5,557

The PGE2-EP Receptor Axis in Colorectal Cancer and Angiogenesis

Federico Feroze Tak

Federico Feroze Tak, Trinity Centre for Health Sciences, St. James’s Hospital, Dublin, Ireland

Correspondence to: Federico Feroze Tak, Trinity Centre for Health Sciences, St. James’s Hospital, Dublin, Ireland.
Email: takf@tcd.ie
Telephone: +353-1896-4099
Received: April 29, 2014
Revised: July 5, 2014
Accepted: July 9, 2014
Published online: August 18, 2014

ABSTRACT

Colorectal Cancer is the third most common cause of cancer-related mortality worldwide with rapidly increasing incidence. Substantial evidence suggests that elevated PGE2 expression, frequently observed in colorectal tumours, enables development of a number of cancer hallmarks, including angiogenesis, which is initiated relatively early in disease and enables tumour progression. However, current therapies targeting COX-2 have an adverse safety profile owing to global prostanoid suppression. Thus, the search for therapies which more specifically modulate components of the PGE2 lifecycle has gained momentum. The prostanoid receptors, specifically the EP-series of receptors via which PGE2 exerts its effects, may be a particularly promising target as they sit at the initiating node of relevant signaling pathways. The enzymes regulating PGE2 synthesis and degradation may also represent potential targets. This article critically examines the literature regarding the PGE2-EP receptor axis and the prospect of targeting its components for anti-angiogenic therapy.

Key words: Colorectal Cancer; inflammation; PGE2; prostanoids; EP1; EP2; EP3; EP4; Angiogenesis; COX-2

© 2014 The Author. Published by ACT Group Ltd.

Tak FF. The PGE2-EP Receptor Axis in Colorectal Cancer and Angiogenesis. Journal of Tumor 2014; 2(8): 208-218 Available from: URL: http://www.ghrnet.org/index.php/JT/article/view/807

INTRODUCTION

Colorectal cancer

Colorectal cancer (CRC) is currently the third most common cancer globally and is responsible for around 8% of cancer deaths. In 2008, over 1.2 million people worldwide were diagnosed with CRC with approximately a 50% mortality rate in the same year[1]. Although males and females have similar incidence rates, there is significant disparity in the geographic distribution of CRC incidence. The incidence of CRC in developed areas such as Australia and North America can be 8 to 10-fold higher than in certain parts of Africa and south-central Asia. CRC is the fourth most common cause of cancer-related death worldwide and stage of detection is the single most important prognostic indicator[2]. Patients diagnosed with stage I disease have five-year survivals of approximately 74% while for patients with stage IV disease this is less than 10%[3]. The vast majority of malignant colon and rectal cancers arise from adenomas following 5-10 year latency in individuals over 50. A number of environmental and genetic risk factors have an established role in CRC development. High visceral adiposity and dietary consumption of red meat and alcohol account for a significant CRC risk in western countries[2]. Inflammatory bowel diseases (IBD) comprising Chron’s Disease and Ulcerative Colitis confer a significantly elevated CRC risk[4,5]. Furthermore, the autosomal dominant hereditary disorders Familial Adenomatous Polyposis (FAP) and heridatry non-polyposis colorectal cancer (HNPCC) together account for 5-10% of CRC cases. Briefly, FAP arises from mutations in APC, a gatekeeper that controls wnt signaling and thus regulates proliferation and differentiation of stem cells in colon crypts[6]. HNPCC arises from mutations in the mismatch repair genes MLH1 and MSH2 that facilitates acquisition of a mutator phenotype[7].

Angiogenesis

The multistep nature of CRC development and progression has been well characterized, with normal colonic epithelium giving rise to adenomas, which progress to carcinoma in situ and finally to invasive disease. Initially, mutations in APC drive neoplasia and subsequent mutations in k-ras and the tumour suppressor P53 lead to a loss of differentiation and increase cell proliferation and survival[8]. As in most solid tumours, nascent colorectal tumours require adequate supply of oxygen and nutrients for sustained growth. The diffusion of growth factors and oxygen from existing capillaries becomes insufficient once tumours grow beyond 1-2 mm[9] and it is necessary for hypoxic transformed cells to release paracrine mediators that enable recruitment of endothelial cells (ECs) from an existing vascular bed into the tumour mass for new vessel formation. This highly-regulated multistep process is referred to as angiogenesis. Normal differentiated adult vasculature is maintained in a quiescent state through the opposing actions of pro-angiogenic growth factors and their anti-angiogenic counterparts. Endogenous Angiogenesis-inducing factors include vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) while angiogenesis-inhibiting factors include angiostatin and vasostatin. For tumour angiogenesis to commence it is necessary for pro-angiogenic mediators to overcome the activity of anti-angiogenic factors, an event referred to as the ‘angiogenic swtich’. Briefly, the steps involved in angiogenesis consist of release of pro-angiogenic factors from tumour cells, binding to cognate receptors on ECs, EC proliferation, extracellular matrix (ECM) degradation, directional migration, tube formation and recruitment of mural cells (smooth muscle cells and pericytes) for stabilization. For an excellent in-depth review detailing the steps in angiogenesis and the regulatory processes see the article by Otrock and colleagues[10].

There is evidence to suggest that angiogenesis begins relatively early in CRC development at the adenoma stage. Staton et al were the first group the examine microvessel density (MVD), a surrogate marker of angiogenesis, in each step of the adenoma-carcinoma sequence in patient samples[11]. Their results indicate a steady increase in expression of VEGF, the archetypal pro-angiogenic peptide, during progression. Angiogenesis was found to be induced early in dysplastic transformation at the adenoma stage. Other studies show MVD to be inversely correlated to overall patient survival, suggesting angiogenesis may accelerate tumour growth and metastasis[12].

CRC and COX-2

Despite our extensive understanding of the molecular mechanisms underlying angiogenesis, the assortment of regulatory factors and receptors that are involved have not been fully characterized. This in part reflects the complexity of the tumour microenvironment, with cells including macrophages, lymphocytes, fibroblasts and ECs themselves playing an important role. Eicosanoids, oxygenated derivatives of omega-6 20-carbon saturated fatty acids, predominantly arachidonic acid (AA), are released from a number of the aforementioned cell types, including tumour cells themselves. Much work in recent years has focused on unraveling the role of eicosanoids in tumour physiology in a bid to identify pertinent therapeutic targets. Eicosanoids include thromboxanes, leukotrienes and prostaglandins that are produced downstream of AA by three major pathways. Intially, AA is released from cell membrane glycerophospholipids through esterification of the sn-2 domain by phospholipase A2[13]. AA can be metabolized via the 5-lipoxygenase (5-LOX) pathway to generate 5-hydroperoxyeicosatetraenoic acid (5-HPETEs), precursors of leukotrienes which have roles in physiologic inflammation, including smooth muscle contraction and leukocyte chemotaxis, but also have been demonstrated to contribute to tumour metastasis[14] and angiogenesis[15-17]. A second pathway for AA metabolism occurs via cytochrome P450 epoxygenases resulting the generation of epoxyeicosatrienoic acids (EETs) which are involved in regulating a number of endothelial processes including calcium flux[18], vascular tone[19] and tissue regeneration[20].

The cyclooxygenase (COX) system accounts for the majority of AA metabolism and is responsible for generating prostaglandins and thromboxanes (collectively termed ‘prostanoids’). Two major COX isoforms exist in mammalian cells, COX-1 and COX-2. COX-1 is constitutively expressed across most tissue types and is involved in housekeeping functions including gastric cytoprotection, platelet aggregation and renal electrolyte homeostasis[21,22]. In contrast, COX-2 constitutive expression is restricted to tissues including the lung, brain and kidney where it performs homeostatic functions[23]. In the majority of healthy tissues, however, COX-2 is not expressed but is rapidly upregulated by inflammatory stimuli, including TLR4 signalling[24] and high levels of reactive oxygen species (ROS)[25]. Both COX-1 and COX-2 function as homodimers to mediate conversion of AA into PGG2, a highly unstable intermediate which is rapidly converted into PGH2 by the peroxidase activity of the COXs[26,27]. As a major source of pro-inflammatory mediators, COX enzymes have long been considered as an important therapeutic target. Non-steroidal anti-inflammatory drugs (NSAIDs), including aspirin, inhibit both COX isoforms and prolonged NSAID use has been associated with loss of gastric mucosal homeostasis, ulceration and renal toxicity[28]. These effects are thought to be a result of COX-1 inhibition, leading to reduced expression of homeostatic prostaglandins. The development of COX-2 specific inhibitors, such as celecoxib, aimed to minimise these complications through inhibition of the inflammatory prostaglandins downstream of COX-2. While successful in a large proportion of patients, selective COX-2 inhibitors have been linked to significantly elevated risk of cardiovascular toxicity, especially myocardial infarction and stroke, across a number of population studies[29,30]. It is believed that downregulation of anti-thrombotic PGI2 expression with sustained levels of COX-1 derived pro-thrombotic TXA2 may underlie these events[31]. Moreover, celecoxib has been shown to interact with non-specific targets including carbonic anhydrase and PDK1[32,33]. These molecules have important physiologic roles and their disruption could lead to unexpected toxicity. The issues described here relating to NSAIDs and COX-2 inhibitors have led to a search for specific therapeutic targets downstream of COX which may be involved in tumour development or progression. Prostanoids, as described below, are involved in various steps of tumorigenesis including proliferation, evasion of apoptosis, metastasis and angiogenesis and therefore represent promising targets.

Prostanoids and their Receptors in Angiogenesis

COX-2 is the rate-limiting enzyme mediating prostaglandin synthesis in inflammation and disease. The endoperoxide PGH2 is converted into the various prostaglandins via terminal prostaglandin synthase (tPGS) enzymes, namely prostaglandin-D synthase (PGDS), PGES, PGFS and PGIS involved in production of prostaglandins D, E, F and I respectively[34]. Following synthesis and release of prostaglandins, these metabolites are able to bind to their cognate receptors receptors in autocrine or paracrine fashion with high specificity. All prostaglandin receptors are rhodopsin-type heptahelical G-protein coupled receptors. Depending on their expression, they are capable of inducing a multitude of intracellular cascades that upregulate pro-inflammatory genes[35] but which are also important in driving tumorigenesis by facilitating acquisition of several hallmarks including proliferation[36], invasion[37] and metastasis[38]. Figure 1 below highlights several important signaling pathways implicated in cancer via PG receptor activation.

Progress in the identification of prostanoid receptors was initially slow owing to the existence of receptor isoforms and varying spatiotemporal expression. Despite data from the 1960’s showing changes in activity and expression of various intracellular secondary messengers following PG receptor ligation, the first prostanoid receptor, that for TXA2, was cloned in 1989[39,40]. Work to characterize the biochemical properties PG receptors rapidly gained momentum. There are currently 8 prostanoid receptors which have been found to be conserved from mouse to humans; DP receptor for PGD2, EPs 1-4 for PGE2, FP for PGF2, IP for PGI2 and TP for TXA2. The PGE2-EP receptor axis is discussed in more detail below

PGE2 and the Tumour microenvironment in Angiogenesis

PGE2 Synthesis

Despite evidence for PGD2, PGI2, PGF2 and TXA2 in angiogenesis and tumorigenesis, PGE2 is the COX-2 metabolite which has been studied most extensively with regards to CRC and is therefore the main focus of this review. The synthesis of PGE2 from PGH2 is mediated by three synthase isoforms, namely cytosolic PGES (cPGES), membrane –bound prostaglandin E synthase 1 (mPGES-1) and mPGES-2. cPGES and mPGES-2 have both characterized as constitutive enzymes, while mPGES-1 levels correlate with COX-2 induction[41,42]. cPGES also appears to preferentially isomerise COX-1 derived PGH2, while mPGES-1 uses COX-2 derived PGH2. mPGES-2 displays no preference as to which endoperoxide substrate to use. The potential of targeting mPGES-1 to selectively inhibit PGE2 synthesis is discussed in more detail later in this review.

Vascular EP Receptors in CRC angiogenesis

Given the abundance of PGE2 in several tumour types, much work has focused on understanding its receptors and downstream signaling pathways in a bid for novel drug targets. The EP receptors consist of four subtypes viz. EP1-4. These GPCRs are coupled to diverse intracellular signaling pathways and regulate a number of tumorigenic processes, of which angiogenesis is just one. Table 1 below summarizes the biochemical properties of each EP receptor and specific effects of their activation on tumour cells, ECs and mural cells.

EP1 is coupled to Gαq and has roles to play in calcium signaling. Both EP2 and EP4 are coupled to Gαs which can elevate levels of cyclic AMP via adenylate cyclase. Paradoxically, it has been shown that EP4 is can also be coupled to Gαi, although this is likely to be cell-type dependent. EP3 is coupled exclusively to Gαi and exerts an inhibitory effect on adenylate cyclase[43].

EP4 is the best characterized receptor with regard to the consequences of its activation on tumour cells and ECs, although its role in smooth muscle cells and pericytes is yet to be elucidated. In vivo it is likely that PGE2 induces hyperproliferation of CRC cells via occupation of EP1, EP2 and EP3 simultaneously. Fujino et al report EP3 to be pro-metastatic for HCA-7 cells, although this is an indirect effect mediated by VEGFR1 expression on ECs[44]. All four EPs regulate various steps of angiogenesis and while EP2 and EP4 independently promote EC migration and survival, no studies as yet report a role for PGE2 in proliferation. Instead, EC proliferation may be induced via the action of VEGF released by tumour cells, stromal cells and ECs themselves later in carcinogenesis. EP1 and EP3 may contribute to the autocrine action of VEGF on ECs through HIF-1α induction[45]. All four EP receptors have been found expressed on vascular smooth muscle cells, which comprise the major cell type mediating vascular tone and providing mechanical support to the vessel. Recently, Fischer et al[46] demonstrated hyaluronan to be induced in smooth muscle cells via EP2 activation. This molecule is a potent angiogenic factor in its own right with roles in EC proliferation, migration and tubologenesis[47]. Currently, no studies have identified expression of EP receptors on vascular pericytes, despite the important paracrine role of these cells in EC survival[48]. Relatively little remains known regarding the action of PGE2 on pericytes, despite higher pericyte investiture for vasculature in advanced CRCs being a negative prognostic indicator[49].

EP Receptors in Tumour Microenvironment-Indirect Angiogenesis

The tumour microenvironment (TME) describes the myriad of cells, soluble factors and ECM components that surround malignant epithelial cells and play a crucial role in regulating the tumour phenotype. Certain cells, including CD8+ T cells, may promote tumour cell death, while other components often mediate tumour progression and metastasis. The roles of TAMs, CAFs and myeloid-derived suppressor cells (MDSCs) with regards to PGE2 signalling and angiogenesis will be examined in more detail below.

Tumor-associated macrophages (TAMs), PGE2 and Angiogenesis

TAMs isolated from colorectal tumours are found to have two distinct phenotypes, denominated M1 and M2. M1 TAMs are characterized by high expression of iNOS, TNF-α, IL-12 and MHC-1/2, while M2 TAMs abundantly express IL-10, TGF-β and arginase1[73]. Owing to their cytokine expression pattern, M1 TAMs are reported to promote an anti-tumour immune response via their ability to activate and polarize CD4+ T cells to a Th1 phenotype which in turn faciliatate CD8+ cytotoxic T cell function, while M2 TAMs actively suppress an anti-tumour immune response and are, by inference, the most abundant TAM subtype in the TME. Nevertheless, there is considerable plasticity in macrophage phenotype, with M1 TAMs undergoing reprogramming to M2 cells (and vice versa) under the influence of local mediators[74].

Several studies report the potential of PGE2, in conjunction with other cytokines, to polarize monocytes towards M2 TAMs[75,76], and in downregulating TNF-α expression[77]. Together, these data suggest an indirect immunomodulatory mechanism for PGE2, although the EP receptors mediating these effects have not been characterised. There has also been considerable interest in the role of TAMs in angiogenesis. Recent work by Jetten and colleagues was found to promote EC proliferation and tubologenesis in vivo, which were dependent on macrophage-derived FGF-2 and PIGF[78]. Contemporaneous work by Zajac et al[79] further demonstrated that M2-macrophage induced angiogenesis is also dependent on expression the zymogen pro-MMP9 which enables ECM degradation. PGE2 has also been demonstrated to increase MMP9 in colorectal tumour cell lines[80], thereby indicating a synergistic effect of PGE2 and TAMs in promoting EC migration. Furthermore, Wu et al demonstrated PGE2 to be a potent inducer of VEGF in M2 TAMs an effect that is exacerbated under hypoxia[81]. It is clear from the above data that PGE2 is a major player in the reciprocal signaling between tumour cells, TAMs and ECs. Understanding the EP signaling pathways regulating M2 macrophage activity may assist in developing drugs which target these pathways to deplete M2 cells while increasing levels of the M1 variety.

Cancer-associated fibroblasts (CAFs), PGE2 and EP receptors

There is considerable interest in understanding the biology of CAFs as they are increasingly being recognized as a central element in the TME, with known roles in inflammation, tumour survival, metabolic reprogramming and angiogenesis[82-84]. Normal fibroblasts play a key role in ECM remodelling and wound healing and are not normally located in tumours. It is not clear if CAFs originate from normal fibroblasts or differentiate from mesenchymal stem cells or other stromal cells[85].

CAFs derived from primary CRCs have been demonstrated to abundantly express COX-2 and this has been implicated in promoting tumour EMT via involvement of downstream NFkB and HIF1 activation[86]. PGE2 expression in CAFs has also been noted, indicating a potential interaction with ECs[87]. In stromal fibroblasts derived from mice with gastric cancer high levels of VEGF secretion were induced under the influence of PGE2, although the involvement of EP receptors was not examined[88]. Few studies have investigated EP receptor expression on CAFs, although Odaka et al[89] identified EP2 and EP4 to be expressed on lung fibroblasts. Owing to the tissue-specific nature of EP expression, it is not clear whether these findings apply to CRC stromal fibroblasts. As CAFs comprise the predominant stromal cell type, further work is necessary to elucidate the interactions between PGE2, fibroblasts and tumour cells.

MDSCs, PGE2 and EP receptors

When MDSCs were initially discovered in the 1970’s they were named ‘natural suppressor’ or null cells owing to a lack of classical monocyte and lymphocyte phenotypic markers. They were later found to have potent immunosuppressive properties and contribute to tumour immune evasion. MDSCs can broadly be defined as the precursors to innate immune components (principally granulocytes, macrophages and dendritic cells) which undergo aberrant clonal expansion in the bone marrow. It is now known that MDSC-specific markers in mice include GR1+ and CD11b+ while in humans they are CD14- CD11b+ and release immunomodulatory factors such as Arg1, IL-10 and iNOS[90]. The last decade has seen an explosion of research on MDSCs and investigation of their roles in a number of cancer hallmarks.

Yang et al[91] noted that MDSCs comprise approximately 5% of tumour volume in mice bearing MC-26 CRC cells. Subsequent work by this group reveals MDSCs to be associated with enhanced MVD and vascular stability. While early work showed prostaglandins to be MDSC inducers, Sinha and colleagues were the first group to characterize the expression of of EP receptors on these cells[92]. They found expression of all four EPs on MDSCs in BALB/c mice, with EP2 critical for MDSC induction and immunosuppressive function. EP2 knockout was associated with a corresponding decrease in tumour size. Other work has since indicated both EP2 and EP4 signalling may be involved in MDSC induction[93]. In comparison, there is little evidence to indicate the importance of EP1 and EP3 expression on MDSCs.

MDSCs express a number of pro-angiogenic factors including VEGF, MMPs 2 and 9 and chemokines CXCL2 and CXCL5[90, 94]. MDSCs are also frequently found to overexpress COX-2 and mPGES-1[93,95], providing a number of drug targets. Interestingly, Xiang et al. noted that released tumour exosomes containing PGE2 and other bioactive molecules were able to fuse with MDSCs to induce expression of VEGF from these cells[96]. While our understanding of exosome release from tumour and stromal cells is in its infancy, it is clear that this represents an important cross-talk mechanism with therapeutic potential. The role of the TME and PGE2 in angiogenesis is summarized in figure 2 below:

Inhibition of the PGE2-EP receptor axis in antiangiogenic therapy

Given the important role of PGE2 in angiogenesis, specific targeting of this prostanoid may yield results in inducing vessel regression, stabilizing existing vasculature and inhibiting tumour growth and metastasis. Such an approach would also attenuate inflammation, chronic levels of which are conducive to tumorigenesis.

mPGES-1 inhibition

As the enzyme directly upstream of PGE2, the potential of inhibiting mPGES-1 has been extensively studied. A number of studies have shown mPGES-1 to be overexpressed in resected colorectal tumours and to be negatively linked to prognosis independent of COX-2 levels. Nakanishi et al[97] demonstrated significantly reduced intestinal cancer growth in APC knockout mice which were also null for mPGES1. Interestingly, mPGES1 deletion was also associated with impaired vascular morphogenesis. This study implies a role for PGE2 in both in cancer cell growth and proper vascularisation of the primary tumour. Lack of PGE2 may reduce tumour size by a loss of both processes. While the data for mPGES1 in tumour growth has been corroborated by other studies[98,99], experiments by Elander et al[100] generated conflicting results showing mPGES1 deletion increases polyp size and frequency in APC knockout mice. This observation may reflect the shunting of PGH2 into parallel pathways that generate alternative eicosanoid species with similar pro-tumour effects[101]. Nevertheless, kamata and colleagues highlight the importance of mPGES-1 expressed in mouse bone marrow-derived cells. Gene knockout significantly impaired angiogenesis in a sponge implantation model, which was strongly correlated with decreased VEGF concentrations in the tumour stroma[102]. This study underlines the interdependence between PGE2 and VEGF in tumour angiogenesis. Recent work by Finnetti et al also shows synergism between mPGES-1 and EGFR in angiogenesis, as pharamacological inhibition with AF3485 (a benzamide derivative) displayed inhibition of mPGES-1 with an approximate 50% decrease in MVD that was also dependent on decreased EGFR signaling[103]. The release of VEGF and FGF-2 in cell lines was also reduced, likely due to a decrease in PGE2. 15-deoxy-Delta(12,14)-prostaglandin J2, the FLAP inhibitor MK886 and the NSAID sulindac have all shown mPGES-1 inhibitory potential[104]. The data with regards to the anti-angiogenic potential of 15-deoxy-Delta(12,14)-prostaglandin J2 are conflicting. While able to induce EC apoptosis via P53 upregulation[105], it also apparently promotes angiogenesis through upregulating VEGF expression from tumour cells in vivo through PPARγ modulation[106]. It is clear that the effects of this compound are cell-type specific. The role of this compound in regulating angiogenesis in the complex tumour mileu requires further investigation in animal models.

The only study to assess the anti-tumour activity of MK886 in CRC cell lines indicates that MK886 in addition to COX-2 inhibitors are necessary to inhibit cell proliferation and angiogenesis via dual block on 5-LOX and COX-2 pathways[107]. Again, further investigation is needed.

Interestingly, DPE, a compound present in olive oil, has been found to decrease expression of mPGES-1 and HIF1-α. This impaired vessel maturation and morphology rather than MVD in vivo[108]. Despite a number of natural compounds displaying mPGES-1 inhibitory activity, including circumin, garcinol and hyperforin, extended administration of these compounds has also shown extended inhibition of the 5-LOX pathway which may limit the biosynthesis of the leukotrienes, essential components of a coordinated inflammatory response[109,110].

Anti-angiogenic Potential of EP Receptor Modulation

The critical role of EP receptors controlling angiogenesis makes them ideal therapeutic targets. Many studies referenced above have utilized compounds that competitively bind these receptors to examine their functions. It is unlikely that highly selective EP antagonists by themselves will have therapeutic utility owing to the redundancy in molecular pathways emanating from these receptors. Nevertheless, selective EP blockade combined with chemotherapy or other anti-angiogenic therapies may prove useful. Dual EP receptor inhibitors (e.g. EP2 and EP4 inhibitors) may also show promise.

While the EP1 antagonist ONO-8713 used by Watanabe et al was shown to reduce CRC growth, there is no data to indicate its potential effects on angiogenesis[111]. The EP1 antagonist SC-51089 has demonstrated anti-neoplastic potential in glioma, but thus far no work has examined this compound in relation to angiogenesis[112]. The paucity of angiogenesis research using EP-1 antagonists probably reflects the limited understanding of this receptor in neovascularisation.

In contrast, a number of interesting EP2 antagonists have emerged for research purposes but there are currently no clinical trials with these compounds. While a number of publications have reported the use of AH6809 to examine the sole impact of EP2 blockade on angiogenesis, this compound is relatively non-specific for EP2 and is known to interact with DP receptors as well as EP1[113]. This lack of specificity has led to conflicts in the literature between studies that used EP2 knockout and those that used AH6809[114]. Recent work by Forselles et al at Pfizer led to the development of PF-04418948. In cell-based assays this compound showed over 2000-fold higher selectivity for EP2 than other EP subtypes, with robust and reproducible findings in vivo[115,116]. While PF-04418948 is clearly a promising research tool in the field of tumour biology, its in vivo pharamacokinetic and safety profiles must be established prior to entering clinical trials. Another antagonist TG4-155 developed by Jiang et al[117] shows similar EP-2 selectivity and may prove useful in this area.

Development of EP3 antagonists began with development and modification of biaryl-acylsulphonamides[118]. This led to development of L-826266 which has been used extensively in cardiovascular research[119,120]. However, the high liphophilicity of this compound renders it unsuitable for clinical use. The compound ONO-AE3-240 was used by Amano et al in characterizing the pro-angiogenic role of EP3 in tumour cells and host stromal cells with proven efficacy in vivo. In spite of this, EP3 antagonists may replicate some of the cardiovascular issues associated with COX inhibitors as other work as shown that firing through the EP3 receptor may be cardioprotective[121].

Antagonism of EP4, like that of EP2, has been extensively explored and the role of EP4 in angiogenesis is perhaps the best understood of all the prostanoid receptors. Zhang et al (2011) used the EP4 antagonist azogrel (AH23848) to demonstrate that EP4 signaling is critical to angiogenesis, a finding that was reproduced in a recent study also using this EP4 antagonist[57]. Nevertheless, all commercially available formulations of this compound also display TP1 inhibitory activity and may lead to adverse effects in vivo. A novel EP4 antagonist developed by RaQualia Pharmaceutics, RQ-15986, has shown efficacy in murine models of breast cancer[122]. While the pharmacologic parameters of this compound must be investigated further, a structurally similar compound RQ7 has shown high safety and selectivity in Phase IIa clinical trials for osteoarthritis pain[123]. Thus RQ-15986 merits further investigation in vivo.

PGT and 15-PGDH modulators

Following its synthesis, PGE2 is exported from the cell predominantly by MRP4, a C group member of the ATP-binding cassette (ABC) family[124]. Extracellular PGE2 is catabolized by a two-step process involving carrier-mediated influx by membrane prostaglandin transporter (PGT) and finally degradation by cytoplasmic 15-hydroxyprostaglandin dehydrogenase (15-PGDH)[125]. There is evidence to indicate that while MRP4 is upregulated in CRC, both PGT and 15-PGDH are suppressed, leading to accumulation of PGE2 in the tumour microenvironment[126]. Thus, pharmacological targeting of these three components may enhance degradation of PGE2 and impede angiogenesis together with other tumour hallmarks.

Several NSAIDs, including indomethacin and ketoprofen, are shown to inhibit MRP4 as part of their mode of action[127]. Studies have also used the small molecule MK571[128,129], although this is non-specific for MRP4 and also inhibits MRP1. More specific inhibitors for MRP4 may be required, although evidence suggests there is a small risk of cholestatic liver injury in patients given concomitant bile salt export pump (BSEP) inhibitors[130].

15- PGDH has been found across several studies to function as a tumour suppressor. Loss of 15-PGDH expression is associated with increased colorectal tumorigenesis in two mouse models of CRC; namely, Min and C57BL/6J[131]. Indeed, this study also found 15-PGDH downregulation early in neoplasia in patients with hereditary predisposition to CRC, thus enabling unrestrained PGE2 biosynthesis. Moreover, 15-PGDH expression levels have been proposed as a biomarker of CRC chemopreventive efficacy for both aspirin and the COX-2-targeted celecoxib, indicating these drugs function primarily by mitigating local PGE2 accummulation in combination with 15-PGDH[132,133]. Evidence also suggests that inhibition of EGFR signaling is able to restore pre-neoplastic 15-PGDH levels[134]. As several studies implicate co-operation between EGFR and the EP4 receptor in CRC progression and angiogenesis, it is feasible that dual inhibition of EGFR and EP4 signalling may effectively reduce PGE2 levels through enhanced 15-PGDH expression[135,136].

The quest to restore normal PGT and 15-PGDH activity will undoubtedly be assisted by understanding the epigenetic mechanisms leading to silencing of these genes. While epigenetic silencing of PGT is postulated[126], no evidence has yet been published to indicate that chromatin remodeling factors or microRNAs play a role in its regulation. By contrast, the genetic and epigenetic mechanisms regulating 15-PGDH have been extensively characterized. Snail, a member of the zinc finger transcription factors activated in EMT , is capable of binding conserved E-box elements in the promoter of 15-PGDH and repressing its expression[137]. Backlund et al[138] used various histone deacetylase inhibitors (HDACIs) in CRC cell lines to suggest HDAC2 plays a role in suppression of 15-PGDH. These findings were extended by Wang et al[139] who recently showed that reactivation of 15-PGDH in lung cancer using HDACs was paralleled by increased COX-2 expression. However, HDACIs together with COX-2 selective inhibitors or EP2/4 antagonists reversed PGE2 accumulation and nullified angiogenesis in vitro. While recapitulation of these results is needed in vivo for CRC, it is clear that a multi-pronged approach targeting various components of the PGE2 axis is desirable.

Conclusions and Future Directions

It is clear that PGE2 and EP receptors play an important role in angiogenesis, with their effects being best characterized on ECs and tumour cells. However, given the complexity of signaling in the TME it is necessary to elucidate the role and tissue expression patterns of prostanoid receptors in TAMs, MDSCs and CAFs. It is also unclear what role EP receptors play in the smooth muscle cells and pericytes which support tumour vasculature. Targeting EP receptors may reduce the pro-angiogenic potential of one cell type but may have adverse consequences on other cell types and whole organ systems. Therefore more studies in vivo in CRC models are necessary, perhaps examining combinations of EP antagonists and FP antagonists together with PGT and 15-PGDH enhancers. Synergism of these compounds with existing chemotherapies and molecular targeted therapies should also be investigated. The development of small molecules which are specific for particular prostanoid receptors will undoubtedly assist in furthering our understanding of prostanoid signaling, and following pharmacokinetic profiling and chemical modification may display potent therapeutic effects in vivo. Progress in this area may also be applied more broadly to other solid malignancies and autoimmune diseases.

CONFLICT OF INTERESTS

There are no conflicts of interest with regard to the present study.

REFERENCES

1 CRUK. 2014 27/02/2014]; Available from: http://www.cancerresearchuk.org/cancer-help/type/bowel-cancer/treatment/statistics-and-outlook-for-bowel-cancer

2 Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg 2009; 22(4): 191-197

3 cancer.org. 2013 27/02/2014]; Available from: http://www.cancer.org/cancer/colonandrectumcancer/detailedguide/colorectal-cancer-survival-rates

4 Jess T, Rungoe C, Peyrin-Biroulet L. Risk of colorectal cancer in patients with ulcerative colitis: a meta-analysis of population-based cohort studies. Clin Gastroenterol Hepatol 2012; 10(6): 639-645

5 Freeman HJ. Colorectal cancer risk in Crohn’s disease. World J Gastroenterol 2008; 14(12): 1810-1811

6 Lang M, Borgmann M, Oberhuber G, Evstatiev R, Jimenez K, Dammann KW, Jambrich M, Khare V, Campregher C, Ristl R, Gasche C. Thymoquinone attenuates tumor growth in ApcMin mice by interference with Wnt-signaling. Mol Cancer 2013; 12(1): 41

7 Muller A, Fishel R. Mismatch repair and the hereditary non-polyposis colorectal cancer syndrome (HNPCC). Cancer Invest 2002; 20(1): 102-9

8 Vogelstein B, Kinzler KW. The multistep nature of cancer. Trends Genet 1993; 9(4): 138-41

9 Dijkgraaf I, Boerman OC. Radionuclide imaging of tumor angiogenesis. Cancer Biother Radiopharm 2009; 24(6): 637-47

10 Otrock, Z.K, Mahfouz, R.A, Makarem, J.A, and Shamseddine, A.I, Understanding the biology of angiogenesis: review of the most important molecular mechanisms. Blood Cells Mol Dis 2007; 39(2): 212-20

11 Staton CA, Chetwood AS, Cameron IC, Cross SS, Brown NJ, Reed MW. The angiogenic switch occurs at the adenoma stage of the adenoma carcinoma sequence in colorectal cancer. Gut 2007; 56(10): 1426-32

12 Des Guetz, G, Uzzan, B, Nicolas, P, Cucherat, M, Morere, J.F, Benamouzig, R, Breau, J.L, and Perret, G.Y, Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer 2006; 94(12): 1823-32

13 Turunen PM, Ekholm ME, Somerharju P, Kukkonen JP. Arachidonic acid release mediated by OX1 orexin receptors. Br J Pharmacol 2010; 159(1): 212-21

14 Seo JM, Park S, Kim JH. Leukotriene B4 receptor-2 promotes invasiveness and metastasis of ovarian cancer cells through signal transducer and activator of transcription 3 (STAT3)-dependent up-regulation of matrix metalloproteinase 2. J Biol Chem 2012; 287(17): 13840-9

15 Xu L, Zhang L, Liu L, Fang S, Lu Y, Wei E, Zhang W. Involvement of cysteinyl leukotriene receptors in angiogenesis in rat thoracic aortic rings. Pharmazie 2010; 65(10): 750-4

16 Kim GY, Lee JW, Cho SH, Seo JM, Kim JH. Role of the low-affinity leukotriene B4 receptor BLT2 in VEGF-induced angiogenesis. Arterioscler Thromb Vasc Biol 2009; 29(6): 915-20

17 Tsopanoglou NE, Pipili-Synetos E, Maragoudakis ME. Leukotrienes C4 and D4 promote angiogenesis via a receptor-mediated interaction. Eur J Pharmacol 1994; 258(1-2): 151-4

18 Imig JD, Pham BT, LeBlanc EA, Reddy KM, Falck JR, Inscho EW. Cytochrome P450 and cyclooxygenase metabolites contribute to the endothelin-1 afferent arteriolar vasoconstrictor and calcium responses. Hypertension 2000; 35(1 Pt 2): 307-12

19 Cazade M, Bidaud I, Hansen PB, Lory P, Chemin J. 5,6-EET potently inhibits T-type calcium channels: implication in the regulation of the vascular tone. Pflugers Arch 2013

20 Panigrahy D, Kalish BT, Huang S, Bielenberg DR, Le HD, Yang J, Edin ML, Lee CR, Benny O, Mudge DK, Butterfield CE, Mammoto A, Mammoto T, Inceoglu B, Jenkins RL, Simpson MA, Akino T, Lih FB, Tomer KB, Ingber DE, Hammock BD, Falck JR, Manthati VL, Kaipainen A, D’Amore PA, Puder M, Zeldin DC, Kieran MW. Epoxyeicosanoids promote organ and tissue regeneration. Proc Natl Acad Sci U S A 2013; 110(33): 13528-33

21 Zidar N, Dolenc-Strazar Z, Jeruc J, Jerse M, Balazic J, Gartner U, Jermol U, Zupanc T, Stajer D. Expression of cyclooxygenase-1 and cyclooxygenase-2 in the normal human heart and in myocardial infarction. Cardiovasc Pathol 2007; 16(5): 300-4

22 Perrone MG, Scilimati A, Simone L, Vitale P. Selective COX-1 inhibition: A therapeutic target to be reconsidered. Curr Med Chem 2010; 17(32): 3769-805

23 Tegeder I, Neupert W, Guhring H, Geisslinger G. Effects of selective and unselective cyclooxygenase inhibitors on prostanoid release from various rat organs. J Pharmacol Exp Ther 2000; 292(3): 1161-8

24 Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, Xu R, Inoue H, Arditi M, Dannenberg AJ, Abreu MT. Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: Role in proliferation and apoptosis in the intestine. Gastroenterology 2006; 131(3): 862-77

25 Barbieri SS, Eligini S, Brambilla M, Tremoli E, Colli S, Reactive oxygen species mediate cyclooxygenase-2 induction during monocyte to macrophage differentiation: critical role of NADPH oxidase. Cardiovasc Res 2003; 60(1): 187-97

26 Rouzer CA, Marnett LJ. Cyclooxygenases: structural and functional insights. J Lipid Res 2009; 50 Suppl: S29-34

27 Liu J, Seibold SA, Rieke CJ, Song I, Cukier RI, Smith WL. Prostaglandin endoperoxide H synthases: peroxidase hydroperoxide specificity and cyclooxygenase activation. J Biol Chem 2007; 282(25): 18233-44

28 Silverstein FE, Faich G, Goldstein JL, Simon LS, Pincus T, Whelton A, Makuch R, Eisen G, Agrawal NM, Stenson WF, Burr AM, Zhao WW, Kent JD, Lefkowith JB, Verburg KM, Geis GS. Gastrointestinal toxicity with celecoxib vs nonsteroidal anti-inflammatory drugs for osteoarthritis and rheumatoid arthritis: the CLASS study: A randomized controlled trial. Celecoxib Long-term Arthritis Safety Study. Jama 2000; 284(10): 1247-55

29 Davies NM, Smith GD, Windmeijer F, Martin RM. COX-2 selective nonsteroidal anti-inflammatory drugs and risk of gastrointestinal tract complications and myocardial infarction: an instrumental variable analysis. Epidemiology 2013; 24(3): 352-62

30 Huang WF, Hsiao FY, Tsai YW, Wen YW, Shih YT. Cardiovascular events associated with long-term use of celecoxib, rofecoxib and meloxicam in Taiwan: an observational study. Drug Saf 2006; 29(3): 261-72

31 Foudi N, Louedec L, Cachina T, Brink C, Norel X. Selective cyclooxygenase-2 inhibition directly increases human vascular reactivity to norepinephrine during acute inflammation. Cardiovasc Res 2009; 81(2): 269-77

32 Knudsen JF, Carlsson U, Hammarstrom P, Sokol GH, Cantilena LR. The cyclooxygenase-2 inhibitor celecoxib is a potent inhibitor of human carbonic anhydrase II. Inflammation 2004; 28(5): 285-90

33 Abdulhameed MD, Hamza A, Zhan CG. Microscopic modes and free energies of 3-phosphoinositide-dependent kinase-1 (PDK1) binding with celecoxib and other inhibitors. J Phys Chem B 2006; 110(51): 26365-74

34 Jabbour HN, Sales KJ. Prostaglandin receptor signalling and function in human endometrial pathology. Trends Endocrinol Metab 2004; 15(8): 398-404

35 Hata AN, Breyer RM. Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation. Pharmacol Ther 2004; 103(2): 147-66

36 Xia D, Wang D, Kim SH, Katoh H, DuBois RN. Prostaglandin E2 promotes intestinal tumor growth via DNA methylation. Nat Med 2012; 18(2): 224-6

37 Li X, Tai HH. Activation of thromboxane A2 receptor (TP) increases the expression of monocyte chemoattractant protein -1 (MCP-1)/chemokine (C-C motif) ligand 2 (CCL2) and recruits macrophages to promote invasion of lung cancer cells. PLoS One 2013; 8(1): e54073

38 Stringfellow DA, Fitzpatrick FA. Prostaglandin D2 controls pulmonary metastasis of malignant melanoma cells. Nature 1979; 282(5734): 76-8

39 Jones RL, Giembycz MA, and Woodward DF. Prostanoid receptor antagonists: development strategies and therapeutic applications. Br J Pharmacol 2009; 158(1): 104-45

40 Ushikubi F, Nakajima M, Hirata M, Okuma M, Fujiwara M, Narumiya S. Purification of the thromboxane A2/prostaglandin H2 receptor from human blood platelets. J Biol Chem 1989; 264(28): 16496-501

41 Nardone G, Rocco A, Vaira D, Staibano S, Budillon A, Tatangelo F, Sciulli MG, Perna F, Salvatore G, Di Benedetto M, De Rosa G, Patrignani P. Expression of COX-2, mPGE-synthase1, MDR-1 (P-gp), and Bcl-xL: a molecular pathway of H pylori-related gastric carcinogenesis. J Pathol 2004; 202(3): 305-12

42 Nakatani Y, Hokonohara Y, Tajima Y, Kudo I, Hara S. Involvement of the constitutive prostaglandin E synthase cPGES/p23 in expression of an initial prostaglandin E2 inactivating enzyme, 15-PGDH. Prostaglandins Other Lipid Mediat 2011; 94(3-4): 112-7

43 Brock T. G Protein-Coupled Receptors for PGE2: Many Effects from One Lipid. 2010 26/02/2014]; Available from: https://www.caymanchem.com/app/template/Article.vm/article/2144.

44 Fujino H, Toyomura K, Chen XB, Regan JW, Murayama T. Prostaglandin E(2) regulates cellular migration via induction of vascular endothelial growth factor receptor-1 in HCA-7 human colon cancer cells. Biochem Pharmacol 2011; 81(3): 379-87

45 Suzuki K, Nishi K, Takabuchi S, Kai S, Matsuyama T, Kurosawa S, Adachi T, Maruyama T, Fukuda K, Hirota K. Differential roles of prostaglandin E-type receptors in activation of hypoxia-inducible factor 1 by prostaglandin E1 in vascular-derived cells under non-hypoxic conditions. PeerJ 2013; 1: e220

46 Fischer JW, Schror K. Regulation of hyaluronan synthesis by vasodilatory prostaglandins. Implications for atherosclerosis. Thromb Haemost 2007; 98(2): 287-95

47 Genasetti A, Vigetti D, Viola M, Karousou E, Moretto P, Rizzi M, Bartolini B, Clerici M, Pallotti F, De Luca G, Passi A. Hyaluronan and human endothelial cell behavior. Connect Tissue Res 2008; 49(3): 120-3

48 Franco M, Roswall P, Cortez E, Hanahan D, Pietras K. Pericytes promote endothelial cell survival through induction of autocrine VEGF-A signaling and Bcl-w expression. Blood 2011; 118(10): 2906-17

49 Yonenaga Y, Mori A, Onodera H, Yasuda S, Oe H, Fujimoto A, Tachibana T, and Imamura M. Absence of smooth muscle actin-positive pericyte coverage of tumor vessels correlates with hematogenous metastasis and prognosis of colorectal cancer patients. Oncology 2005; 69(2): 159-66

50 Kawamori T, Kitamura T, Watanabe K, Uchiya N, Maruyama T, Narumiya S, Sugimura, T, Wakabayashi K. Prostaglandin E receptor subtype EP(1) deficiency inhibits colon cancer development. Carcinogenesis 2005; 26(2): 353-7

51 Wang D, Dubois RN. Prostaglandins and cancer. Gut 2006; 55(1): 15-22

52 Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-beta through a PGE2-dependent mechanisms. Carcinogenesis 2008; 29(11): 2227-35

53 Jadhav V, Jabre A, Lin SZ, Lee TJ. EP1- and EP3-receptors mediate prostaglandin E2-induced constriction of porcine large cerebral arteries. J Cereb Blood Flow Metab 2004; 24(12): 1305-16

54 Sonoshita M, Takaku K, Sasaki N, Sugimoto Y, Ushikubi F, Narumiya S, Oshima M, Taketo MM. Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc(Delta 716) knockout mice. Nat Med 2001; 7(9): 1048-51

55 Baba Y, Nosho K, Shima K, Goessling W, Chan AT, Ng K, Chan JA, Giovannucci EL, Fuchs CS, Ogino S. PTGER2 overexpression in colorectal cancer is associated with microsatellite instability, independent of CpG island methylator phenotype. Cancer Epidemiol Biomarkers Prev 2010; 19(3): 822-31

56 Kamiyama M, Pozzi A, Yang L, DeBusk LM, Breyer RM, Lin PC. EP2, a receptor for PGE2, regulates tumor angiogenesis through direct effects on endothelial cell motility and survival. Oncogene 2006; 25(53): 7019-28

57 Sakurai T, Suzuki K, Yoshie M, Hashimoto K, Tachikawa E, Tamura K. Stimulation of tube formation mediated through the prostaglandin EP2 receptor in rat luteal endothelial cells. J Endocrinol 2011; 209(1): 33-43

58 Yau L, Zahradka P. PGE(2) stimulates vascular smooth muscle cell proliferation via the EP2 receptor. Mol Cell Endocrinol 2003; 203(1-2): 77-90

59 Pantazaka E, Taylor EJ, Bernard WG, Taylor CW. Ca(2+) signals evoked by histamine H1 receptors are attenuated by activation of prostaglandin EP2 and EP4 receptors in human aortic smooth muscle cells. Br J Pharmacol 2013; 169(7): 1624-34

60 Shoji Y, Takahashi M, Kitamura T, Watanabe K, Kawamori T, Maruyama T, Sugimoto Y, Negishi M, Narumiya S, Sugimura T, Wakabayashi K. Downregulation of prostaglandin E receptor subtype EP3 during colon cancer development. Gut 2004; 53(8): 1151-8

61 Salvado MD, Di Gennaro A, Lindbom L, Agerberth B, Haeggstrom JZ. Cathelicidin LL-37 induces angiogenesis via PGE2-EP3 signaling in endothelial cells, in vivo inhibition by aspirin. Arterioscler Thromb Vasc Biol 2013; 33(8): 1965-72

62 Zhang J, Zou F, Tang J, Zhang Q, Gong Y, Wang Q, Shen Y, Xiong L, Breyer RM, Lazarus M, Funk CD, Yu Y. Cyclooxygenase-2-derived prostaglandin E(2) promotes injury-induced vascular neointimal hyperplasia through the E-prostanoid 3 receptor. Circ Res 2013; 113(2): 104-14

63 Doherty GA, Byrne SM, Molloy ES, Malhotra V, Austin SC, Kay EW, Murray FE, Fitzgerald DJ. Proneoplastic effects of PGE2 mediated by EP4 receptor in colorectal cancer. BMC Cancer 2009; 9: 207

64 Cherukuri DP, Chen XB, Goulet AC, Young RN, Han Y, Heimark RL, Regan JW, Meuillet E, Nelson MA. The EP4 receptor antagonist, L-161,982, blocks prostaglandin E2-induced signal transduction and cell proliferation in HCA-7 colon cancer cells. Exp Cell Res 2007; 313(14): 2969-79

65 Leone V, di Palma A, Ricchi P, Acquaviva F, Giannouli M, Di Prisco AM, Iuliano F, Acquaviva AM. PGE2 inhibits apoptosis in human adenocarcinoma Caco-2 cell line through Ras-PI3K association and cAMP-dependent kinase A activation. Am J Physiol Gastrointest Liver Physiol 2007; 293(4): G673-81

66 Chell SD, Witherden IR, Dobson RR, Moorghen M, Herman AA, Qualtrough D, Williams AC, Paraskeva C. Increased EP4 receptor expression in colorectal cancer progression promotes cell growth and anchorage independence. Cancer Res 2006; 66(6): 3106-13

67 Yang L, Huang Y, Porta R, Yanagisawa K, Gonzalez A, Segi E, Johnson DH, Narumiya S, Carbone DP. Host and direct antitumor effects and profound reduction in tumor metastasis with selective EP4 receptor antagonism. Cancer Res 2006; 66(19): 9665-72

68 Yu Y, Chadee K. Prostaglandin E2 stimulates IL-8 gene expression in human colonic epithelial cells by a posttranscriptional mechanism. J Immunol 1998; 161(7): 3746-52

69 Rao R, Redha R, Macias-Perez I, Su Y, Hao C, Zent R, Breyer MD, Pozzi A. Prostaglandin E2-EP4 receptor promotes endothelial cell migration via ERK activation and angiogenesis in vivo. J Biol Chem 2007; 282(23): 16959-68

70 Konya V, Ullen A, Kampitsch N, Theiler A, Philipose S, Parzmair GP, Marsche G, Peskar BA, Schuligoi R, Sattler W, Heinemann A. Endothelial E-type prostanoid 4 receptors promote barrier function and inhibit neutrophil trafficking. J Allergy Clin Immunol 2013; 131(2): 532-40.e1-2

71 Aso H, Ito S, Mori A, Morioka M, Suganuma N, Kondo M, Imaizumi K, Hasegawa Y. Prostaglandin E2 enhances interleukin-8 production via EP4 receptor in human pulmonary microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2012; 302(2): L266-73

72 Zhu Z, Fu C, Li X, Song Y, Li C, Zou M, Guan Y, Zhu Y. Prostaglandin E2 promotes endothelial differentiation from bone marrow-derived cells through AMPK activation. PLoS One 2011; 6(8): e23554

73 Hilchie AL, Wuerth K, Hancock RE. Immune modulation by multifaceted cationic host defense (antimicrobial) peptides. Nat Chem Biol 2013; 9(12): 761-8

74 Lyamina SV, Kruglov SV, Vedenikin TY, Borodovitsyna OA, Suvorova IA, Shimshelashvili Sh L, Malyshev IY. Alternative reprogramming of M1/M2 phenotype of mouse peritoneal macrophages in vitro with interferon-gamma and interleukin-4. Bull Exp Biol Med 2012; 152(4): 548-51

75 Heusinkveld M, de Vos van Steenwijk PJ, Goedemans R, Ramwadhdoebe TH, Gorter A, Welters MJ, van Hall T, van der Burg SH. M2 macrophages induced by prostaglandin E2 and IL-6 from cervical carcinoma are switched to activated M1 macrophages by CD4+ Th1 cells. J Immunol 2011; 187(3): 1157-65

76 Liu L, Ge D, Ma L, Mei J, Liu S, Zhang Q, Ren F, Liao H, Pu Q, Wang T, You Z. Interleukin-17 and prostaglandin E2 are involved in formation of an M2 macrophage-dominant microenvironment in lung cancer. J Thorac Oncol 2012; 7(7): 1091-100

77 Kunkel SL, Spengler M, May MA, Spengler R, Larrick J, Remick D. Prostaglandin E2 regulates macrophage-derived tumor necrosis factor gene expression. J Biol Chem 1988; 263(11): 5380-4

78 Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MP, Donners MM. Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis 2014; 17(1): 109-18

79 Zajac E, Schweighofer B, Kupriyanova TA, Juncker-Jensen A, Minder P, Quigley JP, Deryugina EI. Angiogenic capacity of M1- and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9. Blood 2013; 122(25): 4054-67

80 Hsu HH, Hu WS, Lin YM, Kuo WW, Chen LM, Chen WK, Hwang JM, Tsai FJ, Liu CJ, Huang CY. JNK suppression is essential for 17beta-Estradiol inhibits prostaglandin E2-Induced uPA and MMP-9 expressions and cell migration in human LoVo colon cancer cells. J Biomed Sci 2011; 18: 61

81 Wu WK, Llewellyn OP, Bates DO, Nicholson LB, Dick AD. IL-10 regulation of macrophage VEGF production is dependent on macrophage polarisation and hypoxia. Immunobiology 2010; 215(9-10): 796-803

82 Martinez-Outschoorn UE, Lisanti MP, Sotgia F. Catabolic cancer-associated fibroblasts (CAFs) transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol 2014

83 Chiarugi P. Cancer-associated fibroblasts and macrophages: Friendly conspirators for malignancy. Oncoimmunology 2013; 2(9): e25563

84 Nagasaki T, Hara M, Nakanishi H, Takahashi H, Sato M, Takeyama H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. Br J Cancer 2014; 110(2): 469-78

85 Trosko JE. Induction of iPS cells and of cancer stem cells: the stem cell or reprogramming hypothesis of cancer? Anat Rec (Hoboken) 2014; 297(1): 161-73

86 Giannoni E, Bianchini F, Calorini L, Chiarugi P. Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness. Antioxid Redox Signal 2011; 14(12): 2361-71

87 Li T, Yi S, Liu W, Jia C, Wang G, Hua X, Tai Y, Zhang Q, Chen G. Colorectal carcinoma-derived fibroblasts modulate natural killer cell phenotype and antitumor cytotoxicity. Med Oncol 2013; 30(3): 663

88 Guo X, Oshima H, Kitmura T, Taketo MM, Oshima M. Stromal fibroblasts activated by tumor cells promote angiogenesis in mouse gastric cancer. J Biol Chem 2008; 283(28): 19864-71

89 Odaka T, Kobayashi K, Takahashi K, Nakamura H, Matsuoka T. Effect of prostaglandin E2 on urokinase-type plasminogen activator production by human lung fibroblasts. Scand J Clin Lab Invest 2009; 69(2): 225-33

90 Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9(3): 162-74

91 Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B, Shyr Y, Matrisian LM, Carbone DP, Lin PC. Expansion of myeloid immune suppressor Gr+CD11b+ cells in tumor-bearing host directly promotes tumor angiogenesis. Cancer Cell 2004; 6(4): 409-21

92 Sinha P, Clements VK, Fulton AM, Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res 2007; 67(9): 4507-13

93 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(20): 5498-505

94 Roland CL, Dineen SP, Lynn KD, Sullivan LA, Dellinger MT, Sadegh L, Sullivan JP, Shames DS, Brekken RA. Inhibition of vascular endothelial growth factor reduces angiogenesis and modulates immune cell infiltration of orthotopic breast cancer xenografts. Mol Cancer Ther 2009; 8(7): 1761-71

95 Eruslanov E, Daurkin I, Vieweg J, Daaka Y, Kusmartsev S. Aberrant PGE(2) metabolism in bladder tumor microenvironment promotes immunosuppressive phenotype of tumor-infiltrating myeloid cells. Int Immunopharmacol 2011; 11(7): 848-55

96 Xiang X, Poliakov A, Liu C, Liu Y, Deng ZB, Wang J, Cheng Z, Shah SV, Wang GJ, Zhang L, Grizzle WE, Mobley J, Zhang HG. Induction of myeloid-derived suppressor cells by tumor exosomes. Int J Cancer 2009; 124(11): 2621-33

97 Nakanishi M, Montrose DC, Clark P, Nambiar PR, Belinsky GS, Claffey KP, Xu D, Rosenberg DW. Genetic deletion of mPGES-1 suppresses intestinal tumorigenesis. Cancer Res 2008; 68(9): 3251-9

98 Kamei D, Murakami M, Sasaki Y, Nakatani Y, Majima M, Ishikawa Y, Ishii T, Uematsu S, Akira S, Hara S, Kudo I. Microsomal prostaglandin E synthase-1 in both cancer cells and hosts contributes to tumour growth, invasion and metastasis. Biochem J 2010; 425(2): 361-71

99 Hanaka H, Pawelzik SC, Johnsen JI, Rakonjac M, Terawaki K, Rasmuson A, Sveinbjornsson B, Schumacher MC, Hamberg M, Samuelsson B, Jakobsson PJ, Kogner P, Radmark O. Microsomal prostaglandin E synthase 1 determines tumor growth in vivo of prostate and lung cancer cells. Proc Natl Acad Sci U S A 2009; 106(44): 18757-62

100 Elander N, Zhou J, Ungerback J, Dimberg J, Soderkvist P. Association between adenomatosis polyposis coli functional status and microsomal prostaglandin E synthase-1 expression in colorectal cancer. Mol Carcinog 2009; 48(5): 401-7

101 Kapoor M, Kojima F, Qian M, Yang L, Crofford LJ. Shunting of prostanoid biosynthesis in microsomal prostaglandin E synthase-1 null embryo fibroblasts: regulatory effects on inducible nitric oxide synthase expression and nitrite synthesis. Faseb j 2006; 20(13): 2387-9

102 Kamata H, Hosono K, Suzuki T, Ogawa Y, Kubo H, Katoh H, Ito Y, Uematsu S, Akira S, Watanabe M, Majima M, mPGES-1-expressing bone marrow-derived cells enhance tumor growth and angiogenesis in mice. Biomed Pharmacother 2010; 64(6): 409-16

103 Finetti F, Terzuoli E, Bocci E, Coletta I, Polenzani L, Mangano G, Alisi MA, Cazzolla N, Giachetti A, Ziche M, Donnini S, Pharmacological inhibition of microsomal prostaglandin E synthase-1 suppresses epidermal growth factor receptor-mediated tumor growth and angiogenesis. PLoS One 2012; 7(7): e40576

104 Quraishi O, Mancini JA, Riendeau D. Inhibition of inducible prostaglandin E(2) synthase by 15-deoxy-Delta(12,14)-prostaglandin J(2) and polyunsaturated fatty acids. Biochem Pharmacol 2002; 63(6): 1183-9

105 Ho TC, Chen SL, Yang YC, Chen CY, Feng FP, Hsieh JW, Cheng HC, Tsao YP. 15-deoxy-Delta(12,14)-prostaglandin J2 induces vascular endothelial cell apoptosis through the sequential activation of MAPKS and p53. J Biol Chem 2008; 283(44): 30273-88

106 Chintalgattu V, Harris GS, Akula SM, Katwa LC. PPAR-gamma agonists induce the expression of VEGF and its receptors in cultured cardiac myofibroblasts. Cardiovasc Res 2007; 74(1): 140-50

107 Cianchi F, Cortesini C, Magnelli L, Fanti E, Papucci L, Schiavone N, Messerini L, Vannacci A, Capaccioli S, Perna F, Lulli M, Fabbroni V, Perigli G, Bechi P, Masini E. Inhibition of 5-lipoxygenase by MK886 augments the antitumor activity of celecoxib in human colon cancer cells. Mol Cancer Ther 2006; 5(11): 2716-26

108 Terzuoli E, Donnini S, Giachetti A, Iniguez MA, Fresno M, Melillo G, Ziche M. Inhibition of hypoxia inducible factor-1alpha by dihydroxyphenylethanol, a product from olive oil, blocks microsomal prostaglandin-E synthase-1/vascular endothelial growth factor expression and reduces tumor angiogenesis. Clin Cancer Res 2010; 16(16): 4207-16

109 Koeberle A, Northoff H, Werz O. Identification of 5-lipoxygenase and microsomal prostaglandin E2 synthase-1 as functional targets of the anti-inflammatory and anti-carcinogenic garcinol. Biochem Pharmacol 2009; 77(9): 1513-21

110 Koeberle A, Rossi A, Bauer J, Dehm F, Verotta L, Northoff H, Sautebin L, Werz O. Hyperforin, an Anti-Inflammatory Constituent from St. John’s Wort, Inhibits Microsomal Prostaglandin E(2) Synthase-1 and Suppresses Prostaglandin E(2) Formation in vivo. Front Pharmacol 2011; 2: 7

111 Watanabe K, Kawamori T, Nakatsugi S, Ohta T, Ohuchida S, Yamamoto H, Maruyama T, Kondo K, Narumiya S, Sugimura T, Wakabayashi K. Inhibitory effect of a prostaglandin E receptor subtype EP(1) selective antagonist, ONO-8713, on development of azoxymethane-induced aberrant crypt foci in mice. Cancer Lett 2000; 156(1): 57-61

112 Matsuo M, Yoshida N, Zaitsu M, Ishii K, Hamasaki Y. Inhibition of human glioma cell growth by a PHS-2 inhibitor, NS398, and a prostaglandin E receptor subtype EP1-selective antagonist, SC51089. J Neurooncol 2004; 66(3): 285-92

113 Keery RJ. Lumley P. AH6809, a prostaglandin DP-receptor blocking drug on human platelets. Br J Pharmacol 1988; 94(3): 745-54

114 Birrell MA. Nials AT. At last, a truly selective EP(2) receptor antagonist. Br J Pharmacol 2011; 164(7): 1845-6

115 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(7): 1847-56

116 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(1): 129-38

117 Jiang J, Ganesh T, Du Y, Quan Y, Serrano G, Qui M, Speigel I, Rojas A, Lelutiu N, Dingledine R. Small molecule antagonist reveals seizure-induced mediation of neuronal injury by prostaglandin E2 receptor subtype EP2. Proc Natl Acad Sci U S A 2012; 109(8): 3149-54

118 Gallant M, Carriere MC, Chateauneuf A, Denis D, Gareau Y, Godbout C, Greig G, Juteau H, Lachance N, Lacombe P, Lamontagne S, Metters KM, Rochette C, Ruel R, Slipetz D, Sawyer N, Tremblay N, Labelle M. Structure-activity relationship of biaryl acylsulfonamide analogues on the human EP(3) prostanoid receptor. Bioorg Med Chem Lett 2002; 12(18): 2583-6

119 Foudi N, Kotelevets L, Gomez I, Louedec L, Longrois D, Chastre E, Norel X. Differential reactivity of human mammary artery and saphenous vein to prostaglandin E(2) : implication for cardiovascular grafts. Br J Pharmacol 2011; 163(4): 826-34

120 Kozlowska H, Baranowska-Kuczko M, Schlicker E, Kozlowski M, Zakrzeska A, Grzeda E, Malinowska B. EP(3) receptor-mediated contraction of human pulmonary arteries and inhibition of neurogenic tachycardia in pithed rats. Pharmacol Rep 2012; 64(6): 1526-36

121 Zacharowski K, Olbrich A, Piper J, Hafner G, Kondo K, Thiemermann C. Selective activation of the prostanoid EP(3) receptor reduces myocardial infarct size in rodents. Arterioscler Thromb Vasc Biol 1999; 19(9): 2141-7

122 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(1): e22647

123 EP4 Antagonist. 27/02/2014]; Available from: http://www.raqualia.com/product/ep4.html.

124 Lin ZP, Zhu YL, Johnson DR, Rice KP, Nottoli T, Hains BC, McGrath J, Waxman SG, Sartorelli AC. Disruption of cAMP and prostaglandin E2 transport by multidrug resistance protein 4 deficiency alters cAMP-mediated signaling and nociceptive response. Mol Pharmacol 2008; 73(1): 243-51

125 Nomura T, Chang HY, Lu R, Hankin J, Murphy RC, Schuster VL. Prostaglandin signaling in the renal collecting duct: release, reuptake, and oxidation in the same cell. J Biol Chem 2005; 280(31): 28424-9

126 Holla VR, Backlund MG, Yang P, Newman RA, DuBois RN. Regulation of prostaglandin transporters in colorectal neoplasia. Cancer Prev Res (Phila) 2008; 1(2): 93-9

127 Reid G, Wielinga P, Zelcer N, van der Heijden I, Kuil A, de Haas M, Wijnholds J, Borst P. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc Natl Acad Sci U S A 2003; 100(16): 9244-9

128 Hara Y, Sassi Y, Guibert C, Gambaryan N, Dorfmuller P, Eddahibi S, Lompre AM, Humbert M, Hulot JS. Inhibition of MRP4 prevents and reverses pulmonary hypertension in mice. J Clin Invest 2011; 121(7): 2888-97

129 Sinha C, Ren A, Arora K, Moon CS, Yarlagadda S, Zhang W, Cheepala SB, Schuetz JD, Naren AP. Multi-drug resistance protein 4 (MRP4)-mediated regulation of fibroblast cell migration reflects a dichotomous role of intracellular cyclic nucleotides. J Biol Chem 2013; 288(6): 3786-94

130 Kock K, Ferslew BC, Netterberg I, Yang K, Urban TJ, Swaan PW, Stewart PW, Brouwer KL. Risk Factors for Development of Cholestatic Drug-induced Liver Injury: Inhibition of Hepatic Basolateral Bile Acid Transporters MRP3 and MRP4. Drug Metab Dispos 2013

131 Myung SJ, Rerko RM, Yan M, Platzer P, Guda K, Dotson A, Lawrence E, Dannenberg AJ, Lovgren AK, Luo G, Pretlow TP, Newman RA, Willis J, Dawson D, Markowitz SD, 15-Hydroxyprostaglandin dehydrogenase is an in vivo suppressor of colon tumorigenesis. Proc Natl Acad Sci U S A 2006; 103(32): 12098-102

132 Yan M, Myung SJ, Fink SP, Lawrence E, Lutterbaugh J, Yang P, Zhou X, Liu D, Rerko RM, Willis J, Dawson D, Tai HH, Barnholtz-Sloan JS, Newman RA, Bertagnolli MM, Markowitz SD. 15-Hydroxyprostaglandin dehydrogenase inactivation as a mechanism of resistance to celecoxib chemoprevention of colon tumors. Proc Natl Acad Sci U S A 2009; 106(23): 9409-13

133 Fink SP, Yamauchi M, Nishihara R, Jung S, Kuchiba A, Wu K, Cho E, Giovannucci E, Fuchs CS, Ogino S, Markowitz SD, Chan AT. Aspirin and the risk of colorectal cancer in relation to the expression of 15-hydroxyprostaglandin dehydrogenase (HPGD). Sci Transl Med 2014; 6(233): 233re2

134 Yang L, Amann JM, Kikuchi T, Porta R, Guix M, Gonzalez A, Park KH, Billheimer D, Arteaga CL, Tai HH, DuBois R, Carbone DP, Johnson DH. Inhibition of epidermal growth factor receptor signaling elevates 15-hydroxyprostaglandin dehydrogenase in non-small-cell lung cancer. Cancer Res 2007; 67(12): 5587-93

135 Oshima H, Popivanova BK, Oguma K, Kong D, Ishikawa TO, Oshima, M. Activation of epidermal growth factor receptor signaling by the prostaglandin E(2) receptor EP4 pathway during gastric tumorigenesis. Cancer Sci 2011; 102(4): 713-9

136 Pai R, Soreghan B, Szabo IL, Pavelka M, Baatar D, Tarnawski AS. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. Nat Med 2002; 8(3): 289-93

137 Greenland KJ, Jantke I, Jenatschke S, Bracken KE, Vinson C, Gellersen B. The human NAD+-dependent 15-hydroxyprostaglandin dehydrogenase gene promoter is controlled by Ets and activating protein-1 transcription factors and progesterone. Endocrinology 2000; 141(2): 581-97

138. Backlund MG, Mann JR, Holla VR, Shi Q, Daikoku T, Dey SK, DuBois RN. Repression of 15-hydroxyprostaglandin dehydrogenase involves histone deacetylase 2 and snail in colorectal cancer. Cancer Res 2008; 68(22): 9331-7

139 Wang X, Li G, Wang A, Zhang Z, Merchan JR, Halmos B. Combined histone deacetylase and cyclooxygenase inhibition achieves enhanced antiangiogenic effects in lung cancer cells. Mol Carcinog 2013; 52(3): 218-228

Peer reviewer: Zhenghe (John) Wang, Associate Professor, Department of Genetics and Genome Sciences, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.

Refbacks

  • There are currently no refbacks.