloy N, Aranda F, Castoldi F, Eggermont A, Cremer I, Fridman WH, Fucikova J, Galon J, Marabelle A et al. Trial Watch: Immunomodulatory monoclonal antibodies for oncological indications. Oncoimmunology 2015; 4:e1008814; PMID:26137403; http://dx.doi.org/10.1080/2162402X.2015.1008814
  • Apetoh L, Smyth MJ, Drake CG, Abastado JP, Apte RN, Ayyoub M, Blay JY, Bonneville M, Butterfield LH, Caignard A et al. Consensus nomenclature for CD8 T cell phenotypes in cancer. Oncoimmunology 2015; 4:e998538; PMID:26137416; http://dx.doi.org/10.1080/2162402X.2014.998538
  • Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015; 348:56-61; PMID:25838373; http://dx.doi.org/10.1126/science.aaa8172
  • Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015; 161:205-14; PMID:25860605; http://dx.doi.org/10.1016/j.cell.2015.03.030
  • Galluzzi L, Kroemer G, Eggermont A. Novel immune checkpoint blocker approved for the treatment of advanced melanoma. Oncoimmunology 2014; 3:e967147; PMID:25941597; http://dx.doi.org/10.4161/21624011.2014.967147
  • Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, Latreche S, Bergaya S, Benhamouda N, Tanchot C et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015; 212:139-48; PMID:25601652; http://dx.doi.org/10.1084/jem.20140559
  • Vacchelli E, Pol J, Bloy N, Eggermont A, Cremer I, Fridman WH, Galon J, Marabelle A, Kohrt H, Zitvogel L et al. Trial watch: Tumor-targeting monoclonal antibodies for oncological indications. Oncoimmunology 2015; 4:e985940; PMID:25949870; http://dx.doi.org/10.4161/2162402X.2014.985940
  • Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691; PMID:25941621; http://dx.doi.org/10.4161/21624011.2014.955691
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013; 31:51-72; PMID:23157435; http://dx.doi.org/10.1146/annurev-immunol-032712-100008
  • Gujar SA, Clements D, Lee PW. Two is better than one: Complementing oncolytic virotherapy with gemcitabine to potentiate antitumor immune responses. Oncoimmunology 2014; 3:e27622; PMID:24804161; http://dx.doi.org/10.4161/onci.27622
  • Wu J, Waxman DJ. Metronomic cyclophosphamide eradicates large implanted GL261 gliomas by activating antitumor Cd8 T-cell responses and immune memory. Oncoimmunology 2015; 4:e1005521; PMID:26137402; http://dx.doi.org/10.1080/2162402X.2015.1005521
  • Draghiciu O, Lubbers J, Nijman HW, Daemen T. Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy. Oncoimmunology 2015; 4:e954829; PMID:25949858; http://dx.doi.org/10.4161/21624011.2014.954829
  • Xu M, Liu M, Du X, Li S, Li H, Li X, Li Y, Wang Y, Qin Z, Fu YX et al. Intratumoral Delivery of IL-21 Overcomes Anti-Her2/Neu Resistance through Shifting Tumor-Associated Macrophages from M2 to M1 Phenotype. J Immunol 2015; 194:4997-5006; PMID:25876763; http://dx.doi.org/10.4049/jimmunol.1402603
  • Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of immunogenic cancer cell death. Cytokine Growth Factor Rev 2013; 24:319-33; PMID:23391812; http://dx.doi.org/10.1016/j.cytogfr.2013.01.005
  • Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer 2012; 12:860-75; PMID:23151605; http://dx.doi.org/10.1038/nrc3380
  • 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:e1008866; PMID:26137404; http://dx.doi.org/10.1080/2162402X.2015.1008866
  • Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2014; 3:e27878; PMID:24800173; http://dx.doi.org/10.4161/onci.27878
  • Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res 2010; 70:6171-80; PMID:20631075; http://dx.doi.org/10.1158/0008-5472.CAN-10-0153
  • Lob S, Konigsrainer A, Rammensee HG, Opelz G, Terness P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat Rev Cancer 2009; 9:445-52; PMID:19461669; http://dx.doi.org/10.1038/nrc2639
  • Moffett JR, Namboodiri MA. Tryptophan and the immune response. Immunol Cell Biol 2003; 81:247-65; PMID:12848846; http://dx.doi.org/10.1046/j.1440-1711.2003.t01-1-01177.x
  • Vonka V, Humlova Z, Klamova H, Kujovska-Krcmova L, Petrackova M, Hamsikova E, Krmencikova-Fliegl M, Duskova M, Roth Z. Kynurenine and uric acid levels in chronic myeloid leukemia patients. Oncoimmunology 2015; 4:e992646; PMID:25949913; http://dx.doi.org/10.4161/2162402X.2014.992646
  • Guillonneau C, Hill M, Hubert FX, Chiffoleau E, Herve C, Li XL, Heslan M, Usal C, Tesson L, Ménoret S et al. CD40Ig treatment results in allograft acceptance mediated by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase. J Clin Invest 2007; 117:1096-106; PMID:17404623; http://dx.doi.org/10.1172/JCI28801
  • Hwu P, Du MX, Lapointe R, Do M, Taylor MW, Young HA. Indoleamine 2,3-dioxygenase production by human dendritic cells results in the inhibition of T cell proliferation. J Immunol 2000; 164:3596-9; PMID:10725715; http://dx.doi.org/10.4049/jimmunol.164.7.3596
  • Giannoni P, Pietra G, Travaini G, Quarto R, Shyti G, Benelli R, Ottaggio L, Mingari MC, Zupo S, Cutrona G et al. Chronic lymphocytic leukemia nurse-like cells express hepatocyte growth factor receptor (c-MET) and indoleamine 2,3-dioxygenase and display features of immunosuppressive type 2 skewed macrophages. Haematologica 2014; 99:1078-87; PMID:24561793; http://dx.doi.org/10.3324/haematol.2013.091405
  • Rani R, Jordan MB, Divanovic S, Herbert DR. IFN-gamma-driven IDO production from macrophages protects IL-4Ralpha-deficient mice against lethality during Schistosoma mansoni infection. Am J Pathol 2012; 180:2001-8; PMID:22426339; http://dx.doi.org/10.1016/j.ajpath.2012.01.013
  • Potula R, Poluektova L, Knipe B, Chrastil J, Heilman D, Dou H, Takikawa O, Munn DH, Gendelman HE, Persidsky Y. Inhibition of indoleamine 2,3-dioxygenase (IDO) enhances elimination of virus-infected macrophages in an animal model of HIV-1 encephalitis. Blood 2005; 106:2382-90; PMID:15961516; http://dx.doi.org/10.1182/blood-2005-04-1403
  • Yu J, Wang Y, Yan F, Zhang P, Li H, Zhao H, Yan C, Yan F, Ren X. Noncanonical NF-kappaB activation mediates STAT3-stimulated IDO upregulation in myeloid-derived suppressor cells in breast cancer. J Immunol 2014; 193:2574-86; PMID:25063873; http://dx.doi.org/10.4049/jimmunol.1400833
  • Vigneron N, van Baren N, Van den Eynde BJ. Expression profile of the human IDO1 protein, a cancer drug target involved in tumoral immune resistance. Oncoimmunology 2015; 4:e1003012; PMID:26155395; http://dx.doi.org/10.1080/2162402X.2014.1003012
  • Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL. Inhibition of T cell proliferation by macrophage tryptophan catabolism. J Exp Med 1999; 189:1363-72; PMID:10224276; http://dx.doi.org/10.1084/jem.189.9.1363
  • 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; 281:1191-3; PMID:9712583; http://dx.doi.org/10.1126/science.281.5380.1191
  • Uyttenhove C, Pilotte L, Theate 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; 9:1269-74; PMID:14502282; http://dx.doi.org/10.1038/nm934
  • Fallarino F, Grohmann U, Vacca C, Orabona C, Spreca A, Fioretti MC, Puccetti P. T cell apoptosis by kynurenines. Adv Exp Med Biol 2003; 527:183-90; PMID:15206731; http://dx.doi.org/10.1007/978-1-4615-0135-0_21
  • Hayashi T, Mo JH, Gong X, Rossetto C, Jang A, Beck L, Elliott GI, Kufareva I, Abagyan R, Broide DH et al. 3-Hydroxyanthranilic acid inhibits PDK1 activation and suppresses experimental asthma by inducing T cell apoptosis. Proc Natl Acad Sci U S A 2007; 104:18619-24; PMID:18003900; http://dx.doi.org/10.1073/pnas.0709261104
  • 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; 171:1652-5; PMID:12902462; http://dx.doi.org/10.4049/jimmunol.171.4.1652
  • Baban B, Hansen AM, Chandler PR, Manlapat A, Bingaman A, Kahler DJ, Munn DH, Mellor AL. A minor population of splenic dendritic cells expressing CD19 mediates IDO-dependent T cell suppression via type I IFN signaling following B7 ligation. Int Immunol 2005; 17:909-19; PMID:15967784; http://dx.doi.org/10.1093/intimm/dxh271
  • Molano A, Illarionov PA, Besra GS, Putterman C, Porcelli SA. Modulation of invariant natural killer T cell cytokine responses by indoleamine 2,3-dioxygenase. Immunol Lett 2008; 117:81-90; PMID:18272236; http://dx.doi.org/10.1016/j.imlet.2007.12.013
  • Adikari SB, Lian H, Link H, Huang YM, Xiao BG. Interferon-gamma-modified dendritic cells suppress B cell function and ameliorate the development of experimental autoimmune myasthenia gravis. Clin Exp Immunol 2004; 138:230-6; PMID:15498031; http://dx.doi.org/10.1111/j.1365-2249.2004.02585.x
  • Munn DH, Sharma MD, Lee JR, Jhaver KG, Johnson TS, Keskin DB, Marshall B, Chandler P, Antonia SJ, Burgess R et al. Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase. Science 2002; 297:1867-70; PMID:12228717; http://dx.doi.org/10.1126/science.1073514
  • Fallarino F, Orabona C, Vacca C, Bianchi R, Gizzi S, Asselin-Paturel C, Fioretti MC, Trinchieri G, Grohmann U, Puccetti P. Ligand and cytokine dependence of the immunosuppressive pathway of tryptophan catabolism in plasmacytoid dendritic cells. Int Immunol 2005; 17:1429-38; PMID:16172135; http://dx.doi.org/10.1093/intimm/dxh321
  • Fallarino F, Asselin-Paturel C, Vacca C, Bianchi R, Gizzi S, Fioretti MC, Trinchieri G, Grohmann U, Puccetti P. Murine plasmacytoid dendritic cells initiate the immunosuppressive pathway of tryptophan catabolism in response to CD200 receptor engagement. J Immunol 2004; 173:3748-54; PMID:15356121; http://dx.doi.org/10.4049/jimmunol.173.6.3748
  • Sharma MD, Baban B, Chandler P, Hou DY, Singh N, Yagita H, Azuma M, Blazar BR, Mellor AL, Munn DH. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest 2007; 117:2570-82; PMID:17710230; http://dx.doi.org/10.1172/JCI31911
  • 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; 179:3443-51; PMID:17785778; http://dx.doi.org/10.4049/jimmunol.179.6.3443
  • Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest 2004; 114:280-90; PMID:15254595; http://dx.doi.org/10.1172/JCI200421583
  • Vacchelli E, Aranda F, Eggermont A, Sautes-Fridman C, Tartour E, Kennedy EP, Platten M, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2014; 3:e957994; PMID:25941578; http://dx.doi.org/10.4161/21624011.2014.957994
  • Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol 2013; 34:137-43; PMID:23103127; http://dx.doi.org/10.1016/j.it.2012.10.001
  • Balachandran VP, Cavnar MJ, Zeng S, Bamboat ZM, Ocuin LM, Obaid H, Sorenson EC, Popow R, Ariyan C, Rossi F et al. Imatinib potentiates antitumor T cell responses in gastrointestinal stromal tumor through the inhibition of Ido. Nat Med 2011; 17:1094-100; PMID:21873989; http://dx.doi.org/10.1038/nm.2438
  • Kroemer G, Senovilla L, Galluzzi L, Andre F, Zitvogel L. Natural and therapy-induced immunosurveillance in breast cancer. Nat Med 2015; 21:1128-38; PMID:26444637; http://dx.doi.org/10.1038/nm.3944
  • Quintas-Cardama A, Kantarjian H, Cortes J. Imatinib and beyond–exploring the full potential of targeted therapy for CML. Nat Rev Clin Oncol 2009; 6:535-43; PMID:19652654; http://dx.doi.org/10.1038/nrclinonc.2009.112
  • Capdeville R, Buchdunger E, Zimmermann J, Matter A. Glivec (STI571, imatinib), a rationally developed, targeted anticancer drug. Nat Rev Drug Discov 2002; 1:493-502; PMID:12120256; http://dx.doi.org/10.1038/nrd839
  • 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; 21:5427-33; PMID:26519060; http://dx.doi.org/10.1158/1078-0432.CCR-15-0420
  • Qian F, Liao J, Villella J, Edwards R, Kalinski P, Lele S, Shrikant P, Odunsi K. Effects of 1-methyltryptophan stereoisomers on IDO2 enzyme activity and IDO2-mediated arrest of human T cell proliferation. Cancer Immunol Immunother 2012; 61:2013-20; PMID:22527253; http://dx.doi.org/10.1007/s00262-012-1265-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; 11:312-9; PMID:15711557; http://dx.doi.org/10.1038/nm1196
  • 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; PMID:17234791; http://dx.doi.org/10.1158/0008-5472.CAN-06-2925
  • Holmgaard RB, Zamarin D, Munn DH, Wolchok JD, Allison JP. Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4. J Exp Med 2013; 210:1389-402; PMID:23752227; http://dx.doi.org/10.1084/jem.20130066
  • Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, Cheng Y, Kim JW, Qiao J, Zhang L et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res 2014; 20:5290-301; PMID:24691018; http://dx.doi.org/10.1158/1078-0432.CCR-14-0514
  • Wainwright DA, Lesniak MS. Menage a trois: Sustained therapeutic anti-tumor immunity requires multiple partners in malignant glioma. Oncoimmunology 2014; 3:e28927; PMID:25057450; http://dx.doi.org/10.4161/onci.28927
  • Metz R, Duhadaway JB, Kamasani U, Laury-Kleintop L, Muller AJ, Prendergast GC. Novel tryptophan catabolic enzyme IDO2 is the preferred biochemical target of the antitumor indoleamine 2,3-dioxygenase inhibitory compound D-1-methyl-tryptophan. Cancer Res 2007; 67:7082-7; PMID:17671174; http://dx.doi.org/10.1158/0008-5472.CAN-07-1872
  • 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; 291:326-33; PMID:1952947; http://dx.doi.org/10.1016/0003-9861(91)90142-6
  • Soliman HH, Jackson E, Neuger T, Dees EC, Harvey RD, Han H, Ismail-Khan R, Minton S, Vahanian NN, Link C et al. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 2014; 5:8136-46; PMID:25327557; http://dx.doi.org/10.18632/oncotarget.2357
  • Jackson E, Dees EC, Kauh JS, Harvey RD, Neuger A, Lush R, Antonia JS, Minton SE, Ismail-Khan R, Han HS et al. A phase I study of indoximod in combination with docetaxel in metastatic solid tumors. J Clin Oncol 2013; 31: abstr 3026; PMID:23857967; http://dx.doi.org/10.1200/JCO.2012.47.1235
  • Colman H, Mott F, Spira AI, Johnson TS, Zakharia Y, Vahanian NN, Link CJ, Kennedy EP, Sadek RF, Munn D et al. A phase 1b/2 study of the combination of the IDO pathway inhibitor indoximod and temozolomide for adult patients with temozolomide-refractory primary malignant brain tumors: Safety analysis and preliminary efficacy of the phase 1b component. ASCO Meeting Abstracts 2015; 33:2070
  • Soliman HH, Minton SE, Ismail-Khan R, Han HS, Janssen W, Vahanian NN et al. A phase 2 study of Ad.p53 DC vaccine in combination with indoximod in metastatic solid tumors. J Clin Oncol 2013; 31:abstr 3069; http://dx.doi.org/10.1200/JCO.2013.49.9202
  • Zakharia Y, Johnson TS, Colman H, Vahanian NN, Link CJ, Kennedy E et al. A phase I/II study of the combination of indoximod and temozolomide for adult patients with temozolomide-refractory primary malignant brain tumors. J Clin Oncol 2014; 32: abstr TPS2107
  • Jha GG, Miller JS. A randomized, double-blind phase 2 study of sipuleucel-T followed by indoximod or placebo in the treatment of patients with asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer. ASCO Meeting Abstracts 2014; 32:TPS5111
  • Soliman HH, Minton SE, Ismail-Khan R, Han HS, Vahanian NN, Ramsey WJ et al. A phase 2 study of docetaxel in combination with indoximod in metastatic breast cancer. J Clin Oncol 2014; 32: abstr TPS3124
  • Kennedy E, Rossi GR, Vahanian NN, Link CJ. Phase 1/2 trial of the indoleamine 2,3-dioxygenase pathway (IDO) inhibitor indoximod plus ipilimumab for the treatment of unresectable stage 3 or 4 melanoma. ASCO Meeting Abstracts 2014; 32:TPS9117
  • Koblish HK, Hansbury MJ, Bowman KJ, Yang G, Neilan CL, Haley PJ, Burn TC, Waeltz P, Sparks RB, Yue EW et al. Hydroxyamidine inhibitors of indoleamine-2,3-dioxygenase potently suppress systemic tryptophan catabolism and the growth of IDO-expressing tumors. Mol Cancer Ther 2010; 9:489-98; PMID:20124451; http://dx.doi.org/10.1158/1535-7163.MCT-09-0628
  • Liu X, Shin N, Koblish HK, Yang G, Wang Q, Wang K, Leffet L, Hansbury MJ, Thomas B, Rupar M et al. Selective inhibition of IDO1 effectively regulates mediators of antitumor immunity. Blood 2010; 115:3520-30; PMID:20197554; http://dx.doi.org/10.1182/blood-2009-09-246124
  • Li M, Bolduc AR, Hoda MN, Gamble DN, Dolisca SB, Bolduc AK, Hoang K, Ashley C, McCall D, Rojiani AM et al. The indoleamine 2,3-dioxygenase pathway controls complement-dependent enhancement of chemo-radiation therapy against murine glioblastoma. J Immunother Cancer 2014; 2:21; PMID:25054064; http://dx.doi.org/10.1186/2051-1426-2-21
  • Mautino MR, Jaipuri FA, Waldo J, Kumar S, Adams J, Van Allen C et al. NLG919, a novel indoleamine-2,3-dioxygenase (IDO)-pathway inhibitor drug candidate for cancer therapy. Cancer Res 2013; 73:491; http://dx.doi.org/10.1158/1538-7445.AM2013-491
  • Beatty GL, O'Dwyer PJ, Clark J, Shi JG, Newton RC, Schaub R et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients (pts) with advanced malignancies. ASCO Meeting Abstracts 2013; 31:3025
  • Newton RC, Scherle PA, Bowman K, Liu X, Beatty GL, O'Dwyer PJ et al. Pharmacodynamic assessment of INCB024360, an inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), in advanced cancer patients. ASCO Meeting Abstracts 2012; 30:2500
  • Gibney GT, Hamid O, Gangadhar TC, Lutzky J, Olszanski AJ, Gajewski T et al. Preliminary results from a phase 1/2 study of INCB024360 combined with ipilimumab (ipi) in patients (pts) with melanoma. ASCO Meeting Abstracts 2014; 32:3010
  • Tanaka M, Li X, Hikawa H, Suzuki T, Tsutsumi K, Sato M, Takikawa O, Suzuki H, Yokoyama Y. Synthesis and biological evaluation of novel tryptoline derivatives as indoleamine 2,3-dioxygenase (IDO) inhibitors. Bioorg Med Chem 2013; 21:1159-65; PMID:23337802; http://dx.doi.org/10.1016/j.bmc.2012.12.028
  • Gaspari P, Banerjee T, Malachowski WP, Muller AJ, Prendergast GC, DuHadaway J, Bennett S, Donovan AM. Structure-activity study of brassinin derivatives as indoleamine 2,3-dioxygenase inhibitors. J Med Chem 2006; 49:684-92; PMID:16420054; http://dx.doi.org/10.1021/jm0508888
  • Banerjee T, Duhadaway JB, Gaspari P, Sutanto-Ward E, Munn DH, Mellor AL, Malachowski WP, Prendergast GC, Muller AJ. A key in vivo antitumor mechanism of action of natural product-based brassinins is inhibition of indoleamine 2,3-dioxygenase. Oncogene 2008; 27:2851-7; PMID:18026137; http://dx.doi.org/10.1038/sj.onc.1210939
  • Pereira A, Vottero E, Roberge M, Mauk AG, Andersen RJ. Indoleamine 2,3-dioxygenase inhibitors from the Northeastern Pacific Marine Hydroid Garveia annulata. J Nat Prod 2006; 69:1496-9; PMID:17067170; http://dx.doi.org/10.1021/np060111x
  • Kumar S, Malachowski WP, DuHadaway JB, LaLonde JM, Carroll PJ, Jaller D, Metz R, Prendergast GC, Muller AJ. Indoleamine 2,3-dioxygenase is the anticancer target for a novel series of potent naphthoquinone-based inhibitors. J Med Chem 2008; 51:1706-18; PMID:18318466; http://dx.doi.org/10.1021/jm7014155
  • Volgraf M, Lumb JP, Brastianos HC, Carr G, Chung MK, Munzel M, Mauk AG, Andersen RJ, Trauner D. Biomimetic synthesis of the IDO inhibitors exiguamine A and B. Nat Chem Biol 2008; 4:535-7; PMID:18677305; http://dx.doi.org/10.1038/nchembio.107
  • Brastianos HC, Vottero E, Patrick BO, Van Soest R, Matainaho T, Mauk AG, Andersen RJ. Exiguamine A, an indoleamine-2,3-dioxygenase (IDO) inhibitor isolated from the marine sponge Neopetrosia exigua. J Am Chem Soc 2006; 128:16046-7; PMID:17165752; http://dx.doi.org/10.1021/ja067211+
  • 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; 109:2497-502; PMID:22308364; http://dx.doi.org/10.1073/pnas.1113873109
  • Junger WG. Immune cell regulation by autocrine purinergic signalling. Nat Rev Immunol 2011; 11:201-12; PMID:21331080; http://dx.doi.org/10.1038/nri2938
  • Michaud M, Xie X, Bravo-San Pedro JM, Zitvogel L, White E, Kroemer G. An autophagy-dependent anticancer immune response determines the efficacy of melanoma chemotherapy. Oncoimmunology 2014; 3:e944047; PMID:25610726; http://dx.doi.org/10.4161/21624011.2014.944047
  • Adkins I, Fucikova J, Garg AD, Agostinis P, Spisek R. Physical modalities inducing immunogenic tumor cell death for cancer immunotherapy. Oncoimmunology 2014; 3:e968434; PMID:25964865; http://dx.doi.org/10.4161/21624011.2014.968434
  • Ma Y, Adjemian S, Mattarollo SR, Yamazaki T, Aymeric L, Yang H, Portela Catani JP, Hannani D, Duret H, Steegh K et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity 2013; 38:729-41; PMID:23562161; http://dx.doi.org/10.1016/j.immuni.2013.03.003
  • Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med 2009; 15:1170-8; PMID:19767732; http://dx.doi.org/10.1038/nm.2028
  • Antonioli L, Pacher P, Vizi ES, Hasko G. CD39 and CD73 in immunity and inflammation. Trends Mol Med 2013; 19:355-67; PMID:23601906; http://dx.doi.org/10.1016/j.molmed.2013.03.005
  • Beavis PA, Slaney CY, Milenkovski N, Henderson MA, Loi S, Stagg J, Kershaw MH, Darcy PK. CD73: A potential biomarker for anti-PD-1 therapy. Oncoimmunology 2015; 4:e1046675; PMID:26451321; http://dx.doi.org/10.1080/2162402X.2015.1046675
  • Antonioli L, Blandizzi C, Pacher P, Hasko G. Immunity, inflammation and cancer: a leading role for adenosine. Nat Rev Cancer 2013; 13:842-57; PMID:24226193; http://dx.doi.org/10.1038/nrc3613
  • Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 2007; 204:1257-65; PMID:17502665; http://dx.doi.org/10.1084/jem.20062512
  • Cekic C, Day YJ, Sag D, Linden J. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res 2014; 74:7250-9; PMID:25377469; http://dx.doi.org/10.1158/0008-5472.CAN-13-3583
  • Hilchey SP, Kobie JJ, Cochran MR, Secor-Socha S, Wang JC, Hyrien O, Burack WR, Mosmann TR, Quataert SA, Bernstein SH. Human follicular lymphoma CD39+-infiltrating T cells contribute to adenosine-mediated T cell hyporesponsiveness. J Immunol 2009; 183:6157-66; PMID:19864600; http://dx.doi.org/10.4049/jimmunol.0900475
  • Ohta A, Kini R, Ohta A, Subramanian M, Madasu M, Sitkovsky M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol 2012; 3:190; PMID:22783261; http://dx.doi.org/10.3389/fimmu.2012.00190
  • Bergamin LS, Braganhol E, Figueiro F, Casali EA, Zanin RF, Sevigny J, Battastini AM. Involvement of purinergic system in the release of cytokines by macrophages exposed to glioma-conditioned medium. J Cell Biochem 2015; 116:721-9; PMID:25546398; http://dx.doi.org/10.1002/jcb.25018
  • Csoka B, Selmeczy Z, Koscso B, Nemeth ZH, Pacher P, Murray PJ, Kepka-Lenhart D, Morris SM Jr, Gause WC, Leibovich SJ et al. Adenosine promotes alternative macrophage activation via A2A and A2B receptors. FASEB J 2012; 26:376-86; PMID:21926236; http://dx.doi.org/10.1096/fj.11-190934
  • Ryzhov S, Novitskiy SV, Goldstein AE, Biktasova A, Blackburn MR, Biaggioni I, Dikov MM, Feoktistov I. Adenosinergic regulation of the expansion and immunosuppressive activity of CD11b+Gr1+ cells. J Immunol 2011; 187:6120-9; PMID:22039302; http://dx.doi.org/10.4049/jimmunol.1101225
  • Morello S, Miele L. Targeting the adenosine A2b receptor in the tumor microenvironment overcomes local immunosuppression by myeloid-derived suppressor cells. Oncoimmunology 2014; 3:e27989; PMID:25101221; http://dx.doi.org/10.4161/onci.27989
  • Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY, Blackburn MR, Biaggioni I, Carbone DP, Feoktistov I et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008; 112:1822-31; PMID:18559975; http://dx.doi.org/10.1182/blood-2008-02-136325
  • Sheth S, Brito R, Mukherjea D, Rybak LP, Ramkumar V. Adenosine receptors: expression, function and regulation. Int J Mol Sci 2014; 15:2024-52; PMID:24477263; http://dx.doi.org/10.3390/ijms15022024
  • Bianchi G, Vuerich M, Pellegatti P, Marimpietri D, Emionite L, Marigo I, Bronte V, Di Virgilio F, Pistoia V, Raffaghello L. ATP/P2X7 axis modulates myeloid-derived suppressor cell functions in neuroblastoma microenvironment. Cell Death Dis 2014; 5:e1135; PMID:24651438; http://dx.doi.org/10.1038/cddis.2014.109
  • Chadet S, Ivanes F, Benoist L, Salmon-Gandonniere C, Guibon R, Velge-Roussel F, Babuty D, Baron C, Roger S, Angoulvant D. Hypoxia/Reoxygenation Inhibits P2Y11 Receptor Expression and Its Immunosuppressive Activity in Human Dendritic Cells. J Immunol 2015; 195:651-60; PMID:26078273; http://dx.doi.org/10.4049/jimmunol.1500197
  • Smyth MJ, Ngiow SF, Ribas A, Teng MW. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol 2016; 13:143-58; PMID:26598942; http://dx.doi.org/10.1038/nrclinonc.2015.209
  • Bonnefoy N, Bastid J, Alberici G, Bensussan A, Eliaou JF. CD39: A complementary target to immune checkpoints to counteract tumor-mediated immunosuppression. Oncoimmunology 2015; 4:e1003015; PMID:26155397; http://dx.doi.org/10.1080/2162402X.2014.1003015
  • Baqi Y, Weyler S, Iqbal J, Zimmermann H, Muller CE. Structure-activity relationships of anthraquinone derivatives derived from bromaminic acid as inhibitors of ectonucleoside triphosphate diphosphohydrolases (E-NTPDases). Purinergic Signal 2009; 5:91-106; PMID:18528783; http://dx.doi.org/10.1007/s11302-008-9103-5
  • Lecka J, Gillerman I, Fausther M, Salem M, Munkonda MN, Brosseau JP, Cadot C, Martín-Satué M, d'Orléans-Juste P, Rousseau E et al. 8-BuS-ATP derivatives as specific NTPDase1 inhibitors. Br J Pharmacol 2013; 169:179-96; PMID:23425137; http://dx.doi.org/10.1111/bph.12135
  • Knapp K, Zebisch M, Pippel J, El-Tayeb A, Muller CE, Strater N. Crystal structure of the human ecto-5′-nucleotidase (CD73): insights into the regulation of purinergic signaling. Structure 2012; 20:2161-73; PMID:23142347; http://dx.doi.org/10.1016/j.str.2012.10.001
  • Krasteva M, Barth A. Structures of the Ca2+-ATPase complexes with ATP, AMPPCP and AMPPNP. An FTIR study. Biochim Biophys Acta 2007; 1767:114-23; PMID:17157262; http://dx.doi.org/10.1016/j.bbabio.2006.11.003
  • Bhattarai S, Freundlieb M, Pippel J, Meyer A, Abdelrahman A, Fiene A, Lee SY, Zimmermann H, Yegutkin GG, Sträter N et al. alpha,beta-Methylene-ADP (AOPCP) Derivatives and Analogues: Development of Potent and Selective ecto-5′-Nucleotidase (CD73) Inhibitors. J Med Chem 2015; 58:6248-63; PMID:26147331; http://dx.doi.org/10.1021/acs.jmedchem.5b00802
  • Forte G, Sorrentino R, Montinaro A, Luciano A, Adcock IM, Maiolino P, Arra C, Cicala C, Pinto A, Morello S. Inhibition of CD73 improves B cell-mediated anti-tumor immunity in a mouse model of melanoma. J Immunol 2012; 189:2226-33; PMID:22826317; http://dx.doi.org/10.4049/jimmunol.1200744
  • Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A 2013; 110:14711-6; PMID:23964122; http://dx.doi.org/10.1073/pnas.1308209110
  • Jin D, Fan J, Wang L, Thompson LF, Liu A, Daniel BJ, Shin T, Curiel TJ, Zhang B. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res 2010; 70:2245-55; PMID:20179192; http://dx.doi.org/10.1158/0008-5472.CAN-09-3109
  • Stagg J, Divisekera U, Duret H, Sparwasser T, Teng MW, Darcy PK, Smyth MJ. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res 2011; 71:2892-900; PMID:21292811; http://dx.doi.org/10.1158/0008-5472.CAN-10-4246
  • Yegutkin GG, Marttila-Ichihara F, Karikoski M, Niemela J, Laurila JP, Elima K, Jalkanen S, Salmi M. Altered purinergic signaling in CD73-deficient mice inhibits tumor progression. Eur J Immunol 2011; 41:1231-41; PMID:21469131; http://dx.doi.org/10.1002/eji.201041292
  • Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011; 334:1573-7; PMID:22174255; http://dx.doi.org/10.1126/science.1208347
  • Wang L, Fan J, Thompson LF, Zhang Y, Shin T, Curiel TJ, Zhang B. CD73 has distinct roles in nonhematopoietic and hematopoietic cells to promote tumor growth in mice. J Clin Invest 2011; 121:2371-82; PMID:21537079; http://dx.doi.org/10.1172/JCI45559
  • Ntantie E, Gonyo P, Lorimer EL, Hauser AD, Schuld N, McAllister D, Kalyanaraman B, Dwinell MB, Auchampach JA, Williams CL. An adenosine-mediated signaling pathway suppresses prenylation of the GTPase Rap1B and promotes cell scattering. Sci Signal 2013; 6:ra39; PMID:23716716; http://dx.doi.org/10.1126/scisignal.2003374.
  • Zhou X, Zhi X, Zhou P, Chen S, Zhao F, Shao Z, Ou Z, Yin L. Effects of ecto-5′-nucleotidase on human breast cancer cell growth in vitro and in vivo. Oncol Rep 2007; 17:1341-6; PMID:17487388; http://dx.doi.org/10.3892/or.17.6.1341
  • Bastid J, Regairaz A, Bonnefoy N, Dejou C, Giustiniani J, Laheurte C, Cochaud S, Laprevotte E, Funck-Brentano E, Hemon P et al. Inhibition of CD39 enzymatic function at the surface of tumor cells alleviates their immunosuppressive activity. Cancer Immunol Res 2015; 3:254-65; PMID:25403716; http://dx.doi.org/10.1158/2326-6066.CIR-14-0018
  • Clayton A, Al-Taei S, Webber J, Mason MD, Tabi Z. Cancer exosomes express CD39 and CD73, which suppress T cells through adenosine production. J Immunol 2011; 187:676-83; PMID:21677139; http://dx.doi.org/10.4049/jimmunol.1003884
  • Iannone R, Miele L, Maiolino P, Pinto A, Morello S. Adenosine limits the therapeutic effectiveness of anti-CTLA4 mAb in a mouse melanoma model. Am J Cancer Res 2014; 4:172-81; PMID:24660106
  • Villalona-Calero MA, Wientjes MG, Otterson GA, Kanter S, Young D, Murgo AJ, Fischer B, DeHoff C, Chen D, Yeh TK et al. Phase I study of low-dose suramin as a chemosensitizer in patients with advanced non-small cell lung cancer. Clin Cancer Res 2003; 9:3303-11; PMID:12960116
  • Villalona-Calero MA, Otterson GA, Wientjes MG, Weber F, Bekaii-Saab T, Young D, Murgo AJ, Jensen R, Yeh TK, Wei Y et al. Noncytotoxic suramin as a chemosensitizer in patients with advanced non-small-cell lung cancer: a phase II study. Ann Oncol 2008; 19:1903-9; PMID:18632723; http://dx.doi.org/10.1093/annonc/mdn412
  • George S, Dreicer R, Au JJ, Shen T, Rini BI, Roman S, Cooney MM, Mekhail T, Elson P, Wientjes GM et al. Phase I/II trial of 5-fluorouracil and a noncytotoxic dose level of suramin in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 2008; 6:79-85; PMID:18824429; http://dx.doi.org/10.3816/CGC.2008.n.012
  • Lam ET, Au JL, Otterson GA, Guillaume Wientjes M, Chen L, Shen T, Wei Y, Li X, Bekaii-Saab T, Murgo AJ et al. Phase I trial of non-cytotoxic suramin as a modulator of docetaxel and gemcitabine therapy in previously treated patients with non-small cell lung cancer. Cancer Chemother Pharmacol 2010; 66:1019-29; PMID:20107799; http://dx.doi.org/10.1007/s00280-010-1252-x
  • Lustberg MB, Pant S, Ruppert AS, Shen T, Wei Y, Chen L, Brenner L, Shiels D, Jensen RR, Berger M et al. Phase I/II trial of non-cytotoxic suramin in combination with weekly paclitaxel in metastatic breast cancer treated with prior taxanes. Cancer Chemother Pharmacol 2012; 70:49-56; PMID:22729159; http://dx.doi.org/10.1007/s00280-012-1887-x
  • Meis S, Hamacher A, Hongwiset D, Marzian C, Wiese M, Eckstein N, Royer HD, Communi D, Boeynaems JM, Hausmann R et al. NF546 [4,4′-(carbonylbis(imino-3,1-phenylene-carbonylimino-3,1-(4-methyl-phenylene)-car bonylimino))-bis(1,3-xylene-alpha,alpha'-diphosphonic acid) tetrasodium salt] is a non-nucleotide P2Y11 agonist and stimulates release of interleukin-8 from human monocyte-derived dendritic cells. J Pharmacol Exp Ther 2010; 332:238-47; PMID:19815812; http://dx.doi.org/10.1124/jpet.109.157750
  • Mittal D, Young A, Stannard K, Yong M, Teng MW, Allard B, Stagg J, Smyth MJ. Antimetastatic effects of blocking PD-1 and the adenosine A2A receptor. Cancer Res 2014; 74:3652-8; PMID:24986517; http://dx.doi.org/10.1158/0008-5472.CAN-14-0957
  • Young A, Mittal D, Stannard K, Yong M, Teng MW, Allard B, Stagg J, Smyth MJ. Co-blockade of immune checkpoints and adenosine A receptor suppresses metastasis. Oncoimmunology 2014; 3:e958952; PMID:25941583; http://dx.doi.org/10.4161/21624011.2014.958952
  • Vorovenci RJ, Antonini A. The efficacy of oral adenosine A2A antagonist istradefylline for the treatment of moderate to severe Parkinson's disease. Expert Rev Neurother 2015; 15:1383-90; PMID:26630457; http://dx.doi.org/10.1586/14737175.2015.1113131
  • Jenner P. An overview of adenosine A2A receptor antagonists in Parkinson's disease. Int Rev Neurobiol 2014; 119:71-86; PMID:25175961; http://dx.doi.org/10.1016/B978-0-12-801022-8.00003-9
  • Hauser RA, Stocchi F, Rascol O, Huyck SB, Capece R, Ho TW, Sklar P, Lines C, Michelson D, Hewitt D. Preladenant as an Adjunctive Therapy With Levodopa in Parkinson Disease: Two Randomized Clinical Trials and Lessons Learned. JAMA Neurol 2015; 72:1491-500; PMID:26523919; http://dx.doi.org/10.1001/jamaneurol.2015.2268
  • Factor SA, Wolski K, Togasaki DM, Huyck S, Cantillon M, Ho TW, Hauser RA, Pourcher E. Long-term safety and efficacy of preladenant in subjects with fluctuating Parkinson's disease. Mov Disord 2013; 28:817-20; PMID:23589371; http://dx.doi.org/10.1002/mds.25395
  • Chen JF, Eltzschig HK, Fredholm BB. Adenosine receptors as drug targets–what are the challenges? Nat Rev Drug Discov 2013; 12:265-86; PMID:23535933; http://dx.doi.org/10.1038/nrd3955
  • Iannone R, Miele L, Maiolino P, Pinto A, Morello S. Blockade of A2b adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma. Neoplasia 2013; 15:1400-9; PMID:24403862; http://dx.doi.org/10.1593/neo.131748
  • Kaji W, Tanaka S, Tsukimoto M, Kojima S. Adenosine A(2B) receptor antagonist PSB603 suppresses tumor growth and metastasis by inhibiting induction of regulatory T cells. J Toxicol Sci 2014; 39:191-8; PMID:24646699; http://dx.doi.org/10.2131/jts.39.191
  • Sorrentino C, Miele L, Porta A, Pinto A, Morello S. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 2015; 6:27478-89; PMID:26317647; http://dx.doi.org/10.18632/oncotarget.4393
  • Ochoa-Cortes F, Linan-Rico A, Jacobson KA, Christofi FL. Potential for developing purinergic drugs for gastrointestinal diseases. Inflamm Bowel Dis 2014; 20:1259-87; PMID:24859298; http://dx.doi.org/10.1097/MIB.0000000000000047
  • Polosa R, Blackburn MR. Adenosine receptors as targets for therapeutic intervention in asthma and chronic obstructive pulmonary disease. Trends Pharmacol Sci 2009; 30:528-35; PMID:19762093; http://dx.doi.org/10.1016/j.tips.2009.07.005
  • Kalla RV, Zablocki J. Progress in the discovery of selective, high affinity A(2B) adenosine receptor antagonists as clinical candidates. Purinergic Signal 2009; 5:21-9; PMID:18568423; http://dx.doi.org/10.1007/s11302-008-9119-x
  • Kaidi A, Qualtrough D, Williams AC, Paraskeva C. Direct transcriptional up-regulation of cyclooxygenase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor cell survival and enhances HIF-1 transcriptional activity during hypoxia. Cancer Res 2006; 66:6683-91; PMID:16818642; http://dx.doi.org/10.1158/0008-5472.CAN-06-0425
  • Dennis EA, Norris PC. Eicosanoid storm in infection and inflammation. Nat Rev Immunol 2015; 15:511-23; PMID:26139350; http://dx.doi.org/10.1038/nri3859
  • Huang Q, Li F, Liu X, Li W, Shi W, Liu FF, O'Sullivan B, He Z, Peng Y, Tan AC et al. Caspase 3-mediated stimulation of tumor cell repopulation during cancer radiotherapy. Nat Med 2011; 17:860-6; PMID:21725296; http://dx.doi.org/10.1038/nm.2385
  • Li F, Huang Q, Chen J, Peng Y, Roop DR, Bedford JS, Li CY. Apoptotic cells activate the “phoenix rising” pathway to promote wound healing and tissue regeneration. Sci Signal 2010; 3:ra13; PMID:20179271; http://dx.doi.org/10.1126/scisignal.2000634.
  • Li ZL, Ye SB, OuYang LY, Zhang H, Chen YS, He J, Chen QY, Qian CN, Zhang XS, Cui J et al. COX-2 promotes metastasis in nasopharyngeal carcinoma by mediating interactions between cancer cells and myeloid-derived suppressor cells. Oncoimmunology 2015; 4:e1044712; PMID:26451317; http://dx.doi.org/10.1080/2162402X.2015.1044712
  • Motz GT, Santoro SP, Wang LP, Garrabrant T, Lastra RR, Hagemann IS, Lal P, Feldman MD, Benencia F, Coukos G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat Med 2014; 20:607-15; PMID:24793239; http://dx.doi.org/10.1038/nm.3541
  • Zelenay S, van der Veen AG, Bottcher JP, Snelgrove KJ, Rogers N, Acton SE, Chakravarty P, Girotti MR, Marais R, Quezada SA et al. Cyclooxygenase-Dependent Tumor Growth through Evasion of Immunity. Cell 2015; 162:1257-70; PMID:26343581; http://dx.doi.org/10.1016/j.cell.2015.08.015
  • O'Brien AJ, Fullerton JN, Massey KA, Auld G, Sewell G, James S, Newson J, Karra E, Winstanley A, Alazawi W et al. Immunosuppression in acutely decompensated cirrhosis is mediated by prostaglandin E2. Nat Med 2014; 20:518-23; PMID:24728410; http://dx.doi.org/10.1038/nm.3516
  • Gonnermann D, Oberg HH, Kellner C, Peipp M, Sebens S, Kabelitz D, Wesch D. Resistance of cyclooxygenase-2 expressing pancreatic ductal adenocarcinoma cells against gammadelta T cell cytotoxicity. Oncoimmunology 2015; 4:e988460; PMID:25949900; http://dx.doi.org/10.4161/2162402X.2014.988460
  • Martinet L, Jean C, Dietrich G, Fournie JJ, Poupot R. PGE2 inhibits natural killer and gamma delta T cell cytotoxicity triggered by NKR and TCR through a cAMP-mediated PKA type I-dependent signaling. Biochem Pharmacol 2010; 80:838-45; PMID:20470757; http://dx.doi.org/10.1016/j.bcp.2010.05.002
  • Holt D, Ma X, Kundu N, Fulton A. Prostaglandin E(2) (PGE (2)) suppresses natural killer cell function primarily through the PGE(2) receptor EP4. Cancer Immunol Immunother 2011; 60:1577-86; PMID:21681369; http://dx.doi.org/10.1007/s00262-011-1064-9
  • Mao Y, Sarhan D, Steven A, Seliger B, Kiessling R, Lundqvist A. Inhibition of tumor-derived prostaglandin-e2 blocks the induction of myeloid-derived suppressor cells and recovers natural killer cell activity. Clin Cancer Res 2014; 20:4096-106; PMID:24907113; http://dx.doi.org/10.1158/1078-0432.CCR-14-0635
  • Holt DM, Ma X, Kundu N, Collin PD, Fulton AM. Modulation of host natural killer cell functions in breast cancer via prostaglandin E2 receptors EP2 and EP4. J Immunother 2012; 35:179-88; PMID:22306906; http://dx.doi.org/10.1097/CJI.0b013e318247a5e9
  • Karavitis J, Hix LM, Shi YH, Schultz RF, Khazaie K, Zhang M. Regulation of COX2 expression in mouse mammary tumor cells controls bone metastasis and PGE2-induction of regulatory T cell migration. PLoS One 2012; 7:e46342; PMID:23029485; http://dx.doi.org/10.1371/journal.pone.0046342
  • Obermajer N, Muthuswamy R, Lesnock J, Edwards RP, Kalinski P. Positive feedback between PGE2 and COX2 redirects the differentiation of human dendritic cells toward stable myeloid-derived suppressor cells. Blood 2011; 118:5498-505; PMID:21972293; http://dx.doi.org/10.1182/blood-2011-07-365825
  • Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P. PGE(2)-induced CXCL12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res 2011; 71:7463-70; PMID:22025564; http://dx.doi.org/10.1158/0008-5472.CAN-11-2449
  • Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res 2007; 67:4507-13; PMID:17483367; http://dx.doi.org/10.1158/0008-5472.CAN-06-4174
  • Ma X, Holt D, Kundu N, Reader J, Goloubeva O, Take Y, Fulton AM. A prostaglandin E (PGE) receptor EP4 antagonist protects natural killer cells from PGE-mediated immunosuppression and inhibits breast cancer metastasis. Oncoimmunology 2013; 2:e22647; PMID:23482441; http://dx.doi.org/10.4161/onci.22647
  • Sokolowska M, Chen LY, Liu Y, Martinez-Anton A, Qi HY, Logun C, Alsaaty S, Park YH, Kastner DL, Chae JJ et al. Prostaglandin E2 Inhibits NLRP3 Inflammasome Activation through EP4 Receptor and Intracellular Cyclic AMP in Human Macrophages. J Immunol 2015; 194:5472-87; PMID:25917098; http://dx.doi.org/10.4049/jimmunol.1401343
  • Antonopoulos C, Dubyak GR. Chemotherapy engages multiple pathways leading to IL-1beta production by myeloid leukocytes. Oncoimmunology 2014; 3:e27499; PMID:24800164; http://dx.doi.org/10.4161/onci.27499
  • Galluzzi L, Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ 2015; 22:58-73; PMID:25236395; http://dx.doi.org/10.1038/cdd.2014.137
  • Galluzzi L, Vitale I, Abrams JM, Alnemri ES, Baehrecke EH, Blagosklonny MV, Dawson TM, Dawson VL, El-Deiry WS, Fulda S et al. Molecular definitions of cell death subroutines: recommendations of the Nomenclature Committee on Cell Death 2012. Cell Death Differ 2012; 19:107-20; PMID:21760595; http://dx.doi.org/10.1038/cdd.2011.96
  • Galluzzi L, Bravo-San Pedro JM, Kroemer G. Organelle-specific initiation of cell death. Nat Cell Biol 2014; 16:728-36; PMID:25082195; http://dx.doi.org/10.1038/ncb3005
  • Franchi F, Angiolillo DJ. Novel antiplatelet agents in acute coronary syndrome. Nat Rev Cardiol 2015; 12:30-47; PMID:25286881; http://dx.doi.org/10.1038/nrcardio.2014.156
  • Liao X, Lochhead P, Nishihara R, Morikawa T, Kuchiba A, Yamauchi M, Imamura Y, Qian ZR, Baba Y, Shima K et al. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med 2012; 367:1596-606; PMID:23094721; http://dx.doi.org/10.1056/NEJMoa1207756
  • Algra AM, Rothwell PM. Effects of regular aspirin on long-term cancer incidence and metastasis: a systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol 2012; 13:518-27; PMID:22440112; http://dx.doi.org/10.1016/S1470-2045(12)70112-2
  • Walsh DA, McWilliams DF. Mechanisms, impact and management of pain in rheumatoid arthritis. Nat Rev Rheumatol 2014; 10:581-92; PMID:24861185; http://dx.doi.org/10.1038/nrrheum.2014.64
  • Fujita M, Kohanbash G, Fellows-Mayle W, Hamilton RL, Komohara Y, Decker SA, Ohlfest JR, Okada H. COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res 2011; 71:2664-74; PMID:21324923; http://dx.doi.org/10.1158/0008-5472.CAN-10-3055
  • Veltman JD, Lambers ME, van Nimwegen M, Hendriks RW, Hoogsteden HC, Aerts JG, Hegmans JP. COX-2 inhibition improves immunotherapy and is associated with decreased numbers of myeloid-derived suppressor cells in mesothelioma. Celecoxib influences MDSC function. BMC Cancer 2010; 10:464; PMID:20804550; http://dx.doi.org/10.1186/1471-2407-10-464
  • Zeytin HE, Patel AC, Rogers CJ, Canter D, Hursting SD, Schlom J, Greiner JW. Combination of a poxvirus-based vaccine with a cyclooxygenase-2 inhibitor (celecoxib) elicits antitumor immunity and long-term survival in CEA.Tg/MIN mice. Cancer Res 2004; 64:3668-78; PMID:15150127; http://dx.doi.org/10.1158/0008-5472.CAN-03-3878
  • Kundu N, Walser TC, Ma X, Fulton AM. Cyclooxygenase inhibitors modulate NK activities that control metastatic disease. Cancer Immunol Immunother 2005; 54:981-7; PMID:15891886; http://dx.doi.org/10.1007/s00262-005-0669-2
  • Kosaka A, Ohkuri T, Okada H. Combination of an agonistic anti-CD40 monoclonal antibody and the COX-2 inhibitor celecoxib induces anti-glioma effects by promotion of type-1 immunity in myeloid cells and T-cells. Cancer Immunol Immunother 2014; 63:847-57; PMID:24878890; http://dx.doi.org/10.1007/s00262-014-1561-8
  • Hahn T, Alvarez I, Kobie JJ, Ramanathapuram L, Dial S, Fulton A, Besselsen D, Walker E, Akporiaye ET. Short-term dietary administration of celecoxib enhances the efficacy of tumor lysate-pulsed dendritic cell vaccines in treating murine breast cancer. Int J Cancer 2006; 118:2220-31; PMID:16331615; http://dx.doi.org/10.1002/ijc.21616
  • Carlson LM, Rasmuson A, Idborg H, Segerstrom L, Jakobsson PJ, Sveinbjornsson B, Kogner P. Low-dose aspirin delays an inflammatory tumor progression in vivo in a transgenic mouse model of neuroblastoma. Carcinogenesis 2013; 34:1081-8; PMID:23349014; http://dx.doi.org/10.1093/carcin/bgt009
  • Majumder M, Xin X, Liu L, Girish GV, Lala PK. Prostaglandin E2 receptor EP4 as the common target on cancer cells and macrophages to abolish angiogenesis, lymphangiogenesis, metastasis, and stem-like cell functions. Cancer Sci 2014; 105:1142-51; PMID:24981602; http://dx.doi.org/10.1111/cas.12475
  • Ahn B, Kohanbash G, Ohkuri T, Kosaka A, Chen X, Ikeura M, Wang TC, Okada H. Histamine deficiency promotes accumulation of immunosuppressive immature myeloid cells and growth of murine gliomas. Oncoimmunology 2015; 4:e1047581; PMID:26451324; http://dx.doi.org/10.1080/2162402X.2015.1047581
  • Burdette-Radoux S, Holmes CE, Khan FB, Dittus K, Wilson KM, Wood M. Low-dose metronomic cyclophosphamide/methotrexate (LDCM) and aspirin for patients without pathologic complete response (pCR) after neoadjuvant treatment for stage II-III breast cancer. ASCO Meeting Abstracts 2013; 31:163
  • Burdette-Radoux S, Holmes CE, Khan FB, Dittus K, Wilson KM, Wood ME. Low-dose metronomic cyclophosphamide/methotrexate (LDCM) and aspirin for patients who fail to achieve pathologic complete response (pCR) after neoadjuvant treatment for stage II-III breast cancer. ASCO Meeting Abstracts 2013; 31:e12000
  • Kim JW, Marte JL, Bilusic M, Singh NK, Heery CR, Madan RA et al. Safety profile of poxviral vaccines: NCI experience. ASCO Meeting Abstracts 2013; 31:85
  • Kim JW, Marte JL, Singh NK, Heery CR, Madan RA, Pazdur M et al. Safety profile of recombinant poxviral TRICOM vaccines. ASCO Meeting Abstracts 2013; 31:e16036
  • Ellebaek E, Engell-Noerregaard L, Iversen TZ, Froesig TM, Munir S, Hadrup SR et al. Metastatic melanoma patients treated with dendritic cell vaccination, Interleukin-2 and metronomic cyclophosphamide: results from a phase II trial. Cancer Immunol Immunother 2012; 61:1791-804; PMID:22426890; http://dx.doi.org/10.1007/s00262-012-1242-4
  • af Forselles KJ, Root J, Clarke T, Davey D, Aughton K, Dack K, Pullen N. In vitro and in vivo characterization of PF-04418948, a novel, potent and selective prostaglandin EP(2) receptor antagonist. Br J Pharmacol 2011; 164:1847-56; PMID:21595651; http://dx.doi.org/10.1111/j.1476-5381.2011.01495.x
  • Birrell MA, Maher SA, Buckley J, Dale N, Bonvini S, Raemdonck K, Pullen N, Giembycz MA, Belvisi MG. Selectivity profiling of the novel EP2 receptor antagonist, PF-04418948, in functional bioassay systems: atypical affinity at the guinea pig EP2 receptor. Br J Pharmacol 2013; 168:129-38; PMID:22747912; http://dx.doi.org/10.1111/j.1476-5381.2012.02088.x
  • Kundu N, Ma X, Holt D, Goloubeva O, Ostrand-Rosenberg S, Fulton AM. Antagonism of the prostaglandin E receptor EP4 inhibits metastasis and enhances NK function. Breast Cancer Res Treat 2009; 117:235-42; PMID:18792778; http://dx.doi.org/10.1007/s10549-008-0180-5
  • Antonova M, Wienecke T, Maubach K, Thomas E, Olesen J, Ashina M. The pharmacological effect of BGC20-1531, a novel prostanoid EP4 receptor antagonist, in the prostaglandin E2 human model of headache. J Headache Pain 2011; 12:551-9; PMID:21681585; http://dx.doi.org/10.1007/s10194-011-0358-9
  • West NR, McCuaig S, Franchini F, Powrie F. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol 2015; 15:615-29; PMID:26358393; http://dx.doi.org/10.1038/nri3896
  • Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015; 15:409-25; PMID:26105538; http://dx.doi.org/10.1038/nrc3958
  • Zitvogel L, Galluzzi L, Kepp O, Smyth MJ, Kroemer G. Type I interferons in anticancer immunity. Nat Rev Immunol 2015; 15:405-14; PMID:26027717; http://dx.doi.org/10.1038/nri3845
  • Pickup M, Novitskiy S, Moses HL. The roles of TGFbeta in the tumour microenvironment. Nat Rev Cancer 2013; 13:788-99; PMID:24132110; http://dx.doi.org/10.1038/nrc3603
  • Chen X, Wakefield LM, Oppenheim JJ. Synergistic antitumor effects of a TGFbeta inhibitor and cyclophosphamide. Oncoimmunology 2014; 3:e28247; PMID:25050195; http://dx.doi.org/10.4161/onci.28247
  • Lonning S, Mannick J, McPherson JM. Antibody targeting of TGF-beta in cancer patients. Curr Pharm Biotechnol 2011; 12:2176-89; PMID:21619535; http://dx.doi.org/10.2174/138920111798808392
  • Morris JC, Tan AR, Olencki TE, Shapiro GI, Dezube BJ, Reiss M, Hsu FJ, Berzofsky JA, Lawrence DP. Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-beta (TGFbeta) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One 2014; 9:e90353; PMID:24618589; http://dx.doi.org/10.1371/journal.pone.0090353
  • Kashyap MK, Kumar D, Jones H, Amaya-Chanaga CI, Choi MY, Melo-Cardenas J, Ale-Ali A, Kuhne MR, Sabbatini P, Cohen LJ et al. Ulocuplumab (BMS-936564 / MDX1338): a fully human anti-CXCR4 antibody induces cell death in chronic lymphocytic leukemia mediated through a reactive oxygen species-dependent pathway. Oncotarget 2015; 7:2809-22; PMID:26646452; http://dx.doi.org/10.18632/oncotarget.6465.
  • O'Hayre M, Vazquez-Prado J, Kufareva I, Stawiski EW, Handel TM, Seshagiri S, Gutkind JS. The emerging mutational landscape of G proteins and G-protein-coupled receptors in cancer. Nat Rev Cancer 2013; 13:412-24; PMID:23640210; http://dx.doi.org/10.1038/nrc3521
  • Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 2013; 13:227-42; PMID:23470321; http://dx.doi.org/10.1038/nri3405
  • Feig C, Jones JO, Kraman M, Wells RJ, Deonarine A, Chan DS, Connell CM, Roberts EW, Zhao Q, Caballero OL et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2013; 110:20212-7; PMID:24277834; http://dx.doi.org/10.1073/pnas.1320318110
  • Yan M, Jene N, Byrne D, Millar EK, O'Toole SA, McNeil CM, Bates GJ, Harris AL, Banham AH, Sutherland RL et al. Recruitment of regulatory T cells is correlated with hypoxia-induced CXCR4 expression, and is associated with poor prognosis in basal-like breast cancers. Breast Cancer Res 2011; 13:R47; PMID:21521526; http://dx.doi.org/10.1186/bcr2869.
  • Debnath B, Xu S, Grande F, Garofalo A, Neamati N. Small molecule inhibitors of CXCR4. Theranostics 2013; 3:47-75; PMID:23382786; http://dx.doi.org/10.7150/thno.5376
  • de Nigris F, Schiano C, Infante T, Napoli C. CXCR4 inhibitors: tumor vasculature and therapeutic challenges. Recent Pat Anticancer Drug Discov 2012; 7:251-64; PMID:22376154; http://dx.doi.org/10.2174/157489212801820039
  • Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature 1996; 381:661-6; PMID:8649511; http://dx.doi.org/10.1038/381661a0
  • Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 1996; 272:872-7; PMID:8629022; http://dx.doi.org/10.1126/science.272.5263.872
  • Zhang L, Huang Y, He T, Cao Y, Ho DD. HIV-1 subtype and second-receptor use. Nature 1996; 383:768; PMID:8892998; http://dx.doi.org/10.1038/383768a0
  • Peled A, Wald O, Burger J. Development of novel CXCR4-based therapeutics. Expert Opin Investig Drugs 2012; 21:341-53; PMID:22283809; http://dx.doi.org/10.1517/13543784.2012.656197
  • Devine SM, Flomenberg N, Vesole DH, Liesveld J, Weisdorf D, Badel K, Calandra G, DiPersio JF. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin's lymphoma. J Clin Oncol 2004; 22:1095-102; PMID:15020611; http://dx.doi.org/10.1200/JCO.2004.07.131
  • Devine SM, Vij R, Rettig M, Todt L, McGlauchlen K, Fisher N, Devine H, Link DC, Calandra G, Bridger G et al. Rapid mobilization of functional donor hematopoietic cells without G-CSF using AMD3100, an antagonist of the CXCR4/SDF-1 interaction. Blood 2008; 112:990-8; PMID:18426988; http://dx.doi.org/10.1182/blood-2007-12-130179
  • Azab AK, Runnels JM, Pitsillides C, Moreau AS, Azab F, Leleu X, Jia X, Wright R, Ospina B, Carlson AL et al. CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood 2009; 113:4341-51; PMID:19139079; http://dx.doi.org/10.1182/blood-2008-10-186668
  • Sison EA, Magoon D, Li L, Annesley CE, Rau RE, Small D, Brown P. Plerixafor as a chemosensitizing agent in pediatric acute lymphoblastic leukemia: efficacy and potential mechanisms of resistance to CXCR4 inhibition. Oncotarget 2014; 5:8947-58; PMID:25333254; http://dx.doi.org/10.18632/oncotarget.2407
  • Byrne SN, Sarchio SN. AMD3100 protects from UV-induced skin cancer. Oncoimmunology 2014; 3:e27562; PMID:24744978; http://dx.doi.org/10.4161/onci.27562
  • Cho BS, Zeng Z, Mu H, Wang Z, Konoplev S, McQueen T, Protopopova M, Cortes J, Marszalek JR, Peng SB et al. Antileukemia activity of the novel peptidic CXCR4 antagonist LY2510924 as monotherapy and in combination with chemotherapy. Blood 2015; 126:222-32; PMID:26031918; http://dx.doi.org/10.1182/blood-2015-02-628677
  • Hainsworth JD, Mace JR, Reeves JA, Crane EJ, Hamid O, Stille JR et al. Randomized phase II study of sunitinib + CXCR4 inhibitor LY2510924 versus sunitinib alone in first-line treatment of patients with metastatic renal cell carcinoma. ASCO Meeting Abstracts 2015; 33:4547
  • Salgia R, Weaver RW, McCleod M, Stille JR, Yan SB, Roberson S et al. Evaluation of CXCR4 expression on tumor and circulating tumor cells (CTCs) as predictive response marker for CXCR4 antagonist LY2510924 in combination with carboplatin-etoposide in extensive-disease small cell lung cancer (ED-SCLC). ASCO Meeting Abstracts 2015; 33:7567
  • Stille JR, Flynt A, Peek VL, Gross S, Keij J, Connelly MC et al. CXCR4 expression and circulating tumor cell (CTC) counts evaluated as prognostic markers in extensive disease small cell lung cancer (ED-SCLC) patients (pts). ASCO Meeting Abstracts 2015; 33:e18558
  • Ziarek JJ, Liu Y, Smith E, Zhang G, Peterson FC, Chen J, Yu Y, Chen Y, Volkman BF, Li R. Fragment-based optimization of small molecule CXCL12 inhibitors for antagonizing the CXCL12/CXCR4 interaction. Curr Top Med Chem 2012; 12:2727-40; PMID:23368099; http://dx.doi.org/10.2174/1568026611212240003
  • Liang Z, Zhan W, Zhu A, Yoon Y, Lin S, Sasaki M, Klapproth JM, Yang H, Grossniklaus HE, Xu J et al. Development of a unique small molecule modulator of CXCR4. PLoS One 2012; 7:e34038; PMID:22485156; http://dx.doi.org/10.1371/journal.pone.0034038
  • Gravina GL, Mancini A, Muzi P, Ventura L, Biordi L, Ricevuto E, Pompili S, Mattei C, Di Cesare E, Jannini EA et al. CXCR4 pharmacogical inhibition reduces bone and soft tissue metastatic burden by affecting tumor growth and tumorigenic potential in prostate cancer preclinical models. Prostate 2015; 75:1227-46; PMID:26073897; http://dx.doi.org/10.1002/pros.23007
  • Gil M, Seshadri M, Komorowski MP, Abrams SI, Kozbor D. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases. Proc Natl Acad Sci U S A 2013; 110:E1291-300; PMID:23509246; http://dx.doi.org/10.1073/pnas.1220580110
  • Drenckhan A, Kurschat N, Dohrmann T, Raabe N, Koenig AM, Reichelt U, Kaifi JT, Izbicki JR, Gros SJ. Effective inhibition of metastases and primary tumor growth with CTCE-9908 in esophageal cancer. J Surg Res 2013; 182:250-6; PMID:23117118; http://dx.doi.org/10.1016/j.jss.2012.09.035
  • Hassan S, Buchanan M, Jahan K, Aguilar-Mahecha A, Gaboury L, Muller WJ, Alsawafi Y, Mourskaia AA, Siegel PM, Salvucci O et al. CXCR4 peptide antagonist inhibits primary breast tumor growth, metastasis and enhances the efficacy of anti-VEGF treatment or docetaxel in a transgenic mouse model. Int J Cancer 2011; 129:225-32; PMID:20830712; http://dx.doi.org/10.1002/ijc.25665
  • Porvasnik S, Sakamoto N, Kusmartsev S, Eruslanov E, Kim WJ, Cao W, Urbanek C, Wong D, Goodison S, Rosser CJ. Effects of CXCR4 antagonist CTCE-9908 on prostate tumor growth. Prostate 2009; 69:1460-9; PMID:19588526; http://dx.doi.org/10.1002/pros.21008
  • Huang EH, Singh B, Cristofanilli M, Gelovani J, Wei C, Vincent L, Cook KR, Lucci A. A CXCR4 antagonist CTCE-9908 inhibits primary tumor growth and metastasis of breast cancer. J Surg Res 2009; 155:231-6; PMID:19482312; http://dx.doi.org/10.1016/j.jss.2008.06.044
  • Sison EA, Magoon D, Li L, Annesley CE, Romagnoli B, Douglas GJ, Tuffin G, Zimmermann J, Brown P. POL5551, a novel and potent CXCR4 antagonist, enhances sensitivity to chemotherapy in pediatric ALL. Oncotarget 2015; 6:30902-18; PMID:26360610; http://dx.doi.org/10.18632/oncotarget.5094
  • Xie J. The hedgehog's trick for escaping immunosurveillance: The molecular mechanisms driving myeloid-derived suppressor cell recruitment in hedgehog signaling-dependent tumors. Oncoimmunology 2014; 3:e29180; PMID:25054089; http://dx.doi.org/10.4161/onci.29180
  • Sanford DE, Belt BA, Panni RZ, Mayer A, Deshpande AD, Carpenter D, Mitchem JB, Plambeck-Suess SM, Worley LA, Goetz BD et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: a role for targeting the CCL2/CCR2 axis. Clin Cancer Res 2013; 19:3404-15; PMID:23653148; http://dx.doi.org/10.1158/1078-0432.CCR-13-0525
  • Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri F, Rizzuto GA, Lazarus JJ, Pamer EG, Houghton AN et al. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res 2012; 72:876-86; PMID:22174368; http://dx.doi.org/10.1158/0008-5472.CAN-11-1792
  • Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA, Pollard JW. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011; 475:222-5; PMID:21654748; http://dx.doi.org/10.1038/nature10138
  • Sato Y, Shimizu K, Shinga J, Hidaka M, Kawano F, Kakimi K, Yamasaki S, Asakura M, Fujii SI. Characterization of the myeloid-derived suppressor cell subset regulated by NK cells in malignant lymphoma. Oncoimmunology 2015; 4:e995541; PMID:25949922; http://dx.doi.org/10.1080/2162402X.2014.995541
  • Spary LK, Salimu J, Webber JP, Clayton A, Mason MD, Tabi Z. Tumor stroma-derived factors skew monocyte to dendritic cell differentiation toward a suppressive CD14 PD-L1 phenotype in prostate cancer. Oncoimmunology 2014; 3:e955331; PMID:25941611; http://dx.doi.org/10.4161/21624011.2014.955331
  • Lanca T, Costa MF, Goncalves-Sousa N, Rei M, Grosso AR, Penido C, Silva-Santos B. Protective role of the inflammatory CCR2/CCL2 chemokine pathway through recruitment of type 1 cytotoxic gammadelta T lymphocytes to tumor beds. J Immunol 2013; 190:6673-80; PMID:23686489; http://dx.doi.org/10.4049/jimmunol.1300434
  • Ma Y, Mattarollo SR, Adjemian S, Yang H, Aymeric L, Hannani D, Portela Catani JP, Duret H, Teng MW, Kepp O et al. CCL2/CCR2-dependent recruitment of functional antigen-presenting cells into tumors upon chemotherapy. Cancer Res 2014; 74:436-45; PMID:24302580; http://dx.doi.org/10.1158/0008-5472.CAN-13-1265
  • Ma Y, Adjemian S, Galluzzi L, Zitvogel L, Kroemer G. Chemokines and chemokine receptors required for optimal responses to anticancer chemotherapy. Oncoimmunology 2014; 3:e27663; PMID:24800170; http://dx.doi.org/10.4161/onci.27663
  • Iannello A, Raulet DH. Immunosurveillance of senescent cancer cells by natural killer cells. Oncoimmunology 2014; 3:e27616; PMID:24800169; http://dx.doi.org/10.4161/onci.27616
  • Schwarz MK, Wells TN. New therapeutics that modulate chemokine networks. Nat Rev Drug Discov 2002; 1:347-58; PMID:12120410; http://dx.doi.org/10.1038/nrd795
  • Kalliomaki J, Attal N, Jonzon B, Bach FW, Huizar K, Ratcliffe S, Eriksson B, Janecki M, Danilov A, Bouhassira D et al. A randomized, double-blind, placebo-controlled trial of a chemokine receptor 2 (CCR2) antagonist in posttraumatic neuralgia. Pain 2013; 154:761-7; PMID:23523116; http://dx.doi.org/10.1016/j.pain.2013.02.003
  • de Zeeuw D, Bekker P, Henkel E, Hasslacher C, Gouni-Berthold I, Mehling H, Potarca A, Tesar V, Heerspink HJ, Schall TJ. The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomised trial. Lancet Diabetes Endocrinol 2015; 3:687-96; PMID:26268910; http://dx.doi.org/10.1016/S2213-8587(15)00261-2
  • Sullivan T, Miao Z, Dairaghi DJ, Krasinski A, Wang Y, Zhao BN, Baumgart T, Ertl LS, Pennell A, Seitz L et al. CCR2 antagonist CCX140-B provides renal and glycemic benefits in diabetic transgenic human CCR2 knockin mice. Am J Physiol Renal Physiol 2013; 305:F1288-97; PMID:23986513; http://dx.doi.org/10.1152/ajprenal.00316.2013
  • Sullivan TJ, Miao Z, Zhao BN, Ertl LS, Wang Y, Krasinski A, Walters MJ, Powers JP, Dairaghi DJ, Baumgart T et al. Experimental evidence for the use of CCR2 antagonists in the treatment of type 2 diabetes. Metabolism 2013; 62:1623-32; PMID:23953944; http://dx.doi.org/10.1016/j.metabol.2013.06.008
  • Min SH, Wang Y, Gonsiorek W, Anilkumar G, Kozlowski J, Lundell D, Fine JS, Grant EP. Pharmacological targeting reveals distinct roles for CXCR2/CXCR1 and CCR2 in a mouse model of arthritis. Biochem Biophys Res Commun 2010; 391:1080-6; PMID:20004647; http://dx.doi.org/10.1016/j.bbrc.2009.12.025
  • Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 1996; 381:667-73; PMID:8649512; http://dx.doi.org/10.1038/381667a0
  • Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 1996; 272:1955-8; PMID:8658171; http://dx.doi.org/10.1126/science.272.5270.1955
  • Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, Cerwenka A. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J Immunol 2012; 189:5602-11; PMID:23152559; http://dx.doi.org/10.4049/jimmunol.1201018
  • Tan MC, Goedegebuure PS, Belt BA, Flaherty B, Sankpal N, Gillanders WE, Eberlein TJ, Hsieh CS, Linehan DC. Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. J Immunol 2009; 182:1746-55; PMID:19155524; http://dx.doi.org/10.4049/jimmunol.182.3.1746
  • Weitzenfeld P, Ben-Baruch A. The chemokine system, and its CCR5 and CXCR4 receptors, as potential targets for personalized therapy in cancer. Cancer Lett 2014; 352:36-53; PMID:24141062; http://dx.doi.org/10.1016/j.canlet.2013.10.006
  • Ochoa-Callejero L, Perez-Martinez L, Rubio-Mediavilla S, Oteo JA, Martinez A, Blanco JR. Maraviroc, a CCR5 antagonist, prevents development of hepatocellular carcinoma in a mouse model. PLoS One 2013; 8:e53992; PMID:23326556; http://dx.doi.org/10.1371/journal.pone.0053992
  • Velasco-Velazquez M, Jiao X, De La Fuente M, Pestell TG, Ertel A, Lisanti MP, Pestell RG. CCR5 antagonist blocks metastasis of basal breast cancer cells. Cancer Res 2012; 72:3839-50; PMID:22637726; http://dx.doi.org/10.1158/0008-5472.CAN-11-3917
  • Frankenberger C, Rabe D, Bainer R, Sankarasharma D, Chada K, Krausz T, Gilad Y, Becker L, Rosner MR. Metastasis Suppressors Regulate the Tumor Microenvironment by Blocking Recruitment of Prometastatic Tumor-Associated Macrophages. Cancer Res 2015; 75:4063-73; PMID:26238785; http://dx.doi.org/10.1158/0008-5472.CAN-14-3394
  • Chawla A, Philips AV, Alatrash G, Mittendorf E. Immune checkpoints: A therapeutic target in triple negative breast cancer. Oncoimmunology 2014; 3:e28325; PMID:24843833; http://dx.doi.org/10.4161/onci.28325
  • Opar A. New HIV drug classes on the horizon. Nat Rev Drug Discov 2007; 6:258-9; PMID:17457997; http://dx.doi.org/10.1038/nrd2294
  • Fatkenheuer G, Nelson M, Lazzarin A, Konourina I, Hoepelman AI, Lampiris H, Hirschel B, Tebas P, Raffi F, Trottier B et al. Subgroup analyses of maraviroc in previously treated R5 HIV-1 infection. N Engl J Med 2008; 359:1442-55; PMID:18832245; http://dx.doi.org/10.1056/NEJMoa0803154
  • Choi SW, Reddy P. Current and emerging strategies for the prevention of graft-versus-host disease. Nat Rev Clin Oncol 2014; 11:536-47; PMID:24958183; http://dx.doi.org/10.1038/nrclinonc.2014.102
  • Reshef R, Luger SM, Hexner EO, Loren AW, Frey NV, Nasta SD, Goldstein SC, Stadtmauer EA, Smith J, Bailey S et al. Blockade of lymphocyte chemotaxis in visceral graft-versus-host disease. N Engl J Med 2012; 367:135-45; PMID:22784116; http://dx.doi.org/10.1056/NEJMoa1201248
  • Stellbrink HJ. Novel compounds for the treatment of HIV type-1 infection. Antivir Chem Chemother 2009; 19:189-200; PMID:19483267; http://dx.doi.org/10.1177/095632020901900502
  • Rotstein DM, Gabriel SD, Manser N, Filonova L, Padilla F, Sankuratri S, Ji C, deRosier A, Dioszegi M, Heilek G et al. Synthesis, SAR and evaluation of [1,4′]-bipiperidinyl-4-yl-imidazolidin-2-one derivatives as novel CCR5 antagonists. Bioorg Med Chem Lett 2010; 20:3219-22; PMID:20457517; http://dx.doi.org/10.1016/j.bmcl.2010.04.077
  • Rotstein DM, Gabriel SD, Makra F, Filonova L, Gleason S, Brotherton-Pleiss C, Setti LQ, Trejo-Martin A, Lee EK, Sankuratri S et al. Spiropiperidine CCR5 antagonists. Bioorg Med Chem Lett 2009; 19:5401-6; PMID:19674898; http://dx.doi.org/10.1016/j.bmcl.2009.07.122
  • Pyonteck SM, Akkari L, Schuhmacher AJ, Bowman RL, Sevenich L, Quail DF, Olson OC, Quick ML, Huse JT, Teijeiro V et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med 2013; 19:1264-72; PMID:24056773; http://dx.doi.org/10.1038/nm.3337
  • Ruffell B, Coussens LM. Macrophages and therapeutic resistance in cancer. Cancer Cell 2015; 27:462-72; PMID:25858805; http://dx.doi.org/10.1016/j.ccell.2015.02.015
  • DeNardo DG, Brennan DJ, Rexhepaj E, Ruffell B, Shiao SL, Madden SF, Gallagher WM, Wadhwani N, Keil SD, Junaid SA et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov 2011; 1:54-67; PMID:22039576; http://dx.doi.org/10.1158/2159-8274.CD-10-0028
  • Cavnar MJ, DeMatteo RP. Sarcoma response to targeted therapy dynamically polarizes tumor-associated macrophages. Oncoimmunology 2014; 3:e28463; PMID:25050212; http://dx.doi.org/10.4161/onci.28463
  • Mok S, Tsoi J, Koya RC, Hu-Lieskovan S, West BL, Bollag G, Graeber TG, Ribas A. Inhibition of colony stimulating factor-1 receptor improves antitumor efficacy of BRAF inhibition. BMC Cancer 2015; 15:356; PMID:25939769; http://dx.doi.org/10.1186/s12885-015-1377-8
  • Shiao SL, Ruffell B, DeNardo DG, Faddegon BA, Park CC, Coussens LM. TH2-Polarized CD4(+) T Cells and Macrophages Limit Efficacy of Radiotherapy. Cancer Immunol Res 2015; 3:518-25; PMID:25716473; http://dx.doi.org/10.1158/2326-6066.CIR-14-0232
  • Patwardhan PP, Surriga O, Beckman MJ, de Stanchina E, Dematteo RP, Tap WD, Schwartz GK. Sustained inhibition of receptor tyrosine kinases and macrophage depletion by PLX3397 and rapamycin as a potential new approach for the treatment of MPNSTs. Clin Cancer Res 2014; 20:3146-58; PMID:24718867; http://dx.doi.org/10.1158/1078-0432.CCR-13-2576
  • Sluijter M, van der Sluis TC, van der Velden PA, Versluis M, West BL, van der Burg SH, van Hall T. Inhibition of CSF-1R supports T-cell mediated melanoma therapy. PLoS One 2014; 9:e104230; PMID:25110953; http://dx.doi.org/10.1371/journal.pone.0104230
  • Mok S, Koya RC, Tsui C, Xu J, Robert L, Wu L, Graeber TG, West BL, Bollag G, Ribas A. Inhibition of CSF-1 Receptor Improves the Antitumor Efficacy of Adoptive Cell Transfer Immunotherapy. Cancer Res 2014; 74:153-61; PMID:24247719; http://dx.doi.org/10.1158/0008-5472.CAN-13-1816
  • Tap WD, Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C, Healey JH, Chmielowski B, Staddon AP, Cohn AL, Shapiro GI et al. Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell Tumor. N Engl J Med 2015; 373:428-37; PMID:26222558; http://dx.doi.org/10.1056/NEJMoa1411366
  • Butowski NA, Colman H, De Groot JF, Omuro AMP, Nayak L, Cloughesy TF, Marimuthu A, Perry A, Phillips JJ, West B. A phase 2 study of orally administered PLX3397 in patients with recurrent glioblastoma. ASCO Meeting Abstracts 2014; 32:2023
  • Butowski N, Colman H, De Groot JF, Omuro AM, Nayak L, Wen PY, Cloughesy TF, Marimuthu A, Haidar S, Perry A et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro Oncol 2015; PMID:26449250; http://dx.doi.org/10.1093/neuonc/nov245
  • Sharma N, Wesolowski R, Reebel L, Rodal MB, Peck A, West B, Karlin DA, Dowlati A, Le MH, Coussens LM et al. A phase 1b study to assess the safety of PLX3397, a CSF-1 receptor inhibitor, and paclitaxel in patients with advanced solid tumors. ASCO Meeting Abstracts 2014; 32:TPS3127
  • Strachan DC, Ruffell B, Oei Y, Bissell MJ, Coussens LM, Pryer N, Daniel D. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8 T cells. Oncoimmunology 2013; 2:e26968; PMID:24498562; http://dx.doi.org/10.4161/onci.26968
  • Moughon DL, He H, Schokrpur S, Jiang ZK, Yaqoob M, David J, Lin C, Iruela-Arispe ML, Dorigo O, Wu L. Macrophage Blockade Using CSF1R Inhibitors Reverses the Vascular Leakage Underlying Malignant Ascites in Late-Stage Epithelial Ovarian Cancer. Cancer Res 2015; 75:4742-52; PMID:26471360; http://dx.doi.org/10.1158/0008-5472.CAN-14-3373
  • Achyut BR, Shankar A, Iskander AS, Ara R, Angara K, Zeng P, Knight RA, Scicli AG, Arbab AS. Bone marrow derived myeloid cells orchestrate antiangiogenic resistance in glioblastoma through coordinated molecular networks. Cancer Lett 2015; 369:416-26; PMID:26404753; http://dx.doi.org/10.1016/j.canlet.2015.09.004
  • Boulter L, Guest RV, Kendall TJ, Wilson DH, Wojtacha D, Robson AJ, Ridgway RA, Samuel K, Van Rooijen N, Barry ST et al. WNT signaling drives cholangiocarcinoma growth and can be pharmacologically inhibited. J Clin Invest 2015; 125:1269-85; PMID:25689248; http://dx.doi.org/10.1172/JCI76452
  • Garcia AJ, Ruscetti M, Arenzana TL, Tran LM, Bianci-Frias D, Sybert E, Priceman SJ, Wu L, Nelson PS, Smale ST et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression. Mol Cell Biol 2014; 34:2017-28; PMID:24662052; http://dx.doi.org/10.1128/MCB.00090-14
  • Ryder M, Gild M, Hohl TM, Pamer E, Knauf J, Ghossein R, Joyce JA, Fagin JA. Genetic and pharmacological targeting of CSF-1/CSF-1R inhibits tumor-associated macrophages and impairs BRAF-induced thyroid cancer progression. PLoS One 2013; 8:e54302; PMID:23372702; http://dx.doi.org/10.1371/journal.pone.0054302
  • Komohara Y, Horlad H, Ohnishi K, Fujiwara Y, Bai B, Nakagawa T, Suzu S, Nakamura H, Kuratsu J, Takeya M. Importance of direct macrophage-tumor cell interaction on progression of human glioma. Cancer Sci 2012; 103:2165-72; PMID:22957741; http://dx.doi.org/10.1111/cas.12015
  • Xu J, Escamilla J, Mok S, David J, Priceman S, West B, Bollag G, McBride W, Wu L. CSF1R signaling blockade stanches tumor-infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res 2013; 73:2782-94; PMID:23418320; http://dx.doi.org/10.1158/0008-5472.CAN-12-3981
  • Weizman N, Krelin Y, Shabtay-Orbach A, Amit M, Binenbaum Y, Wong RJ, Gil Z. Macrophages mediate gemcitabine resistance of pancreatic adenocarcinoma by upregulating cytidine deaminase. Oncogene 2014; 33:3812-9; PMID:23995783; http://dx.doi.org/10.1038/onc.2013.357
  • Priceman SJ, Sung JL, Shaposhnik Z, Burton JB, Torres-Collado AX, Moughon DL, Johnson M, Lusis AJ, Cohen DA, Iruela-Arispe ML et al. Targeting distinct tumor-infiltrating myeloid cells by inhibiting CSF-1 receptor: combating tumor evasion of antiangiogenic therapy. Blood 2010; 115:1461-71; PMID:20008303; http://dx.doi.org/10.1182/blood-2009-08-237412
  • Ohnuki H, Tosato G. Notch and TGFbeta: Functional partners facilitating tumor progression. Oncoimmunology 2014; 3:e29029; PMID:25114830; http://dx.doi.org/10.4161/onci.29029
  • Liu FL, Mo EP, Yang L, Du J, Wang HS, Zhang H, Kurihara H, Xu J, Cai SH. Autophagy is involved in TGF-beta1-induced protective mechanisms and formation of cancer-associated fibroblasts phenotype in tumor microenvironment. Oncotarget 2015; PMID:26716641; http://dx.doi.org/10.18632/oncotarget.6702
  • Kloss S, Chambron N, Gardlowski T, Arseniev L, Koch J, Esser R, Glienke W, Seitz O, Köhl U. Increased sMICA and TGFbeta levels in HNSCC patients impair NKG2D-dependent functionality of activated NK cells. Oncoimmunology 2015; 4:e1055993; PMID:26451327; http://dx.doi.org/10.1080/2162402X.2015.1055993
  • Anderton MJ, Mellor HR, Bell A, Sadler C, Pass M, Powell S, Steele SJ, Roberts RR, Heier A. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol 2011; 39:916-24; PMID:21859884; http://dx.doi.org/10.1177/0192623311416259
  • Bueno L, de Alwis DP, Pitou C, Yingling J, Lahn M, Glatt S, Trocóniz IF. Semi-mechanistic modelling of the tumour growth inhibitory effects of LY2157299, a new type I receptor TGF-beta kinase antagonist, in mice. Eur J Cancer 2008; 44:142-50; PMID:18039567; http://dx.doi.org/10.1016/j.ejca.2007.10.008
  • Bhola NE, Balko JM, Dugger TC, Kuba MG, Sanchez V, Sanders M, Stanford J, Cook RS, Arteaga CL. TGF-beta inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest 2013; 123:1348-58; PMID:23391723; http://dx.doi.org/10.1172/JCI65416
  • Yoon JH, Jung SM, Park SH, Kato M, Yamashita T, Lee IK, Sudo K, Nakae S, Han JS, Kim OH et al. Activin receptor-like kinase5 inhibition suppresses mouse melanoma by ubiquitin degradation of Smad4, thereby derepressing eomesodermin in cytotoxic T lymphocytes. EMBO Mol Med 2013; 5:1720-39; PMID:24127404; http://dx.doi.org/10.1002/emmm.201302524
  • Herbertz S, Sawyer JS, Stauber AJ, Gueorguieva I, Driscoll KE, Estrem ST, Cleverly AL, Desaiah D, Guba SC, Benhadji KA et al. Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway. Drug Des Devel Ther 2015; 9:4479-99; PMID:26309397; http://dx.doi.org/10.2147/DDDT.S86621
  • Fujiwara Y, Nokihara H, Yamada Y, Yamamoto N, Sunami K, Utsumi H, Asou H, TakahashI O, Ogasawara K, Gueorguieva I et al. Phase 1 study of galunisertib, a TGF-beta receptor I kinase inhibitor, in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol 2015; 76:1143-52; PMID:26526984; http://dx.doi.org/10.1007/s00280-015-2895-4
  • Rodon J, Carducci MA, Sepulveda JM, Azaro A, Calvo E, Seoane J, Brana I, Sicart E, Gueorguieva I, Cleverly A et al. Integrated data review of the first-in-human dose (FHD) study evaluating safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral transforming growth factor-beta (TGF-ss) receptor I kinase inhibitor, LY2157299 monohydrate (LY). ASCO Meeting Abstracts 2013; 31:2016
  • Rodon J, Carducci M, Sepulveda-Sanchez JM, Azaro A, Calvo E, Seoane J, Braña I, Sicart E, Gueorguieva I, Cleverly A et al. Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor-beta receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Invest New Drugs 2015; 33:357-70; PMID:25529192; http://dx.doi.org/10.1007/s10637-014-0192-4
  • Rodon J, Carducci MA, Sepulveda-Sanchez JM, Azaro A, Calvo E, Seoane J, Braña I, Sicart E, Gueorguieva I, Cleverly AL et al. First-in-human dose study of the novel transforming growth factor-beta receptor I kinase inhibitor LY2157299 monohydrate in patients with advanced cancer and glioma. Clin Cancer Res 2015; 21:553-60; PMID:25424852; http://dx.doi.org/10.1158/1078-0432.CCR-14-1380
  • Kovacs RJ, Maldonado G, Azaro A, Fernandez MS, Romero FL, Sepulveda-Sanchez JM, Corretti M, Carducci M, Dolan M, Gueorguieva I et al. Cardiac Safety of TGF-beta Receptor I Kinase Inhibitor LY2157299 Monohydrate in Cancer Patients in a First-in-Human Dose Study. Cardiovasc Toxicol 2015; 15:309-23; PMID:25488804; http://dx.doi.org/10.1007/s12012-014-9297-4
  • Suarez C, Rodon J, Desjardins A, Forsyth PAJ, Gueorguieva I, Cleverly A et al. Phase Ib study evaluating safety and pharmacokinetics (PK) of the oral transforming growth factor-beta (TGF-ss) receptor I kinase inhibitor LY2157299 monohydrate (LY) when combined with chemoradiotherapy in newly diagnosed malignant gliomas. ASCO Meeting Abstracts 2013; 31:2039
  • Kozloff M, Carbonero R, Nadal T, Gueorguieva I, Cleverly A, Desaiah D et al. Phase Ib study evaluating safety and pharmacokinetics (PK) of the oral transforming growth factor-beta (TGF-ss) receptor I kinase inhibitor LY2157299 monohydrate (LY) when combined with gemcitabine in patients with advanced cancer. ASCO Meeting Abstracts 2013; 31:2563
  • Brandes AA, Carpentier AF, Kesari S, Sepulveda J, Wheeler H, Chinot OL et al. A phase II study of galunisertib monotherapy or galunisertib plus lomustine compared to lomustine monotherapy in recurrent glioblastoma. ASCO Meeting Abstracts 2015; 33:2014
  • Carpentier AF, Brandes AA, Kesari S, Sepulveda JM, Wheeler H, Chinot OL et al. Safety interim data from a three-arm phase II study evaluating safety and pharmacokinetics of the oral transforming growth factor-beta (TGF-ss) receptor I kinase inhibitor LY2157299 monohydrate in patients with glioblastoma at first progression. ASCO Meeting Abstracts 2013; 31:2061
  • Faivre SJ, Santoro A, Kelley RK, Merle P, Gane E, Douillard J-Y et al. Randomized dose comparison phase II study of the oral transforming growth factor-beta (TGF-ss) receptor I kinase inhibitor LY2157299 monohydrate (LY) in patients with advanced hepatocellular carcinoma (HCC). ASCO Meeting Abstracts 2013; 31:4118
  • Sharda DR, Yu S, Ray M, Squadrito ML, De Palma M, Wynn TA, Morris SM Jr, Hankey PA. Regulation of macrophage arginase expression and tumor growth by the Ron receptor tyrosine kinase. J Immunol 2011; 187:2181-92; PMID:21810604; http://dx.doi.org/10.4049/jimmunol.1003460
  • Chaudhuri A. Regulation of Macrophage Polarization by RON Receptor Tyrosine Kinase Signaling. Front Immunol 2014; 5:546; PMID:25400637; http://dx.doi.org/10.3389/fimmu.2014.00546
  • Thorpe LM, Yuzugullu H, Zhao JJ. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer 2015; 15:7-24; PMID:25533673; http://dx.doi.org/10.1038/nrc3860
  • Fruman DA, Rommel C. PI3K and cancer: lessons, challenges and opportunities. Nat Rev Drug Discov 2014; 13:140-56; PMID:24481312; http://dx.doi.org/10.1038/nrd4204
  • Rodon J, Dienstmann R, Serra V, Tabernero J. Development of PI3K inhibitors: lessons learned from early clinical trials. Nat Rev Clin Oncol 2013; 10:143-53; PMID:23400000; http://dx.doi.org/10.1038/nrclinonc.2013.10
  • Gopal AK, Kahl BS, de Vos S, Wagner-Johnston ND, Schuster SJ, Jurczak WJ, Flinn IW, Flowers CR, Martin P, Viardot A, Flinn IW, Flowers CR, Martin P, Viardot A et al. PI3Kdelta inhibition by idelalisib in patients with relapsed indolent lymphoma. N Engl J Med 2014; 370:1008-18; PMID:24450858; http://dx.doi.org/10.1056/NEJMoa1314583
  • Furman RR, Sharman JP, Coutre SE, Cheson BD, Pagel JM, Hillmen P, Barrientos JC, Zelenetz AD, Kipps TJ, Flinn I et al. Idelalisib and rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med 2014; 370:997-1007; PMID:24450857; http://dx.doi.org/10.1056/NEJMoa1315226
  • Ali K, Soond DR, Pineiro R, Hagemann T, Pearce W, Lim EL, Bouabe H, Scudamore CL, Hancox T, Maecker H et al. Inactivation of PI(3)K p110delta breaks regulatory T-cell-mediated immune tolerance to cancer. Nature 2014; 510:407-11; PMID:24919154; http://dx.doi.org/10.1038/nature13444
  • Fu H, Kishore M, Gittens B, Wang G, Coe D, Komarowska I, Infante E, Ridley AJ, Cooper D, Perretti M et al. Self-recognition of the endothelium enables regulatory T-cell trafficking and defines the kinetics of immune regulation. Nat Commun 2014; 5:3436; PMID:24625653; http://dx.doi.org/10.1038/ncomms4436
  • Schmid MC, Avraamides CJ, Dippold HC, Franco I, Foubert P, Ellies LG, Acevedo LM, Manglicmot JR, Song X, Wrasidlo W et al. Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3kgamma, a single convergent point promoting tumor inflammation and progression. Cancer Cell 2011; 19:715-27; PMID:21665146; http://dx.doi.org/10.1016/j.ccr.2011.04.016
  • Shah A, Mangaonkar A. Idelalisib: A Novel PI3Kdelta Inhibitor for Chronic Lymphocytic Leukemia. Ann Pharmacother 2015; 49:1162-70; PMID:26185276; http://dx.doi.org/10.1177/1060028015594813
  • Cheah CY, Nastoupil LJ, Neelapu SS, Forbes SG, Oki Y, Fowler NH. Lenalidomide, idelalisib, and rituximab are unacceptably toxic in patients with relapsed/refractory indolent lymphoma. Blood 2015; 125:3357-9; PMID:25999447; http://dx.doi.org/10.1182/blood-2015-03-633156
  • Hewett YG, Uprety D, Shah BK. Idelalisib- a PI3Kdelta targeting agent for B-cell malignancies. J Oncol Pharm Pract 2016; 22:284-8; PMID:25712626; http://dx.doi.org/10.1177/1078155215572933
  • Kahl BS, Spurgeon SE, Furman RR, Flinn IW, Coutre SE, Brown JR, Benson DM, Byrd JC, Peterman S, Cho Y et al. A phase 1 study of the PI3Kdelta inhibitor idelalisib in patients with relapsed/refractory mantle cell lymphoma (MCL). Blood 2014; 123:3398-405; PMID:24615778; http://dx.doi.org/10.1182/blood-2013-11-537555
  • Brown JR, Byrd JC, Coutre SE, Benson DM, Flinn IW, Wagner-Johnston ND, Spurgeon SE, Kahl BS, Bello C, Webb HK et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110delta, for relapsed/refractory chronic lymphocytic leukemia. Blood 2014; 123:3390-7; PMID:24615777; http://dx.doi.org/10.1182/blood-2013-11-535047
  • Flinn IW, Kahl BS, Leonard JP, Furman RR, Brown JR, Byrd JC, Wagner-Johnston ND, Coutre SE, Benson DM, Peterman S et al. Idelalisib, a selective inhibitor of phosphatidylinositol 3-kinase-delta, as therapy for previously treated indolent non-Hodgkin lymphoma. Blood 2014; 123:3406-13; PMID:24615776; http://dx.doi.org/10.1182/blood-2013-11-538546
  • Sikalidis AK. Amino acids and immune response: a role for cysteine, glutamine, phenylalanine, tryptophan and arginine in T-cell function and cancer? Pathol Oncol Res 2015; 21:9-17; PMID:25351939; http://dx.doi.org/10.1007/s12253-014-9860-0
  • Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM et al. Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res 2004; 64:5839-49; PMID:15313928; http://dx.doi.org/10.1158/0008-5472.CAN-04-0465
  • Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol 2005; 5:641-54; PMID:16056256; http://dx.doi.org/10.1038/nri1668
  • Bogdan C. Nitric oxide synthase in innate and adaptive immunity: an update. Trends Immunol 2015; 36:161-78; PMID:25687683; http://dx.doi.org/10.1016/j.it.2015.01.003
  • Serafini P. Myeloid derived suppressor cells in physiological and pathological conditions: the good, the bad, and the ugly. Immunol Res 2013; 57:172-84; PMID:24203443; http://dx.doi.org/10.1007/s12026-013-8455-2
  • Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D, De Palma A, Mauri P, Monegal A, Rescigno M et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med 2011; 208:1949-62; PMID:21930770; http://dx.doi.org/10.1084/jem.20101956
  • Gallina G, Dolcetti L, Serafini P, De Santo C, Marigo I, Colombo MP, Basso G, Brombacher F, Borrello I, Zanovello P et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest 2006; 116:2777-90; PMID:17016559; http://dx.doi.org/10.1172/JCI28828
  • Van Zandt MC, Whitehouse DL, Golebiowski A, Ji MK, Zhang M, Beckett RP, Jagdmann GE, Ryder TR, Sheeler R, Andreoli M et al. Discovery of (R)-2-amino-6-borono-2-(2-(piperidin-1-yl)ethyl)hexanoic acid and congeners as highly potent inhibitors of human arginases I and II for treatment of myocardial reperfusion injury. J Med Chem 2013; 56:2568-80; PMID:23472952; http://dx.doi.org/10.1021/jm400014c
  • De Santo C, Serafini P, Marigo I, Dolcetti L, Bolla M, Del Soldato P, Melani C, Guiducci C, Colombo MP, Iezzi M et al. Nitroaspirin corrects immune dysfunction in tumor-bearing hosts and promotes tumor eradication by cancer vaccination. Proc Natl Acad Sci U S A 2005; 102:4185-90; PMID:15753302; http://dx.doi.org/10.1073/pnas.0409783102
  • Bratasz A, Weir NM, Parinandi NL, Zweier JL, Sridhar R, Ignarro LJ, Kuppusamy P. Reversal to cisplatin sensitivity in recurrent human ovarian cancer cells by NCX-4016, a nitro derivative of aspirin. Proc Natl Acad Sci U S A 2006; 103:3914-9; PMID:16497833; http://dx.doi.org/10.1073/pnas.0511250103
  • Chu GH, Le Bourdonnec B, Gu M, Ajello CW, Leister LK, Sellitto I, Cassel JA, Tuthill PA, O' Hare H, Dehaven RN et al. Design and Synthesis of Imidazopyrimidine Derivatives as Potent iNOS Dimerization Inhibitors. Open Med Chem J 2009; 3:8-13; PMID:19966921; http://dx.doi.org/10.2174/1874104500903010008
  • Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W, Dolcetti L, Bronte V, Borrello I. Phosphodiesterase-5 inhibition augments endogenous antitumor immunity by reducing myeloid-derived suppressor cell function. J Exp Med 2006; 203:2691-702; PMID:17101732; http://dx.doi.org/10.1084/jem.20061104
  • Califano JA, Khan Z, Noonan KA, Rudraraju L, Zhang Z, Wang H, Goodman S, Gourin CG, Ha PK, Fakhry C et al. Tadalafil augments tumor specific immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res 2015; 21:30-8; PMID:25564570; http://dx.doi.org/10.1158/1078-0432.CCR-14-1716
  • Weed DT, Vella JL, Reis IM, De la Fuente AC, Gomez C, Sargi Z, Nazarian R, Califano J, Borrello I, Serafini P. Tadalafil reduces myeloid-derived suppressor cells and regulatory T cells and promotes tumor immunity in patients with head and neck squamous cell carcinoma. Clin Cancer Res 2015; 21:39-48; PMID:25320361; http://dx.doi.org/10.1158/1078-0432.CCR-14-1711
  • Semeraro M, Galluzzi L. Novel insights into the mechanism of action of lenalidomide. Oncoimmunology 2014; 3:e28386; PMID:25340011; http://dx.doi.org/10.4161/onci.28386
  • Frye SV, Arkin MR, Arrowsmith CH, Conn PJ, Glicksman MA, Hull-Ryde EA, Slusher BS. Tackling reproducibility in academic preclinical drug discovery. Nat Rev Drug Discov 2015; 14:733-4; PMID:26388229; http://dx.doi.org/10.1038/nrd4737
  • Gharwan H, Groninger H. Kinase inhibitors and monoclonal antibodies in oncology: clinical implications. Nat Rev Clin Oncol 2016; 13:209-27; PMID:26718105; http://dx.doi.org/10.1038/nrclinonc.2015.213
  • Frail DE, Brady M, Escott KJ, Holt A, Sanganee HJ, Pangalos MN, Watkins C, Wegner CD. Pioneering government-sponsored drug repositioning collaborations: progress and learning. Nat Rev Drug Discov 2015; 14:833-41; PMID:26585533; http://dx.doi.org/10.1038/nrd4707

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.